1
|
Baazaoui N, Y Alfaifi M, Ben Saad R, Garzoli S. Potential role of long noncoding RNA maternally expressed gene 3 (MEG3) in the process of neurodegeneration. Neuroscience 2025; 565:487-498. [PMID: 39675694 DOI: 10.1016/j.neuroscience.2024.12.023] [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: 03/24/2024] [Revised: 10/28/2024] [Accepted: 12/12/2024] [Indexed: 12/17/2024]
Abstract
Neurodegenerative diseases (ND) are complex diseases of still unknown etiology. Lately, long non-coding RNAs (lncRNAs) have become increasingly popular and implicated in several pathologies as they have several roles and appear to be involved in all biological processes such as cell signaling and cycle control as well as translation and transcription. MEG3 is one of these and acts by binding proteins or directly or competitively binding miRNAs. It has a crucial role in controlling cell death, inflammatory process, oxidative stress, endoplasmic reticulum stress, epithelial-mesenchymal transition and other processes. Recent reports showed that MEG3 is a major driving force of the necrosis phenomena in AD, causing the death of neurons, and its upregulation in cancer patients was linked to tumor suppression. Dysregulation of MEG3 affects neuronal cell death, inflammatory process, smooth muscle cell proliferation and consequently leads to the initiation or the acceleration of the disease. This review examines the current state of knowledge concerning the level of expression and the regulatory function of MEG3 in relation to several NDs. In addition, we examined the relation of MEG3 with neurotrophic factors such as Tumor growth factor β (TGFβ) and its possible mechanism of action. A comprehensive and in-depth analysis of the role of MEG3 in ND could give a clearer picture about the initiation of the process of neuronal death and help develop an alternative therapy that targets MEG3.
Collapse
Affiliation(s)
- Narjes Baazaoui
- Central Labs, King Khalid University, AlQura'a, Abha, P.O. Box 960, Saudi Arabia; Biology Department, Faculty of Science, King Khalid University, Abha, Saudi Arabia; Tissue Culture and Cancer Biology Research Laboratory, King Khalid University, Abha 9004, Saudi Arabia
| | - Mohammad Y Alfaifi
- Central Labs, King Khalid University, AlQura'a, Abha, P.O. Box 960, Saudi Arabia; Biology Department, Faculty of Science, King Khalid University, Abha, Saudi Arabia; Tissue Culture and Cancer Biology Research Laboratory, King Khalid University, Abha 9004, Saudi Arabia
| | - Rania Ben Saad
- Biotechnology and Plant Improvement Laboratory, Center of Biotechnology of Sfax, B.P "1177", Sfax 3018, Tunisia
| | - Stefania Garzoli
- Department of Chemistry and Technologies of Drug, Sapienza University, P. le Aldo Moro 5, 00185 Rome, Italy.
| |
Collapse
|
2
|
Kaur R, Pandey S, Gupta S, Singh J. Harnessing the potential of long non-coding RNAs in the pathophysiology of Alzheimer's disease. Exp Neurol 2024; 385:115134. [PMID: 39740737 DOI: 10.1016/j.expneurol.2024.115134] [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: 07/02/2024] [Revised: 12/08/2024] [Accepted: 12/27/2024] [Indexed: 01/02/2025]
Abstract
Alzheimer's disease (AD), a diverse neurodegenerative disease, is the leading cause of dementia, accounting for 60-80 % of all cases. The pathophysiology of Alzheimer's disease is unknown, and there is no cure at this time. Recent developments in transcriptome-wide profiling have led to the identification of a number of non-coding RNAs (ncRNAs). Among these, long non-coding RNAs (lncRNAs)-long transcripts that don't seem to be able to code for proteins-have drawn attention because they function as regulatory agents in a variety of biological processes. Recent research suggests that lncRNAs play a role in the pathogenesis of Alzheimer's disease by modulating tau hyperphosphorylation, amyloid production, synaptic impairment, neuroinflammation, mitochondrial dysfunction, and oxidative stress, though their precise effects on the disorder are unknown. The biology and modes of action of the best-characterized lncRNAs in AD will be outlined here, with an emphasis on their possible involvement in the pathophysiology of the disease. As lncRNAs may offer prospective prognostic/diagnostic biomarkers and therapeutic targets for the treatment of AD, a greater comprehension of the molecular processes and the intricate network of interactions in which they are implicated could pave the way for future research.
Collapse
Affiliation(s)
- Rasanpreet Kaur
- Department of Biotechnology, Institute of Applied Sciences & Humanities, GLA University, Chaumuhan, Mathura 281406, Uttar Pradesh, India; Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam, India
| | - Swadha Pandey
- Department of Biotechnology, Institute of Applied Sciences & Humanities, GLA University, Chaumuhan, Mathura 281406, Uttar Pradesh, India
| | - Saurabh Gupta
- Department of Biotechnology, Institute of Applied Sciences & Humanities, GLA University, Chaumuhan, Mathura 281406, Uttar Pradesh, India.
| | - Jitendra Singh
- Department of Translational Medicine, All India Institute of Medical Sciences (AIIMS)Bhopal, Saket Nagar, Bhopal 462020, Madhya Pradesh, India
| |
Collapse
|
3
|
Wang C, Liu J, Zheng M, Hu M, Li Q, Zhang X, Gu L. Andrographolide mitigates neurotoxicity induced by lipopolysaccharide or amyloid-β through modulation of miR-222-mediated p62 and NF-κBp65 expression. Eur J Pharmacol 2024; 988:177224. [PMID: 39734037 DOI: 10.1016/j.ejphar.2024.177224] [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: 06/23/2024] [Revised: 12/18/2024] [Accepted: 12/22/2024] [Indexed: 12/31/2024]
Abstract
MicroRNA-222 (miR-222) plays a crucial role in neurodegeneration and is up-regulated in Alzheimer's disease (AD) patients. Andrographolide (Andro) has been reported to have anti-inflammatory and neuroprotective effects, showing potential for treating AD. The relationship between Andro's anti-AD mechanism and the regulation of miR-222 was discussed in this study. Andro protected against cytotoxicity induced by lipopolysaccharide (LPS) or amyloid-β, accompanied by upregulating p62 and Nrf2 mRNA and protein, downregulating TLR4 and NF-κBp65 mRNA and protein, and increasing LC3Ⅱ protein in vitro. miRNA and mRNA sequencing results showed that Andro downregulated miR-222 and upregulated sqstm1/p62. Andro was observed to inhibit the expression of miR-222 and the phosphorylation of NF-κBp65, while simultaneously enhancing the levels of p62 and LC3Ⅱ proteins, decreasing Aβ levels, and attenuating the release of inflammatory factors in the 3xTg-AD mice. MiR-222 mimic increased NF-κBp65 mRNA and protein levels in LPS-induced cells, while miR-222 inhibitors increased p62 mRNA and protein levels as well as Nrf2 and LC3Ⅱ protein, and decreased p-NF-κBp65 protein level in LPS-induced cells. Furthermore, miR-222 mimic reversed the increase in p62 and LC3Ⅱ protein and the decrease in NF-κBp65 mRNA and protein, as well as the decrease in Tau protein levels induced by Andro in LPS-induced cells. These findings suggest that Andro plays a neuroprotective role through downregulation of miR-222 to promote p62 expression while inhibiting NF-kB p65 expression, providing new insights into the mechanism of action of Andro for treating AD.
Collapse
Affiliation(s)
- Can Wang
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang, 310013, P.R. China
| | - Jiayi Liu
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang, 310013, P.R. China
| | - Miao Zheng
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang, 310013, P.R. China
| | - Min Hu
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang, 310013, P.R. China
| | - Qin Li
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang, 310013, P.R. China
| | - Xinyue Zhang
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang, 310013, P.R. China.
| | - Lili Gu
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang, 310013, P.R. China.
| |
Collapse
|
4
|
Wang F, Li Y, Shen H, Martinez-Feduchi P, Ji X, Teng P, Krishnakumar S, Hu J, Chen L, Feng Y, Yao B. Identification of pathological pathways centered on circRNA dysregulation in association with irreversible progression of Alzheimer's disease. Genome Med 2024; 16:129. [PMID: 39529134 PMCID: PMC11552301 DOI: 10.1186/s13073-024-01404-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Circular RNAs (circRNAs) are highly stable regulators, often accumulated in mammalian brains and thought to serve as "memory molecules" that govern the long process of aging. Mounting evidence demonstrated circRNA dysregulation in the brains of Alzheimer's disease (AD) patients. However, whether and how circRNA dysregulation underlies AD progression remains unexplored. METHODS We combined Poly(A)-tailing/RNase R digestion experimental approach with CARP, our published computational framework using pseudo-reference alignment for more sensitive and accurate circRNA detection to identify genome-wide circRNA dysregulation and their downstream pathways in the 5xFAD mouse cerebral cortex between 5 and 7 months of age, a critical window marks the transition from reversible to irreversible pathogenic progression. Dysregulated circRNAs and pathways associated with disease progression in 5xFAD cortex were systematically compared with circRNAs affected in postmortem subcortical areas of a large human AD cohort. A top-ranked circRNA conserved and commonly affected in AD patients and 5xFAD mice was depleted in cultured cells to examine AD-relevant molecular and cellular changes. RESULTS We discovered genome-wide circRNA alterations specifically in 5xFAD cortex associated with AD progression, many of which are commonly dysregulated in the subcortical areas of AD patients. Among these circRNAs, circGigyf2 is highly conserved and showed the highest net reduction specifically in the 7-month 5xFAD cortex. CircGIGYF2 level in AD patients' cortices negatively correlated with dementia severity. Mechanistically, we found multiple AD-affected splicing factors that are essential for circGigyf2 biogenesis. Functionally, we identified and experimentally validated the conserved roles of circGigyf2 in sponging AD-relevant miRNAs and AD-associated RNA binding proteins (RBPs), including the cleavage and polyadenylation factor 6 (CPSF6). Moreover, circGigyf2 downregulation in AD promoted silencing activities of its sponged miRNAs and enhanced polyadenylation site processing efficiency of CPSF6 targets. Furthermore, circGigyf2 depletion in a mouse neuronal cell line dysregulated circGigyf2-miRNA and circGigyf2-CPSF6 axes and potentiated apoptotic responses upon insults, which strongly support the causative roles of circGigyf2 deficiency in AD neurodegeneration. CONCLUSIONS Together, our results unveiled brain circRNAs associated with irreversible disease progression in an AD mouse model that is also affected in AD patients and identified novel molecular mechanisms underlying the dysregulation of conserved circRNA pathways contributing to AD pathogenesis.
Collapse
Affiliation(s)
- Feng Wang
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Yangping Li
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Huifeng Shen
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Paula Martinez-Feduchi
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Xingyu Ji
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Peng Teng
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Siddharth Krishnakumar
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Jian Hu
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Li Chen
- Department of Biostatistics, College of Public Health and Health Professions & College of Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Yue Feng
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| | - Bing Yao
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| |
Collapse
|
5
|
Nurkolis F, Utami TW, Alatas AI, Wicaksono D, Kurniawan R, Ratmandhika SR, Sukarno KT, Pahu YGP, Kim B, Tallei TE, Tjandrawinata RR, Alhasyimi AA, Surya R, Helen H, Halim P, Muhar AM, Syahputra RA. Can salivary and skin microbiome become a biodetector for aging-associated diseases? Current insights and future perspectives. FRONTIERS IN AGING 2024; 5:1462569. [PMID: 39484071 PMCID: PMC11524912 DOI: 10.3389/fragi.2024.1462569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 10/02/2024] [Indexed: 11/03/2024]
Abstract
Growth and aging are fundamental elements of human development. Aging is defined by a decrease in physiological activities and higher illness vulnerability. Affected by lifestyle, environmental, and hereditary elements, aging results in disorders including cardiovascular, musculoskeletal, and neurological diseases, which accounted for 16.1 million worldwide deaths in 2019. Stress-induced cellular senescence, caused by DNA damage, can reduce tissue regeneration and repair, promoting aging. The root cause of many age-related disorders is inflammation, encouraged by the senescence-associated secretory phenotype (SASP). Aging's metabolic changes and declining immune systems raise illness risk via promoting microbiome diversity. Stable, individual-specific skin and oral microbiomes are essential for both health and disease since dysbiosis is linked with periodontitis and eczema. Present from birth to death, the human microbiome, under the influence of diet and lifestyle, interacts symbiotically with the body. Poor dental health has been linked to Alzheimer's and Parkinson's diseases since oral microorganisms and systemic diseases have important interactions. Emphasizing the importance of microbiome health across the lifetime, this study reviews the understanding of the microbiome's role in aging-related diseases that can direct novel diagnosis and treatment approaches.
Collapse
Affiliation(s)
- Fahrul Nurkolis
- Department of Biological Sciences, Faculty of Sciences and Technology, State Islamic University of Sunan Kalijaga (UIN Sunan Kalijaga), Yogyakarta, Indonesia
| | - Trianna Wahyu Utami
- Department of Dental Biomedical Sciences, Faculty of Dentistry, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Aiman Idrus Alatas
- Program of Clinical Microbiology Residency, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Danar Wicaksono
- Alumnus Department of Dermatology and Venereology, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Rudy Kurniawan
- Graduate School of Medicine, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | | | | | | | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Trina Ekawati Tallei
- Department of Biology, Faculty of Mathematics and Natural Sciences, Sam Ratulangi University, Manado, Indonesia
| | | | - Ananto Ali Alhasyimi
- Department of Orthodontics, Faculty of Dentistry, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Reggie Surya
- Department of Food Technology, Faculty of Engineering, Bina Nusantara University, Jakarta, Indonesia
| | - Helen Helen
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan, Indonesia
| | - Princella Halim
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan, Indonesia
| | - Adi Muradi Muhar
- Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - Rony Abdi Syahputra
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan, Indonesia
| |
Collapse
|
6
|
Black CM, Braden AA, Nasim S, Tripathi M, Xiao J, Khan MM. The Association between Long Non-Coding RNAs and Alzheimer's Disease. Brain Sci 2024; 14:818. [PMID: 39199508 PMCID: PMC11353078 DOI: 10.3390/brainsci14080818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 08/05/2024] [Accepted: 08/13/2024] [Indexed: 09/01/2024] Open
Abstract
Neurodegeneration occurs naturally as humans age, but the presence of additional pathogenic mechanisms yields harmful and consequential effects on the brain. Alzheimer's disease (AD), the most common form of dementia, is a composite of such factors. Despite extensive research to identify the exact causes of AD, therapeutic approaches for treating the disease continue to be ineffective, indicating important gaps in our understanding of disease mechanisms. Long non-coding RNAs (lncRNAs) are an endogenous class of regulatory RNA transcripts longer than 200 nucleotides, involved in various regulatory networks, whose dysregulation is evident in several neural and extraneural diseases. LncRNAs are ubiquitously expressed across all tissues with a wide range of functions, including controlling cell differentiation and development, responding to environmental stimuli, and other physiological processes. Several lncRNAs have been identified as potential contributors in worsening neurodegeneration due to altered regulation during abnormal pathological conditions. Within neurological disease, lncRNAs are prime candidates for use as biomarkers and pharmacological targets. Gender-associated lncRNA expression is altered in a gender-dependent manner for AD, suggesting more research needs to be focused on this relationship. Overall, research on lncRNAs and their connection to neurodegenerative disease is growing exponentially, as commercial enterprises are already designing and employing RNA therapeutics. In this review we offer a comprehensive overview of the current state of knowledge on the role of lncRNAs in AD and discuss the potential implications of lncRNA as potential therapeutic targets and diagnostic biomarkers in patients with Alzheimer's disease.
Collapse
Affiliation(s)
- Carson M. Black
- Departments of Neurology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (C.M.B.); (J.X.)
| | - Anneliesse A. Braden
- Departments of Neurology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (C.M.B.); (J.X.)
- Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Samia Nasim
- Departments of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Science Center, Memphis, TN 38163, USA;
| | - Manish Tripathi
- Medicine and Oncology, University of Texas Rio Grande Valley, McAllen, TX 78504, USA;
| | - Jianfeng Xiao
- Departments of Neurology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (C.M.B.); (J.X.)
| | - Mohammad Moshahid Khan
- Departments of Neurology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (C.M.B.); (J.X.)
- Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Division of Regenerative and Rehabilitation Sciences, Department of Physical Therapy, Center for Muscle, Metabolism and Neuropathology, College of Health Professions, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| |
Collapse
|
7
|
Han L, Chen W, Zong Y, Zhao Y, Li J, He Z, Du R. Analysis of the mechanism of fibrauretine alleviating Alzheimer's disease based on transcriptomics and proteomics. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2024; 28:361-377. [PMID: 38926843 PMCID: PMC11211758 DOI: 10.4196/kjpp.2024.28.4.361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 01/17/2024] [Accepted: 01/18/2024] [Indexed: 06/28/2024]
Abstract
The dried rattan stem of the Fibraurea Recisa Pierre plant contains the active ingredient known as fibrauretine (FN). Although it greatly affects Alzheimer's disease (AD), the mechanism of their effects still remains unclear. Proteomics and transcriptomics analysis methods were used in this study to determine the mechanism of FN in the treatment of AD. AD model is used through bilateral hippocampal injection of Aβ1-40. After successful modeling, FN was given for 30 days. The results showed that FN could improve the cognitive dysfunction of AD model rats, reduce the expression of Aβ and P-Tau, increase the content of acetylcholine and reduce the activity of acetylcholinesterase. The Kyoto Encyclopedia of Genes and Genomes enriched differentially expressed genes and proteins are involved in signaling pathways including metabolic pathway, AD, pathway in cancer, PI3K-AKT signaling pathway, and cAMP signaling pathway. Transcriptomics and proteomics sequencing resulted in 19 differentially expressed genes and proteins. Finally, in contrast to the model group, after FN treatment, the protein expressions and genes associated with the PI3K-AKT pathway were significantly improved in RT-qPCR and Western blot and assays. This is consistent with the findings of transcriptomic and proteomic analyses. Our study found that, FN may improve some symptoms of AD model rats through PI3K-AKT signaling pathway.
Collapse
Affiliation(s)
- Lu Han
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Weijia Chen
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Ying Zong
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Yan Zhao
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Jianming Li
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Zhongmei He
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
- Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education of China, Changchun 130118, China
- Jilin Provincial Engineering Research Center for Efficient Breeding and Product Development of Sika Deer of China, Changchun 130118, China
| | - Rui Du
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
- Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education of China, Changchun 130118, China
- Jilin Provincial Engineering Research Center for Efficient Breeding and Product Development of Sika Deer of China, Changchun 130118, China
| |
Collapse
|
8
|
Shademan B, Yousefi H, Nourazarian A. Methylglyoxal Affects the Expression of miR-125b, miR-107, and Oxidative Stress Pathway-associated Genes in the SH-SY5Y Cell Line. Adv Pharm Bull 2024; 14:419-425. [PMID: 39206401 PMCID: PMC11347743 DOI: 10.34172/apb.2024.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 01/03/2024] [Accepted: 01/07/2024] [Indexed: 09/04/2024] Open
Abstract
Purpose Alzhеimеr's disеasе (AD) is thе most prеvalеnt form of dеmеntia globally. Rеsеarch links thе incrеasе of rеactivе oxidativе spеciеs (ROS) to thе pathogеnеsis of AD; thus, this study invеstigatеd thе impact of mеthylglyoxal (MGO) on thе еxprеssion of miR-125b, miR-107, and gеnеs involvеd in oxidativе strеss signaling in SH-SY5Y cеlls. Methods Thе MTT assay assеssеd MGO's еffеcts on SH-SY5Y viability. miR-125b and miR-107 еxprеssion was analyzеd via rеal-timе PCR. Additionally, thе Human Oxidativе Strеss Pathway Plus RT2 Profilеr PCR array quantifiеd oxidativе pathway gеnе еxprеssion. Results MGO concеntrations undеr 700μM did not significantly rеducе SH-SY5Y viability. MiR-125b and miR-107 еxprеssion in SH-SY5Y cеlls incrеasеd and dеcrеasеd rеspеctivеly (P<0.05). Cеlls trеatеd with 700μM MGO еxhibitеd incrеasеd CCS, CYBB, PRDX3, SPINK1, CYGB, DHCR24 and BAG2 еxprеssion (P<0.05). Thosе trеatеd with 1400μM MGO showеd incrеasеd CCS, CYBB, PRDX3, SPINK1, DUSP1, EPHX2, EPX, FOXM1, and GPX3 еxprеssion (P<0.05). Conclusion MGO altеrs oxidativе strеss pathway gеnе, miR-125b, and miR-107 еxprеssion in SH-SY5Y cеlls. Targеting MGO or miR-125b and miR-107 may providе novеl AD thеrapеutic stratеgiеs or improvе sеvеrе symptoms. Furthеr rеsеarch should еlucidatе thе prеcisе mеchanisms.
Collapse
Affiliation(s)
- Behrouz Shademan
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hadi Yousefi
- Department of Basic Medical Sciences, Khoy University of Medical Sciences, Khoy, Iran
| | - Alireza Nourazarian
- Department of Basic Medical Sciences, Khoy University of Medical Sciences, Khoy, Iran
| |
Collapse
|
9
|
Kim YA, Mellen M, Kizil C, Santa-Maria I. Mechanisms linking cerebrovascular dysfunction and tauopathy: Adding a layer of epiregulatory complexity. Br J Pharmacol 2024; 181:879-895. [PMID: 37926507 DOI: 10.1111/bph.16280] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 09/11/2023] [Accepted: 09/30/2023] [Indexed: 11/07/2023] Open
Abstract
Intracellular accumulation of hyperphosphorylated misfolded tau proteins are found in many neurodegenerative tauopathies, including Alzheimer's disease (AD). Tau pathology can impact cerebrovascular physiology and function through multiple mechanisms. In vitro and in vivo studies have shown that alterations in the blood-brain barrier (BBB) integrity and function can result in synaptic abnormalities and neuronal damage. In the present review, we will summarize how tau proteostasis dysregulation contributes to vascular dysfunction and, conversely, we will examine the factors and pathways leading to tau pathological alterations triggered by cerebrovascular dysfunction. Finally, we will highlight the role epigenetic and epitranscriptomic factors play in regulating the integrity of the cerebrovascular system and the progression of tauopathy including a few observartions on potential therapeutic interventions. LINKED ARTICLES: This article is part of a themed issue From Alzheimer's Disease to Vascular Dementia: Different Roads Leading to Cognitive Decline. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v181.6/issuetoc.
Collapse
Affiliation(s)
- Yoon A Kim
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, New York, USA
- Department of Pathology and Cell Biology, Columbia University, New York, New York, USA
| | - Marian Mellen
- Facultad de Ciencias Experimentales, Universidad Francisco de Vitoria, Pozuelo de Alarcon, Madrid, Spain
| | - Caghan Kizil
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, New York, USA
- Department of Neurology, Columbia University Irving Medical Center, New York, New York, USA
| | - Ismael Santa-Maria
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, New York, USA
- Department of Pathology and Cell Biology, Columbia University, New York, New York, USA
- Facultad de Ciencias Experimentales, Universidad Francisco de Vitoria, Pozuelo de Alarcon, Madrid, Spain
| |
Collapse
|
10
|
Kumar A, Nader MA, Deep G. Emergence of Extracellular Vesicles as "Liquid Biopsy" for Neurological Disorders: Boom or Bust. Pharmacol Rev 2024; 76:199-227. [PMID: 38351075 PMCID: PMC10877757 DOI: 10.1124/pharmrev.122.000788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 11/11/2023] [Accepted: 11/27/2023] [Indexed: 02/16/2024] Open
Abstract
Extracellular vesicles (EVs) have emerged as an attractive liquid biopsy approach in the diagnosis and prognosis of multiple diseases and disorders. The feasibility of enriching specific subpopulations of EVs from biofluids based on their unique surface markers has opened novel opportunities to gain molecular insight from various tissues and organs, including the brain. Over the past decade, EVs in bodily fluids have been extensively studied for biomarkers associated with various neurological disorders, such as Alzheimer's disease, Parkinson's disease, schizophrenia, bipolar disorder, major depressive disorders, substance use disorders, human immunodeficiency virus-associated neurocognitive disorder, and cancer/treatment-induced neurodegeneration. These studies have focused on the isolation and cargo characterization of either total EVs or brain cells, such as neuron-, astrocyte-, microglia-, oligodendrocyte-, pericyte-, and endothelial-derived EVs from biofluids to achieve early diagnosis and molecular characterization and to predict the treatment and intervention outcomes. The findings of these studies have demonstrated that EVs could serve as a repetitive and less invasive source of valuable molecular information for these neurological disorders, supplementing existing costly neuroimaging techniques and relatively invasive measures, like lumbar puncture. However, the initial excitement surrounding blood-based biomarkers for brain-related diseases has been tempered by challenges, such as lack of central nervous system specificity in EV markers, lengthy protocols, and the absence of standardized procedures for biological sample collection, EV isolation, and characterization. Nevertheless, with rapid advancements in the EV field, supported by improved isolation methods and sensitive assays for cargo characterization, brain cell-derived EVs continue to offer unparallel opportunities with significant translational implications for various neurological disorders. SIGNIFICANCE STATEMENT: Extracellular vesicles present a less invasive liquid biopsy approach in the diagnosis and prognosis of various neurological disorders. Characterizing these vesicles in biofluids holds the potential to yield valuable molecular information, thereby significantly impacting the development of novel biomarkers for various neurological disorders. This paper has reviewed the methodology employed to isolate extracellular vesicles derived from various brain cells in biofluids, their utility in enhancing the molecular understanding of neurodegeneration, and the potential challenges in this research field.
Collapse
Affiliation(s)
- Ashish Kumar
- Departments of Cancer Biology (A.K., G.D.), Physiology and Pharmacology (M.A.N.), Radiology (M.A.N.), and Center for Addiction Research (M.A.N., G.D.), Wake Forest University School of Medicine, Winston-Salem, North Carolina; Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, North Carolina (G.D.); and Sticht Center for Healthy Aging and Alzheimer's Prevention, Wake Forest School of Medicine, Winston-Salem, North Carolina (G.D.)
| | - Michael A Nader
- Departments of Cancer Biology (A.K., G.D.), Physiology and Pharmacology (M.A.N.), Radiology (M.A.N.), and Center for Addiction Research (M.A.N., G.D.), Wake Forest University School of Medicine, Winston-Salem, North Carolina; Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, North Carolina (G.D.); and Sticht Center for Healthy Aging and Alzheimer's Prevention, Wake Forest School of Medicine, Winston-Salem, North Carolina (G.D.)
| | - Gagan Deep
- Departments of Cancer Biology (A.K., G.D.), Physiology and Pharmacology (M.A.N.), Radiology (M.A.N.), and Center for Addiction Research (M.A.N., G.D.), Wake Forest University School of Medicine, Winston-Salem, North Carolina; Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, North Carolina (G.D.); and Sticht Center for Healthy Aging and Alzheimer's Prevention, Wake Forest School of Medicine, Winston-Salem, North Carolina (G.D.)
| |
Collapse
|
11
|
Chimthanawala NMA, Haria A, Sathaye S. Non-invasive Biomarkers for Early Detection of Alzheimer's Disease: a New-Age Perspective. Mol Neurobiol 2024; 61:212-223. [PMID: 37596437 DOI: 10.1007/s12035-023-03578-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 08/14/2023] [Indexed: 08/20/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that primarily affects the elderly population. It gradually leads to memory loss, loss of thinking ability, and an overall cognitive decline. However, exhaustive literature is available to suggest that pathological changes in the brain occur decades before the first clinical symptoms appear. This review provides insight into the non-invasive biomarkers for early detection of AD that have been successfully studied in populations across the globe. These biomarkers have been detected in the blood, saliva, breath, and urine samples. Retinal imaging techniques are also reported. In this study, PubMed and Google scholar were the databases employed using keywords "Alzheimer's disease," "neurodegeneration," "non-invasive biomarkers," "early diagnosis," "blood-based biomarkers," and "preclinical AD," among others. The evaluation of these biomarkers will provide early diagnosis of AD in the preclinical stages due to their positive correlation with brain pathology in AD. Early diagnosis with reliable and timely intervention can effectively manage this disease.
Collapse
Affiliation(s)
- Niyamat M A Chimthanawala
- Department of Pharmaceutical Sciences & Technology, Institute of Chemical Technology, Nathalal Parekh Marg, Mumbai, 400019, Maharashtra, India
| | - Akash Haria
- Department of Pharmaceutical Sciences & Technology, Institute of Chemical Technology, Nathalal Parekh Marg, Mumbai, 400019, Maharashtra, India
| | - Sadhana Sathaye
- Department of Pharmaceutical Sciences & Technology, Institute of Chemical Technology, Nathalal Parekh Marg, Mumbai, 400019, Maharashtra, India.
| |
Collapse
|
12
|
Zhou Z, Ye Q, Ren H, Zhang Y, Han B, Yao H, Bai Y. CircDYM attenuates microglial apoptosis via CEBPB/ZC3H4 axis in LPS-induced mouse model of depression. Int J Biol Macromol 2024; 254:127922. [PMID: 37944732 DOI: 10.1016/j.ijbiomac.2023.127922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 11/04/2023] [Accepted: 11/04/2023] [Indexed: 11/12/2023]
Abstract
Major depressive disorder (MDD) is a highly prevalent condition and one of the most common psychiatric disorders worldwide. Circular RNA (circRNA) has been increasingly implicated in MDD. However, a comprehensive understanding of circRNA and microglial apoptosis in depression is incomplete. Here, we show that circDYM inhibits microglial apoptosis induced by LPS via CEBPB/ZC3H4 axis. CircDYM prevents the translocation of CEBPB from cytoplasm to the nucleus by binding with CEBPB. Moreover, LPS-induced CEBPB nuclear entry downregulates the expression of ZC3H4, in which promotes autophagy and apoptosis in microglia. Taken together, our findings provide new insights into the relationship between circDYM and microglial apoptosis and shed new light on the function of this novel mechanism in depression-associated complex changes in the brain.
Collapse
Affiliation(s)
- Zhongqiu Zhou
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing, China
| | - Qingqing Ye
- Department of Pharmacy, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Hui Ren
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing, China
| | - Yuan Zhang
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing, China
| | - Bing Han
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing, China
| | - Honghong Yao
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China; Institute of Life Sciences, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Ying Bai
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing, China.
| |
Collapse
|
13
|
Sharma H, Kaushik M, Goswami P, Sreevani S, Chakraborty A, Ashique S, Pal R. Role of miRNAs in Brain Development. Microrna 2024; 13:96-109. [PMID: 38571343 DOI: 10.2174/0122115366287127240322054519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 02/09/2024] [Accepted: 02/21/2024] [Indexed: 04/05/2024]
Abstract
Non-coding RNAs that are small in size, called microRNAs (miRNAs), exert a consequence in neutralizing gene activity after transcription. The nervous system is a massively expressed organ, and an expanding body of research reveals the vital functions that miRNAs play in the brain's growth and neural activity. The significant benefit of miRNAs on the development of the central nervous system is currently shown through new scientific methods that concentrate on targeting and eradicating vital miRNA biogenesis pathways the elements involving Dicer and DGCR8. Modulation of miRNA has been associated with numerous essential cellular processes on neural progenitors, like differentiation, proliferation, and destiny determination. Current research discoveries that emphasize the significance of miRNAs in the complex process of brain development are included in this book. The miRNA pathway plays a major role in brain development, its operational dynamics, and even diseases. Recent studies on miRNA-mediated gene regulation within neural discrepancy, the circadian period and synaptic remodeling are signs of this. We also discussed how these discoveries may affect our comprehension of the fundamental processes behind brain diseases, highlighting the novel therapeutic opportunities miRNAs provide for treating various human illnesses.
Collapse
Affiliation(s)
- Himanshu Sharma
- Teerthanker Mahaveer College of Pharmacy, Teerthanker Mahaveer University, Moradabad (UP), 244001, India
| | - Monika Kaushik
- Amity Institute of Pharmacy, Amity University Gwalior, 474005, Madhya Pradesh, India
| | - Priyanka Goswami
- Department of Pharmacognosy, Maharashtra Educational Society's H. K. College of Pharmacy, Mumbai, Maharashtra, 400102, India
| | - Sanakattula Sreevani
- Department of Pharmacology, Vivekananda College of Pharmacy, Rajajinagar, Bengaluru, Karnataka, 560055, India
| | - Ananya Chakraborty
- Department of Biotechnology, Maulana Abul Kalam Azad University of Technology, West Bengal, 700064, India
| | - Sumel Ashique
- Department of Pharmaceutical Sciences, Bengal College of Pharmaceutical Sciences & Research, Durgapur, 713212, West Bengal, India
| | - Radheshyam Pal
- Department of Pharmaceutical Sciences, Bengal College of Pharmaceutical Sciences & Research, Durgapur, 713212, West Bengal, India
| |
Collapse
|
14
|
Lin J, Chen J, Huang C. Systematic identification of key basement membrane related genes as potential new biomarkers in Alzheimer's disease. Clin Neurol Neurosurg 2024; 236:108094. [PMID: 38154381 DOI: 10.1016/j.clineuro.2023.108094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 12/13/2023] [Accepted: 12/17/2023] [Indexed: 12/30/2023]
Abstract
OBJECTIVE The study aimed to identify biomarkers associated with basement membranes (BMs)-related genes (BMGs) in Alzheimer's disease (AD) and investigate their potential role in the progression of AD pathology. METHODS Gene expression profiles were retrieved from Gene Expression Omnibus database. 222 human BMGs were collected from the relevant literature. Subsequently, the differentially expressed BMGs (DE-BMGs) were filtered, and the key DE-BMGs were identified using weighted gene correlation network analysis (WGCNA) and two machine learning algorithms. The expression levels, diagnostic values, clinical significances, enrichment analyses and regulatory networks of these candidate biomarkers were further examined. RESULTS A total of 44 DE-BMGs were acquired by comparing AD temporal cortex with nondemented controls. Using WGCNA and machine learning, versiscan (VCAN), tissue inhibitor of metalloproteinase 1 (TIMP1), structural maintenance of chromosome 3 (SMC3), and laminin β2 (LAMB2) were ultimately identified as candidate biomarkers, and they were verified in a murine model. These biomarkers had high diagnostic value (area under the curve (AUC)>0.8). The diagnostic value of the four gene combination was then evaluated in multiple databases, yielding AUCs ranging from 0.688 to 1. Furthermore, a meaningful correlation between these biomarkers and AD pathology progression was observed. Finally, comprehensive analyses involving Hallmark pathway enrichment, immune cell infiltration analysis, transcriptional regulatory, and competitive endogenous RNA networks indicated that key DE-BMGs closely correlated with oxidative stress and immune dysfunction. CONCLUSION Our study comprehensively identified four candidate BMGs and their combination model that play a crucial part in the diagnosis and pathogenesis of AD.
Collapse
Affiliation(s)
- Jia'xing Lin
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Jing Chen
- Department of Rheumatology, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Cheng Huang
- Department of Neurology, Clinical Neuroscience Institute, The First Affiliated Hospital, Jinan University, Guangzhou, China
| |
Collapse
|
15
|
Mir FA, Amanullah A, Jain BP, Hyderi Z, Gautam A. Neuroepigenetics of ageing and neurodegeneration-associated dementia: An updated review. Ageing Res Rev 2023; 91:102067. [PMID: 37689143 DOI: 10.1016/j.arr.2023.102067] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 09/01/2023] [Accepted: 09/06/2023] [Indexed: 09/11/2023]
Abstract
Gene expression is tremendously altered in the brain during memory acquisition, recall, and forgetfulness. However, non-genetic factors, including environmental elements, epigenetic changes, and lifestyle, have grabbed significant attention in recent years regarding the etiology of neurodegenerative diseases (NDD) and age-associated dementia. Epigenetic modifications are essential in regulating gene expression in all living organisms in a DNA sequence-independent manner. The genes implicated in ageing and NDD-related memory disorders are epigenetically regulated by processes such as DNA methylation, histone acetylation as well as messenger RNA editing machinery. The physiological and optimal state of the epigenome, especially within the CNS of humans, plays an intricate role in helping us adjust to the changing environment, and alterations in it cause many brain disorders, but the mechanisms behind it still need to be well understood. When fully understood, these epigenetic landscapes could act as vital targets for pharmacogenetic rescue strategies for treating several diseases, including neurodegeneration- and age-induced dementia. Keeping this objective in mind, this updated review summarises the epigenetic changes associated with age and neurodegeneration-associated dementia.
Collapse
Affiliation(s)
- Fayaz Ahmad Mir
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | - Zeeshan Hyderi
- Department of Biotechnology, Alagappa University, Karaikudi, India
| | - Akash Gautam
- Centre for Neural and Cognitive Sciences, University of Hyderabad, Hyderabad, India.
| |
Collapse
|
16
|
Lepolard C, Rombaut C, Jaouen F, Borges A, Caccomo-Garcia E, Popa N, Gascon E. Optimized miR-124 reporters uncover differences in miR-124 expression among neuronal populations in vitro. Front Neurosci 2023; 17:1257599. [PMID: 37920296 PMCID: PMC10619730 DOI: 10.3389/fnins.2023.1257599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 09/26/2023] [Indexed: 11/04/2023] Open
Abstract
Introduction Although intensively studied in the last decades, how microRNAs (miRNAs) are expressed across different cell types in the brain remains largely unknown. Materials To address this issue, we sought to develop optimized fluorescence reporters that could be expressed in precise cellular subsets and used to accurately quantify miR contents in vivo. Results Focusing on miR-124, we tested different reporter designs whose efficiency was confirmed in different in vitro settings including cell lines and primary neuronal cultures from different brain structures. Unlike previous reporters, we provide experimental evidence that our optimized designs can faithfully translate miR levels in vitro. Discussion Tools developed here would enable assessing miRNA expression at the single cell resolution and are expected to significantly contribute to future miRNA research in vivo.
Collapse
Affiliation(s)
- Catherine Lepolard
- Aix Marseille University, CNRS, INT, Institute of Neuroscience of la Timone, Marseille, France
| | - Cynthia Rombaut
- Aix Marseille University, CNRS, INT, Institute of Neuroscience of la Timone, Marseille, France
| | - Florence Jaouen
- Aix Marseille University, CNRS, INT, Institute of Neuroscience of la Timone, Marseille, France
- Neurobiotools Facility (Neurovir), Aix Marseille University, CNRS, INT, Institute of Neuroscience of la Timone, Marseille, France
| | - Ana Borges
- Aix Marseille University, CNRS, INT, Institute of Neuroscience of la Timone, Marseille, France
| | - Elodie Caccomo-Garcia
- Aix Marseille University, CNRS, INT, Institute of Neuroscience of la Timone, Marseille, France
| | - Natalia Popa
- Aix Marseille University, CNRS, INT, Institute of Neuroscience of la Timone, Marseille, France
| | - Eduardo Gascon
- Aix Marseille University, CNRS, INT, Institute of Neuroscience of la Timone, Marseille, France
| |
Collapse
|
17
|
Zuniga G, Frost B. Selective neuronal vulnerability to deficits in RNA processing. Prog Neurobiol 2023; 229:102500. [PMID: 37454791 DOI: 10.1016/j.pneurobio.2023.102500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/30/2023] [Accepted: 07/10/2023] [Indexed: 07/18/2023]
Abstract
Emerging evidence indicates that errors in RNA processing can causally drive neurodegeneration. Given that RNA produced from expressed genes of all cell types undergoes processing (splicing, polyadenylation, 5' capping, etc.), the particular vulnerability of neurons to deficits in RNA processing calls for careful consideration. The activity-dependent transcriptome remodeling associated with synaptic plasticity in neurons requires rapid, multilevel post-transcriptional RNA processing events that provide additional opportunities for dysregulation and consequent introduction or persistence of errors in RNA transcripts. Here we review the accumulating evidence that neurons have an enhanced propensity for errors in RNA processing alongside grossly insufficient defenses to clear misprocessed RNA compared to other cell types. Additionally, we explore how tau, a microtubule-associated protein implicated in Alzheimer's disease and related tauopathies, contributes to deficits in RNA processing and clearance.
Collapse
Affiliation(s)
- Gabrielle Zuniga
- Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, San Antonio, TX, USA; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health San Antonio, San Antonio, TX, USA; Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Bess Frost
- Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, San Antonio, TX, USA; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health San Antonio, San Antonio, TX, USA; Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, TX, USA.
| |
Collapse
|
18
|
Singh R, Hussain J, Kaur A, Jamdare BG, Pathak D, Garg K, Kaur R, Shankar S, Sunkaria A. The hidden players: Shedding light on the significance of post-translational modifications and miRNAs in Alzheimer's disease development. Ageing Res Rev 2023; 90:102002. [PMID: 37423542 DOI: 10.1016/j.arr.2023.102002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/29/2023] [Accepted: 07/03/2023] [Indexed: 07/11/2023]
Abstract
Alzheimer's disease (AD) is the most prevalent, expensive, lethal, and burdening neurodegenerative disease of this century. The initial stages of this disease are characterized by a reduced ability to encode and store new memories. Subsequent cognitive and behavioral deterioration occurs during the later stages. Abnormal cleavage of amyloid precursor protein (APP) resulting in amyloid-beta (Aβ) accumulation along with hyperphosphorylation of tau protein are the two characteristic hallmarks of AD. Recently, several post-translational modifications (PTMs) have been identified on both Aβ as well as tau proteins. However, a complete understanding of how different PTMs influence the structure and function of proteins in both healthy and diseased conditions is still lacking. It has been speculated that these PTMs might play vital roles in the progression of AD. In addition, several short non-coding microRNA (miRNA) sequences have been found to be deregulated in the peripheral blood of Alzheimer patients. The miRNAs are single-stranded RNAs that control gene expression by causing mRNA degradation, deadenylation, or translational repression and have been implicated in the regulation of several neuronal and glial activities. The lack of comprehensive understanding regarding disease mechanisms, biomarkers, and therapeutic targets greatly hampers the development of effective strategies for early diagnosis and the identification of viable therapeutic targets. Moreover, existing treatment options for managing the disease have proven to be ineffective and provide only temporary relief. Therefore, understanding the role of miRNAs and PTMs in AD can provide valuable insights into disease mechanisms, aid in the identification of biomarkers, facilitate the discovery of novel therapeutic targets, and inspire innovative treatments for this challenging condition.
Collapse
Affiliation(s)
- Ravinder Singh
- Department of Biotechnology, Guru Nanak Dev University, Amritsar 143005, Punjab, India
| | - Julfequar Hussain
- Department of Biotechnology, Guru Nanak Dev University, Amritsar 143005, Punjab, India
| | - Amandeep Kaur
- Department of Biotechnology, Guru Nanak Dev University, Amritsar 143005, Punjab, India
| | - Balaji Gokul Jamdare
- Department of Biotechnology, Guru Nanak Dev University, Amritsar 143005, Punjab, India
| | - Deepti Pathak
- Department of Biotechnology, Guru Nanak Dev University, Amritsar 143005, Punjab, India
| | - Kanchan Garg
- Department of Biotechnology, Guru Nanak Dev University, Amritsar 143005, Punjab, India
| | - Ramanpreet Kaur
- Department of Biotechnology, Guru Nanak Dev University, Amritsar 143005, Punjab, India
| | - Shivani Shankar
- Department of Biotechnology, Guru Nanak Dev University, Amritsar 143005, Punjab, India
| | - Aditya Sunkaria
- Department of Biotechnology, Guru Nanak Dev University, Amritsar 143005, Punjab, India.
| |
Collapse
|
19
|
Kunze R, Fischer S, Marti HH, Preissner KT. Brain alarm by self-extracellular nucleic acids: from neuroinflammation to neurodegeneration. J Biomed Sci 2023; 30:64. [PMID: 37550658 PMCID: PMC10405513 DOI: 10.1186/s12929-023-00954-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 07/22/2023] [Indexed: 08/09/2023] Open
Abstract
Neurological disorders such as stroke, multiple sclerosis, as well as the neurodegenerative diseases Parkinson's or Alzheimer's disease are accompanied or even powered by danger associated molecular patterns (DAMPs), defined as endogenous molecules released from stressed or damaged tissue. Besides protein-related DAMPs or "alarmins", numerous nucleic acid DAMPs exist in body fluids, such as cell-free nuclear and mitochondrial DNA as well as different species of extracellular RNA, collectively termed as self-extracellular nucleic acids (SENAs). Among these, microRNA, long non-coding RNAs, circular RNAs and extracellular ribosomal RNA constitute the majority of RNA-based DAMPs. Upon tissue injury, necrosis or apoptosis, such SENAs are released from neuronal, immune and other cells predominantly in association with extracellular vesicles and may be translocated to target cells where they can induce intracellular regulatory pathways in gene transcription and translation. The majority of SENA-induced signaling reactions in the brain appear to be related to neuroinflammatory processes, often causally associated with the onset or progression of the respective disease. In this review, the impact of the diverse types of SENAs on neuroinflammatory and neurodegenerative diseases will be discussed. Based on the accumulating knowledge in this field, several specific antagonistic approaches are presented that could serve as therapeutic interventions to lower the pathological outcome of the indicated brain disorders.
Collapse
Affiliation(s)
- Reiner Kunze
- Institute of Physiology and Pathophysiology, Department of Cardiovascular Physiology, Ruprecht-Karls-University, Im Neuenheimer Feld 326, 69120 Heidelberg, Germany
| | - Silvia Fischer
- Department of Biochemistry, Medical School, Justus-Liebig-University, Giessen, Germany
| | - Hugo H. Marti
- Institute of Physiology and Pathophysiology, Department of Cardiovascular Physiology, Ruprecht-Karls-University, Im Neuenheimer Feld 326, 69120 Heidelberg, Germany
| | - Klaus T. Preissner
- Department of Biochemistry, Medical School, Justus-Liebig-University, Giessen, Germany
- Kerckhoff-Heart-Research-Institute, Department of Cardiology, Medical School, Justus-Liebig-University, Giessen, Germany
| |
Collapse
|
20
|
Datta N, Johnson C, Kao D, Gurnani P, Alexander C, Polytarchou C, Monaghan TM. MicroRNA-based therapeutics for inflammatory disorders of the microbiota-gut-brain axis. Pharmacol Res 2023; 194:106870. [PMID: 37499702 DOI: 10.1016/j.phrs.2023.106870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 07/22/2023] [Accepted: 07/24/2023] [Indexed: 07/29/2023]
Abstract
An emerging but less explored shared pathophysiology across microbiota-gut-brain axis disorders is aberrant miRNA expression, which may represent novel therapeutic targets. miRNAs are small, endogenous non-coding RNAs that are important transcriptional repressors of gene expression. Most importantly, they regulate the integrity of the intestinal epithelial and blood-brain barriers and serve as an important communication channel between the gut microbiome and the host. A well-defined understanding of the mode of action, therapeutic strategies and delivery mechanisms of miRNAs is pivotal in translating the clinical applications of miRNA-based therapeutics. Accumulating evidence links disorders of the microbiota-gut-brain axis with a compromised gut-blood-brain-barrier, causing gut contents such as immune cells and microbiota to enter the bloodstream leading to low-grade systemic inflammation. This has the potential to affect all organs, including the brain, causing central inflammation and the development of neurodegenerative and neuropsychiatric diseases. In this review, we have examined in detail miRNA biogenesis, strategies for therapeutic application, delivery mechanisms, as well as their pathophysiology and clinical applications in inflammatory gut-brain disorders. The research data in this review was drawn from the following databases: PubMed, Google Scholar, and Clinicaltrials.gov. With increasing evidence of the pathophysiological importance for miRNAs in microbiota-gut-brain axis disorders, therapeutic targeting of cross-regulated miRNAs in these disorders displays potentially transformative and translational potential. Further preclinical research and human clinical trials are required to further advance this area of research.
Collapse
Affiliation(s)
- Neha Datta
- School of Medicine, University of Nottingham, Nottingham, UK
| | - Charlotte Johnson
- NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK; Nottingham Digestive Diseases Centre, School of Medicine, University of Nottingham, Nottingham, UK
| | - Dina Kao
- Division of Gastroenterology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Pratik Gurnani
- Division of Molecular Therapeutics & Formulation, School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Cameron Alexander
- Division of Molecular Therapeutics & Formulation, School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Christos Polytarchou
- Department of Biosciences, John van Geest Cancer Research Centre, School of Science & Technology, Nottingham Trent University, Nottingham, UK.
| | - Tanya M Monaghan
- NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK; Nottingham Digestive Diseases Centre, School of Medicine, University of Nottingham, Nottingham, UK.
| |
Collapse
|
21
|
Irwin AB, Martina V, Jago SCS, Bahabry R, Schreiber AM, Lubin FD. The lncRNA Neat1 is associated with astrocyte reactivity and memory deficits in a mouse model of Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.03.539260. [PMID: 37205548 PMCID: PMC10187170 DOI: 10.1101/2023.05.03.539260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Dysregulation of long non-coding RNAs (lncRNAs) have been associated with Alzheimer's disease (AD). However, the functional role of lncRNAs in AD remains unclear. Here, we report a crucial role for the lncRNA Neat1 in astrocyte dysfunction and memory deficits associated with AD. Transcriptomics analysis show abnormally high expression levels of NEAT1 in the brains of AD patients relative to aged-matched healthy controls, with the most significantly elevated levels in glial cells. In a human transgenic APP-J20 (J20) mouse model of AD, RNA-fluorescent in situ hybridization characterization of Neat1 expression in hippocampal astrocyte versus non-astrocyte cell populations revealed a significant increase in Neat1 expression in astrocytes of male, but not female, mice. This corresponded with increased seizure susceptibility in J20 male mice. Interestingly, Neat1 deficiency in the dCA1 in J20 male mice did not alter seizure threshold. Mechanistically, Neat1 deficiency in the dorsal area CA1 of the hippocampus (dCA1) J20 male mice significantly improved hippocampus-dependent memory. Neat1 deficiency also remarkably reduced astrocyte reactivity markers suggesting that Neat1 overexpression is associated with astrocyte dysfunction induced by hAPP/Aβ in the J20 mice. Together, these findings indicate that abnormal Neat1 overexpression may contribute to memory deficits in the J20 AD model not through altered neuronal activity, but through astrocyte dysfunction.
Collapse
Affiliation(s)
- Ashleigh B Irwin
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294 USA
| | - Verdion Martina
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294 USA
| | - Silvienne C Sint Jago
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294 USA
| | - Rudhab Bahabry
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294 USA
| | - Anna Maria Schreiber
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294 USA
| | - Farah D. Lubin
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294 USA
| |
Collapse
|
22
|
Xiong W, Li D, Feng Y, Jia C, Zhang X, Liu Z. CircLPAR1 Promotes Neuroinflammation and Oxidative Stress in APP/PS1 Mice by Inhibiting SIRT1/Nrf-2/HO-1 Axis Through Destabilizing GDF-15 mRNA. Mol Neurobiol 2023; 60:2236-2251. [PMID: 36646968 DOI: 10.1007/s12035-022-03177-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 12/16/2022] [Indexed: 01/18/2023]
Abstract
Circular RNA LPAR1 (circLPAR1) was revealed to be elevated in Alzheimer's disease (AD); nevertheless, its role and mechanisms in AD remain unknown. Memory performance of APP/PS1 mice was assessed by Morris water maze test. Expression of circLPAR1 and indicated messenger RNA (mRNA) in mouse brain tissues or/and SH-SY5Y cells were tested by quantitative real-time PCR (qRT-PCR). Protein expression of indicated gene was examined by western blot. Production of proinflammatory cytokines (tumor necrosis factor-α, TNF-α; interleukin-6, IL-6; interleukin-1β, IL-1β; and interleukin-8, IL-8) and oxidative stress-related factors (reactive oxygen species, ROS; malondialdehyde, MDA; superoxide dismutase, SOD; and glutathione, GSH) were assessed by commercial kits. RNA pull down and RNA immunoprecipitation were performed to verify the interplay between up-frameshift protein 1 (UPF1) and circLPAR1 or growth differentiation factor 15 (GDF-15). CircLPAR1 was elevated, while GDF-15 was decreased in both APP/PS1 mice and Aβ-treated SH-SY5Y cells. Knockdown of circLPAR1 and overexpression of GDF-15 protected cells against Aβ-caused inflammation, oxidative stress, and neuronal apoptosis. CircLPAR1 knockdown was also proved to improve AD-related pathological traits and ameliorate cognitive dysfunctions in vivo. In mechanism, we found that circLPAR1 repressed GDF-15 expression by decreasing GDF-15 mRNA stability through UPF1 recruitment. Rescue assays suggested that sirtuin 1 (SIRT1) knockdown reversed GDF-15 overexpression-induced inhibition on Aβ-induced neuronal damage and nuclear factor E2-related factor (Nrf-2)/heme oxygenase-1 (HO-1) pathway inhibition. Moreover, the protective effect of circLPAR1 knockdown against Aβ-induced apoptosis was abolished by GDF-15 knockdown, and SIRT1 overexpression could counteract this effect of GDF-15 knockdown. CircLPAR1 knockdown improved AD-related pathological traits in vitro and in vivo by inhibiting SIRT1/Nrf-2/HO-1 axis through GDF-15.
Collapse
Affiliation(s)
- Wenping Xiong
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, No. 169, East Lake Road, Wuchang District, Wuhan, 430071, Hubei Province, People's Republic of China
| | - Dongming Li
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei Province, People's Republic of China
| | - Yu Feng
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, No. 169, East Lake Road, Wuchang District, Wuhan, 430071, Hubei Province, People's Republic of China
| | - Chenguang Jia
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, No. 169, East Lake Road, Wuchang District, Wuhan, 430071, Hubei Province, People's Republic of China
| | - Xiangyu Zhang
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, No. 169, East Lake Road, Wuchang District, Wuhan, 430071, Hubei Province, People's Republic of China
| | - Zheng Liu
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, No. 169, East Lake Road, Wuchang District, Wuhan, 430071, Hubei Province, People's Republic of China.
| |
Collapse
|
23
|
Balmorez T, Sakazaki A, Murakami S. Genetic Networks of Alzheimer's Disease, Aging, and Longevity in Humans. Int J Mol Sci 2023; 24:ijms24065178. [PMID: 36982253 PMCID: PMC10049434 DOI: 10.3390/ijms24065178] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 03/01/2023] [Accepted: 03/04/2023] [Indexed: 03/30/2023] Open
Abstract
Human genomic analysis and genome-wide association studies (GWAS) have identified genes that are risk factors for early and late-onset Alzheimer's disease (AD genes). Although the genetics of aging and longevity have been extensively studied, previous studies have focused on a specific set of genes that have been shown to contribute to or are a risk factor for AD. Thus, the connections among the genes involved in AD, aging, and longevity are not well understood. Here, we identified the genetic interaction networks (referred to as pathways) of aging and longevity within the context of AD by using a gene set enrichment analysis by Reactome that cross-references more than 100 bioinformatic databases to allow interpretation of the biological functions of gene sets through a wide variety of gene networks. We validated the pathways with a threshold of p-value < 1.00 × 10-5 using the databases to extract lists of 356 AD genes, 307 aging-related (AR) genes, and 357 longevity genes. There was a broad range of biological pathways involved in AR and longevity genes shared with AD genes. AR genes identified 261 pathways within the threshold of p < 1.00 × 10-5, of which 26 pathways (10% of AR gene pathways) were further identified by overlapping genes among AD and AR genes. The overlapped pathways included gene expression (p = 4.05 × 10-11) including ApoE, SOD2, TP53, and TGFB1 (p = 2.84 × 10-10); protein metabolism and SUMOylation, including E3 ligases and target proteins (p = 1.08 × 10-7); ERBB4 signal transduction (p = 2.69 × 10-6); the immune system, including IL-3 and IL-13 (p = 3.83 × 10-6); programmed cell death (p = 4.36 × 10-6); and platelet degranulation (p = 8.16 × 10-6), among others. Longevity genes identified 49 pathways within the threshold, of which 12 pathways (24% of longevity gene pathways) were further identified by overlapping genes among AD and longevity genes. They include the immune system, including IL-3 and IL-13 (p = 7.64 × 10-8), plasma lipoprotein assembly, remodeling and clearance (p < 4.02 × 10-6), and the metabolism of fat-soluble vitamins (p = 1.96 × 10-5). Thus, this study provides shared genetic hallmarks of aging, longevity, and AD backed up by statistical significance. We discuss the significant genes involved in these pathways, including TP53, FOXO, SUMOylation, IL4, IL6, APOE, and CEPT, and suggest that mapping the gene network pathways provide a useful basis for further medical research on AD and healthy aging.
Collapse
Affiliation(s)
- Timothy Balmorez
- Department of Basic Sciences, College of Osteopathic Medicine, Touro University California, Vallejo, CA 94592, USA
| | - Amy Sakazaki
- Department of Basic Sciences, College of Osteopathic Medicine, Touro University California, Vallejo, CA 94592, USA
| | - Shin Murakami
- Department of Basic Sciences, College of Osteopathic Medicine, Touro University California, Vallejo, CA 94592, USA
| |
Collapse
|
24
|
Song S, Lee JU, Jeon MJ, Kim S, Lee CN, Sim SJ. Precise profiling of exosomal biomarkers via programmable curved plasmonic nanoarchitecture-based biosensor for clinical diagnosis of Alzheimer's disease. Biosens Bioelectron 2023; 230:115269. [PMID: 37001292 DOI: 10.1016/j.bios.2023.115269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 03/20/2023] [Accepted: 03/26/2023] [Indexed: 03/30/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease of complex pathogenesis, with overt symptoms following disease progression. Early AD diagnosis is challenging due to the lack of robust biomarkers and limited patient access to diagnostics via neuroimaging and cerebrospinal fluid (CSF) tests. Exosomes present in body fluids are attracting attention as diagnostic biomarkers that directly reflect neuropathological features within the brain. In particular, exosomal miRNAs (exomiRs) signatures are involved in AD pathogenesis, showing a different expression between patients and the healthy controls (HCs). However, low yield and high homologous nature impede the accuracy and reproducibility of exosome blood-based AD diagnostics. Here, we developed a programmable curved plasmonic nanoarchitecture-based biosensor to analyze exomiRs in clinical serum samples for accurate AD diagnosis. To allow the detection of exomiRs in serum at attomolar levels, nanospaces (e.g., nanocrevice and nanocavity) were introduced into the nanostructures to dramatically increase the spectral sensitivity by adjusting the bending angle of the plasmonic nanostructure through sodium chloride concentration control. The developed biosensor classifies individuals into AD, mild cognitive impairment (MCI) patients, and HCs through profiling and quantifying exomiRs. Furthermore, integrating analysis expression patterns of multiple exosomal biomarkers improved serum-based diagnostic performance (average accuracy of 98.22%). Therefore, precise, highly sensitive serum-derived exosomal biomarker detection-based plasmonic biosensor has a robust capacity to predict the molecular pathologic of neurodegenerative disease, progression of cognitive decline, MCI/AD conversion, as well as early diagnosis and treatment.
Collapse
|
25
|
Sun X, Deng Y, Ge P, Peng Q, Soufiany I, Zhu L, Duan R. Diminazene Ameliorates Neuroinflammation by Suppression of Astrocytic miRNA-224-5p/NLRP3 Axis in Alzheimer's Disease Model. J Inflamm Res 2023; 16:1639-1652. [PMID: 37092127 PMCID: PMC10120828 DOI: 10.2147/jir.s401385] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 04/12/2023] [Indexed: 04/25/2023] Open
Abstract
Purpose ACE2/Ang(1-7)/Mas Receptor, the momentous component of the renin-angiotensin system, has been shown to be involved in Alzheimer's disease (AD). We had previously found that enhancing brain ACE2 activity ameliorated cognitive impairment and attenuated brain neuroinflammation in SAMP8 mice, an animal model of AD. However, the exact mechanism of action of Diminazene (DIZE) has not been revealed. Methods APP/PS1 mice were injected intraperitoneally with DIZE. Cognitive functions, neuronal and synaptic integrity, and inflammation-related markers were assessed by Morris water maze, Nissl staining, Western blotting and ELISA, respectively. Since astrocytes played a crucial role in AD-related neuroinflammation whilst miRNAs were reported to participate in modulating inflammatory responses, astrocytes of APP/PS1 mice were then isolated for high-throughput miRNAs sequencing to identify the most differentially expressed miRNA following DIZE treatment. Afterward, the downstream pathway of this miRNA in the anti-inflammatory action of DIZE was investigated using primary astrocytes. Results The results showed that DIZE alleviated cognitive impairment and neuronal and synaptic damage in APP/PS1 mice. Simultaneously, DIZE suppressed the secretion of pro-inflammatory cytokines and the expression of NLRP3 inflammasome. Importantly, miR-224-5p was significantly up-regulated in the astrocytes of APP/PS1 mice treated by DIZE, and NLRP3 is one of the targets of miR-224-5p. Upregulation of miR-224-5p inhibited the expression of NLRP3 in Aβ1-42-stimulated cells, whereas miR-224-5p downregulation reversed this effect. Furthermore, the inhibition of miR-224-5p could reverse the inhibitory effect of DIZE on astrocytic NLRP3 inflammasome. Conclusion These findings firstly suggested that DIZE could inhibit astrocyte-regulated neuroinflammation via miRNA-224-5p/NLRP3 pathway. Furthermore, our study reveals the underlying mechanism by which DIZE suppresses neuroinflammatory responses in AD mice and uncovers the potential of DIZE in AD treatment.
Collapse
Affiliation(s)
- XiaoJin Sun
- Faculty of Pharmacy, Bengbu Medical College, Anhui Engineering Technology Research Center of Biochemical Pharmaceuticals, Bengbu, Anhui, People’s Republic of China
| | - Yang Deng
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, People’s Republic of China
| | - PengXin Ge
- Department of Pharmacy, Anhui Provincial Cancer Hospital, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, People’s Republic of China
| | - Qiang Peng
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, People’s Republic of China
| | - Ismatullah Soufiany
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, People’s Republic of China
| | - Lin Zhu
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, People’s Republic of China
- Correspondence: Lin Zhu; Rui Duan, Email ;
| | - Rui Duan
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, People’s Republic of China
| |
Collapse
|
26
|
Kim YA, Siddiqui T, Blaze J, Cosacak MI, Winters T, Kumar A, Tein E, Sproul AA, Teich AF, Bartolini F, Akbarian S, Kizil C, Hargus G, Santa-Maria I. RNA methyltransferase NSun2 deficiency promotes neurodegeneration through epitranscriptomic regulation of tau phosphorylation. Acta Neuropathol 2023; 145:29-48. [PMID: 36357715 PMCID: PMC9807547 DOI: 10.1007/s00401-022-02511-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 10/06/2022] [Accepted: 10/07/2022] [Indexed: 11/12/2022]
Abstract
Epitranscriptomic regulation adds a layer of post-transcriptional control to brain function during development and adulthood. The identification of RNA-modifying enzymes has opened the possibility of investigating the role epitranscriptomic changes play in the disease process. NOP2/Sun RNA methyltransferase 2 (NSun2) is one of the few known brain-enriched methyltransferases able to methylate mammalian non-coding RNAs. NSun2 loss of function due to autosomal-recessive mutations has been associated with neurological abnormalities in humans. Here, we show NSun2 is expressed in adult human neurons in the hippocampal formation and prefrontal cortex. Strikingly, we unravel decreased NSun2 protein expression and an increased ratio of pTau/NSun2 in the brains of patients with Alzheimer's disease (AD) as demonstrated by Western blotting and immunostaining, respectively. In a well-established Drosophila melanogaster model of tau-induced toxicity, reduction of NSun2 exacerbated tau toxicity, while overexpression of NSun2 partially abrogated the toxic effects. Conditional ablation of NSun2 in the mouse brain promoted a decrease in the miR-125b m6A levels and tau hyperphosphorylation. Utilizing human induced pluripotent stem cell (iPSC)-derived neuronal cultures, we confirmed NSun2 deficiency results in tau hyperphosphorylation. We also found that neuronal NSun2 levels decrease in response to amyloid-beta oligomers (AβO). Notably, AβO-induced tau phosphorylation and cell toxicity in human neurons could be rescued by overexpression of NSun2. Altogether, these results indicate that neuronal NSun2 deficiency promotes dysregulation of miR-125b and tau phosphorylation in AD and highlights a novel avenue for therapeutic targeting.
Collapse
Affiliation(s)
- Yoon A Kim
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, USA
- Department of Pathology and Cell Biology, Columbia University, New York, USA
| | - Tohid Siddiqui
- German Center for Neurodegenerative Diseases (DZNE), Helmholtz Association, Dresden, Germany
| | - Jennifer Blaze
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Mehmet Ilyas Cosacak
- German Center for Neurodegenerative Diseases (DZNE), Helmholtz Association, Dresden, Germany
| | - Tristan Winters
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, USA
- Department of Pathology and Cell Biology, Columbia University, New York, USA
| | - Atul Kumar
- Department of Pathology and Cell Biology, Columbia University, New York, USA
| | - Ellen Tein
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, USA
| | - Andrew A Sproul
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, USA
- Department of Pathology and Cell Biology, Columbia University, New York, USA
| | - Andrew F Teich
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, USA
- Department of Pathology and Cell Biology, Columbia University, New York, USA
| | - Francesca Bartolini
- Department of Pathology and Cell Biology, Columbia University, New York, USA
| | - Schahram Akbarian
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Caghan Kizil
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, USA
- German Center for Neurodegenerative Diseases (DZNE), Helmholtz Association, Dresden, Germany
- Department of Neurology, College of Physicians and Surgeons, Columbia University and the New York Presbyterian Hospital, New York, USA
| | - Gunnar Hargus
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, USA.
- Department of Pathology and Cell Biology, Columbia University, New York, USA.
| | - Ismael Santa-Maria
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, USA.
- Department of Pathology and Cell Biology, Columbia University, New York, USA.
- Facultad de Ciencias Experimentales, Universidad Francisco de Vitoria, Edificio E, Pozuelo de Alarcón, Madrid, 28223, Spain.
| |
Collapse
|
27
|
Hajjo R, Sabbah DA, Abusara OH, Al Bawab AQ. A Review of the Recent Advances in Alzheimer's Disease Research and the Utilization of Network Biology Approaches for Prioritizing Diagnostics and Therapeutics. Diagnostics (Basel) 2022; 12:diagnostics12122975. [PMID: 36552984 PMCID: PMC9777434 DOI: 10.3390/diagnostics12122975] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 11/16/2022] [Accepted: 11/18/2022] [Indexed: 11/29/2022] Open
Abstract
Alzheimer's disease (AD) is a polygenic multifactorial neurodegenerative disease that, after decades of research and development, is still without a cure. There are some symptomatic treatments to manage the psychological symptoms but none of these drugs can halt disease progression. Additionally, over the last few years, many anti-AD drugs failed in late stages of clinical trials and many hypotheses surfaced to explain these failures, including the lack of clear understanding of disease pathways and processes. Recently, different epigenetic factors have been implicated in AD pathogenesis; thus, they could serve as promising AD diagnostic biomarkers. Additionally, network biology approaches have been suggested as effective tools to study AD on the systems level and discover multi-target-directed ligands as novel treatments for AD. Herein, we provide a comprehensive review on Alzheimer's disease pathophysiology to provide a better understanding of disease pathogenesis hypotheses and decipher the role of genetic and epigenetic factors in disease development and progression. We also provide an overview of disease biomarkers and drug targets and suggest network biology approaches as new tools for identifying novel biomarkers and drugs. We also posit that the application of machine learning and artificial intelligence to mining Alzheimer's disease multi-omics data will facilitate drug and biomarker discovery efforts and lead to effective individualized anti-Alzheimer treatments.
Collapse
Affiliation(s)
- Rima Hajjo
- Department of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah University of Jordan, P.O. Box 130, Amman 11733, Jordan
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, The University of North Carlina at Chapel Hill, Chapel Hill, NC 27599, USA
- National Center for Epidemics and Communicable Disease Control, Amman 11118, Jordan
- Correspondence:
| | - Dima A. Sabbah
- Department of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah University of Jordan, P.O. Box 130, Amman 11733, Jordan
| | - Osama H. Abusara
- Department of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah University of Jordan, P.O. Box 130, Amman 11733, Jordan
| | - Abdel Qader Al Bawab
- Department of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah University of Jordan, P.O. Box 130, Amman 11733, Jordan
| |
Collapse
|
28
|
Aerqin Q, Wang ZT, Wu KM, He XY, Dong Q, Yu JT. Omics-based biomarkers discovery for Alzheimer's disease. Cell Mol Life Sci 2022; 79:585. [PMID: 36348101 DOI: 10.1007/s00018-022-04614-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 10/22/2022] [Accepted: 10/26/2022] [Indexed: 11/09/2022]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorders presenting with the pathological hallmarks of amyloid plaques and tau tangles. Over the past few years, great efforts have been made to explore reliable biomarkers of AD. High-throughput omics are a technology driven by multiple levels of unbiased data to detect the complex etiology of AD, and it provides us with new opportunities to better understand the pathophysiology of AD and thereby identify potential biomarkers. Through revealing the interaction networks between different molecular levels, the ultimate goal of multi-omics is to improve the diagnosis and treatment of AD. In this review, based on the current AD pathology and the current status of AD diagnostic biomarkers, we summarize how genomics, transcriptomics, proteomics and metabolomics are all conducing to the discovery of reliable AD biomarkers that could be developed and used in clinical AD management.
Collapse
Affiliation(s)
- Qiaolifan Aerqin
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, 200040, China
| | - Zuo-Teng Wang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Kai-Min Wu
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, 200040, China
| | - Xiao-Yu He
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, 200040, China
| | - Qiang Dong
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, 200040, China
| | - Jin-Tai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, 200040, China.
| |
Collapse
|
29
|
Kalra RS, Vijayan M, Ghosal S. Editorial: Non-coding RNA in Alzheimer's pathology and diagnosis. Front Aging Neurosci 2022; 14:1030742. [PMID: 36204555 PMCID: PMC9531995 DOI: 10.3389/fnagi.2022.1030742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 09/05/2022] [Indexed: 11/13/2022] Open
Affiliation(s)
- Rajkumar Singh Kalra
- Okinawa Institute of Science and Technology, Graduate University, Okinawa, Japan
- *Correspondence: Rajkumar Singh Kalra
| | - Murali Vijayan
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Murali Vijayan
| | - Suman Ghosal
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
30
|
Profile of TREM2-Derived circRNA and mRNA Variants in the Entorhinal Cortex of Alzheimer’s Disease Patients. Int J Mol Sci 2022; 23:ijms23147682. [PMID: 35887031 PMCID: PMC9320643 DOI: 10.3390/ijms23147682] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/04/2022] [Accepted: 07/04/2022] [Indexed: 02/04/2023] Open
Abstract
Genetic variants in TREM2, a microglia-related gene, are well-known risk factors for Alzheimer’s disease (AD). Here, we report that TREM2 originates from circular RNAs (circRNAs), a novel class of non-coding RNAs characterized by a covalent and stable closed-loop structure. First, divergent primers were designed to amplify circRNAs by RT-PCR, which were further assessed by Sanger sequencing. Then, additional primer sets were used to confirm back-splicing junctions. In addition, HMC3 cells were used to assess the microglial expression of circTREM2s. Three candidate circTREM2s were identified in control and AD human entorhinal samples. One of the circRNAs, circTREM2_1, was consistently amplified by all divergent primer sets in control and AD entorhinal cortex samples as well as in HMC3 cells. In AD cases, a moderate negative correlation (r = −0.434) was found between the global average area of Aβ deposits in the entorhinal cortex and circTREM2_1 expression level. In addition, by bioinformatics tools, a total of 16 miRNAs were predicted to join with circTREM2s. Finally, TREM2 mRNA corresponding to four isoforms was profiled by RT-qPCR. TREM2 mRNA levels were found elevated in entorhinal samples of AD patients with low or intermediate ABC scores compared to controls. To sum up, a novel circRNA derived from the TREM2 gene, circTREM2_1, has been identified in the human entorhinal cortex and TREM2 mRNA expression has been detected to increase in AD compared to controls. Unraveling the molecular genetics of the TREM2 gene may help to better know the innate immune response in AD.
Collapse
|
31
|
The Role of Non-Coding RNAs in the Pathogenesis of Parkinson’s Disease: Recent Advancement. Pharmaceuticals (Basel) 2022; 15:ph15070811. [PMID: 35890110 PMCID: PMC9315906 DOI: 10.3390/ph15070811] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/19/2022] [Accepted: 06/21/2022] [Indexed: 01/27/2023] Open
Abstract
Parkinson’s disease (PD) is a prevalent neurodegenerative aging disorder that manifests as motor and non-motor symptoms, and its etiopathogenesis is influenced by non-coding RNAs (ncRNAs). Signal pathway and gene sequence studies have proposed that alteration of ncRNAs is relevant to the occurrence and development of PD. Furthermore, many studies on brain tissues and body fluids from patients with PD indicate that variations in ncRNAs and their target genes could trigger or exacerbate neurodegenerative pathogenesis and serve as potential non-invasive biomarkers of PD. Numerous ncRNAs have been considered regulators of apoptosis, α-syn misfolding and aggregation, mitochondrial dysfunction, autophagy, and neuroinflammation in PD etiology, and evidence is mounting for the determination of the role of competing endogenous RNA (ceRNA) mechanisms in disease development. In this review, we discuss the current knowledge regarding the regulation and function of ncRNAs as well as ceRNA networks in PD pathogenesis, focusing on microRNAs, long ncRNAs, and circular RNAs to increase the understanding of the disease and propose potential target identification and treatment in the early stages of PD.
Collapse
|
32
|
Alvia M, Aytan N, Spencer KR, Foster ZW, Rauf NA, Guilderson L, Robey I, Averill JG, Walker SE, Alvarez VE, Huber BR, Mathais R, Cormier KA, Nicks R, Pothast M, Labadorf A, Agus F, Alosco ML, Mez J, Kowall NW, McKee AC, Brady CB, Stein TD. MicroRNA Alterations in Chronic Traumatic Encephalopathy and Amyotrophic Lateral Sclerosis. Front Neurosci 2022; 16:855096. [PMID: 35663558 PMCID: PMC9160996 DOI: 10.3389/fnins.2022.855096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 03/09/2022] [Indexed: 11/13/2022] Open
Abstract
Repetitive head impacts (RHI) and traumatic brain injuries are risk factors for the neurodegenerative diseases chronic traumatic encephalopathy (CTE) and amyotrophic lateral sclerosis (ALS). ALS and CTE are distinct disorders, yet in some instances, share pathology, affect similar brain regions, and occur together. The pathways involved and biomarkers for diagnosis of both diseases are largely unknown. MicroRNAs (miRNAs) involved in gene regulation may be altered in neurodegeneration and be useful as stable biomarkers. Thus, we set out to determine associations between miRNA levels and disease state within the prefrontal cortex in a group of brain donors with CTE, ALS, CTE + ALS and controls. Of 47 miRNAs previously implicated in neurological disease and tested here, 28 (60%) were significantly different between pathology groups. Of these, 21 (75%) were upregulated in both ALS and CTE, including miRNAs involved in inflammatory, apoptotic, and cell growth/differentiation pathways. The most significant change occurred in miR-10b, which was significantly increased in ALS, but not CTE or CTE + ALS. Overall, we found patterns of miRNA expression that are common and unique to CTE and ALS and that suggest shared and distinct mechanisms of pathogenesis.
Collapse
Affiliation(s)
- Marcela Alvia
- Boston University Alzheimer’s Disease Research Center, Boston University CTE Center, Boston University School of Medicine, Boston, MA, United States
| | - Nurgul Aytan
- Boston University Alzheimer’s Disease Research Center, Boston University CTE Center, Boston University School of Medicine, Boston, MA, United States
- Department of Neurology, Boston University School of Medicine, Boston, MA, United States
| | | | | | | | | | - Ian Robey
- Southern Arizona VA Healthcare System, Tucson, AZ, United States
| | - James G. Averill
- Southern Arizona VA Healthcare System, Tucson, AZ, United States
| | - Sean E. Walker
- Southern Arizona VA Healthcare System, Tucson, AZ, United States
| | - Victor E. Alvarez
- Boston University Alzheimer’s Disease Research Center, Boston University CTE Center, Boston University School of Medicine, Boston, MA, United States
- Department of Neurology, Boston University School of Medicine, Boston, MA, United States
- VA Boston Healthcare System, Boston, MA, United States
- Department of Veterans Affairs Medical Center, Bedford, MA, United States
| | - Bertrand R. Huber
- Boston University Alzheimer’s Disease Research Center, Boston University CTE Center, Boston University School of Medicine, Boston, MA, United States
- Department of Neurology, Boston University School of Medicine, Boston, MA, United States
- VA Boston Healthcare System, Boston, MA, United States
| | - Rebecca Mathais
- Boston University Alzheimer’s Disease Research Center, Boston University CTE Center, Boston University School of Medicine, Boston, MA, United States
| | - Kerry A. Cormier
- Boston University Alzheimer’s Disease Research Center, Boston University CTE Center, Boston University School of Medicine, Boston, MA, United States
- VA Boston Healthcare System, Boston, MA, United States
- Department of Veterans Affairs Medical Center, Bedford, MA, United States
| | - Raymond Nicks
- Boston University Alzheimer’s Disease Research Center, Boston University CTE Center, Boston University School of Medicine, Boston, MA, United States
| | - Morgan Pothast
- Boston University Alzheimer’s Disease Research Center, Boston University CTE Center, Boston University School of Medicine, Boston, MA, United States
| | - Adam Labadorf
- Department of Neurology, Boston University School of Medicine, Boston, MA, United States
- VA Boston Healthcare System, Boston, MA, United States
| | - Filisia Agus
- Department of Neurology, Boston University School of Medicine, Boston, MA, United States
| | - Michael L. Alosco
- Boston University Alzheimer’s Disease Research Center, Boston University CTE Center, Boston University School of Medicine, Boston, MA, United States
- Department of Neurology, Boston University School of Medicine, Boston, MA, United States
| | - Jesse Mez
- Boston University Alzheimer’s Disease Research Center, Boston University CTE Center, Boston University School of Medicine, Boston, MA, United States
- Department of Neurology, Boston University School of Medicine, Boston, MA, United States
| | - Neil W. Kowall
- Boston University Alzheimer’s Disease Research Center, Boston University CTE Center, Boston University School of Medicine, Boston, MA, United States
- Department of Neurology, Boston University School of Medicine, Boston, MA, United States
- VA Boston Healthcare System, Boston, MA, United States
| | - Ann C. McKee
- Boston University Alzheimer’s Disease Research Center, Boston University CTE Center, Boston University School of Medicine, Boston, MA, United States
- Department of Neurology, Boston University School of Medicine, Boston, MA, United States
- VA Boston Healthcare System, Boston, MA, United States
- Department of Veterans Affairs Medical Center, Bedford, MA, United States
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, United States
| | - Christopher B. Brady
- Department of Neurology, Boston University School of Medicine, Boston, MA, United States
- VA Boston Healthcare System, Boston, MA, United States
- Department of Veterans Affairs Medical Center, Bedford, MA, United States
| | - Thor D. Stein
- Boston University Alzheimer’s Disease Research Center, Boston University CTE Center, Boston University School of Medicine, Boston, MA, United States
- VA Boston Healthcare System, Boston, MA, United States
- Department of Veterans Affairs Medical Center, Bedford, MA, United States
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
33
|
Lukiw WJ. Fission Impossible: Stabilized miRNA-Based Analogs in Neurodegenerative Disease. Front Neurosci 2022; 16:875957. [PMID: 35592255 PMCID: PMC9111010 DOI: 10.3389/fnins.2022.875957] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 04/04/2022] [Indexed: 01/18/2023] Open
Affiliation(s)
- Walter J. Lukiw
- LSU Neuroscience Center, Louisiana State University Health Science Center, New Orleans, LA, United States
- Department of Ophthalmology, Louisiana State University Health Science Center, New Orleans, LA, United States
- Department of Neurology, Louisiana State University Health Science Center, New Orleans, LA, United States
- *Correspondence: Walter J. Lukiw
| |
Collapse
|
34
|
Yoon S, Kim SE, Ko Y, Jeong GH, Lee KH, Lee J, Solmi M, Jacob L, Smith L, Stickley A, Carvalho AF, Dragioti E, Kronbichler A, Koyanagi A, Hong SH, Thompson T, Oh H, Salazar de Pablo G, Radua J, Shin JI, Fusar-Poli P. Differential expression of MicroRNAs in Alzheimer's disease: a systematic review and meta-analysis. Mol Psychiatry 2022; 27:2405-2413. [PMID: 35264731 DOI: 10.1038/s41380-022-01476-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 01/24/2022] [Accepted: 02/08/2022] [Indexed: 11/10/2022]
Abstract
Alzheimer's disease (AD) results in progressive cognitive decline owing to the accumulation of amyloid plaques and hyperphosphorylated tau. MicroRNAs (miRNAs) have attracted attention as a putative diagnostic and therapeutic target for neurodegenerative diseases. However, existing meta-analyses on AD and its association with miRNAs have produced inconsistent results. The primary objective of this study is to evaluate the magnitude and consistency of differences in miRNA levels between AD patients, mild cognitive impairment (MCI) patients and healthy controls (HC). Articles investigating miRNA levels in blood, brain tissue, or cerebrospinal fluid (CSF) of AD and MCI patients versus HC were systematically searched in PubMed/Medline from inception to February 16th, 2021. Fixed- and random-effects meta-analyses were complemented with the I2 statistic to measure the heterogeneity, assessment of publication bias, sensitivity subgroup analyses (AD severity, brain region, post-mortem versus ante-mortem specimen for CSF and type of analysis used to quantify miRNA) and functional enrichment pathway analysis. Of the 1512 miRNAs included in 61 articles, 425 meta-analyses were performed on 334 miRNAs. Fifty-six miRNAs were significantly upregulated (n = 40) or downregulated (n = 16) in AD versus HC and all five miRNAs were significantly upregulated in MCI versus HC. Functional enrichment analysis confirmed that pathways related to apoptosis, immune response and inflammation were statistically enriched with upregulated pathways in participants with AD relative to HC. This study confirms that miRNAs' expression is altered in AD and MCI compared to HC. These findings open new diagnostic and therapeutic perspectives for this disorder.
Collapse
Affiliation(s)
- Sojung Yoon
- Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sung Eun Kim
- Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Younhee Ko
- Division of Biomedical Engineering, Hankuk University of Foreign Studies, Kyoungki-do, Republic of Korea
| | - Gwang Hun Jeong
- College of Medicine, Gyeongsang National University, Jinju, Republic of Korea
| | - Keum Hwa Lee
- Department of Pediatrics, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jinhee Lee
- Department of Psychiatry, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - Marco Solmi
- Department of Psychiatry, University of Ottawa, Ontario, ON, Canada.,Department of Mental Health, The Ottawa Hospital, Ontario, ON, Canada.,Clinical Epidemiology Program, Ottawa Hospital Research Institute (OHRI), Ottawa, ON, Canada.,School of Epidemiology and Public Health, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Louis Jacob
- Faculty of Medicine, University of Versailles Saint-Quentin-en-Yvelines, Montigny-le-Bretonneux, France.,Parc Sanitari Sant Joan de Déu/CIBERSAM, Universitat de Barcelona, Fundació Sant Joan de Déu, Sant Boi de Llobregat, Barcelona, Spain
| | - Lee Smith
- Centre for Health, Performance, and Wellbeing, Anglia Ruskin University, Cambridge, UK
| | - Andrew Stickley
- Department of Preventive Intervention for Psychiatric Disorders, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo, Japan.,Stockholm Center for Health and Social Change (SCOHOST), Södertörn University, Huddinge, Sweden
| | - Andre F Carvalho
- Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada.,Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Elena Dragioti
- Pain and Rehabilitation Centre and Department of Health, Medicine and Caring Sciences, Linkoping University, Linkoping, Sweden
| | | | - Ai Koyanagi
- Parc Sanitari Sant Joan de Déu/CIBERSAM, Universitat de Barcelona, Fundació Sant Joan de Déu, Sant Boi de Llobregat, Barcelona, Spain.,ICREA, Pg. Lluis Companys 23, Barcelona, Spain
| | - Sung Hwi Hong
- Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Trevor Thompson
- Centre of Chronic Illness and Ageing, University of Greenwich, London, UK
| | - Hans Oh
- Suzanne Dworak-Peck School of Social Work, University of Southern California, Los Angeles, CA, 90015, USA
| | - Gonzalo Salazar de Pablo
- Early Psychosis: Interventions and Clinical-detection (EPIC) lab, Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK.,Department of Child and Adolescent Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK.,Institute of Psychiatry and Mental Health. Department of Child and Adolescent Psychiatry, Hospital General Universitario Gregorio Marañón School of Medicine, Universidad Complutense, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), CIBERSAM, Madrid, Spain
| | - Joaquim Radua
- Early Psychosis: Interventions and Clinical-detection (EPIC) lab, Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK.,Imaging of Mood- and Anxiety-Related Disorders (IMARD) Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), CIBERSAM, Barcelona, Spain.,Department of Clinical Neuroscience, Centre for Psychiatric Research and Education, Karolinska Institutet, Stockholm, Sweden
| | - Jae Il Shin
- Department of Pediatrics, Yonsei University College of Medicine, Seoul, Republic of Korea.
| | - Paolo Fusar-Poli
- Early Psychosis: Interventions and Clinical-detection (EPIC) lab, Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK.,Department of Brain and Behavioural Sciences, University of Pavia, Pavia, Italy.,OASIS service, South London and Maudsley NHS Foundation Trust, London, UK.,National Institute of Health Research Maudsley Biomedical Research Centre, South London and Maudsley NHS Foundation Trust, London, UK
| |
Collapse
|
35
|
Yan H, Yan Y, Gao Y, Zhang N, Kumar G, Fang Q, Li Z, Li J, Zhang Y, Song L, Wang J, Sun J, Zhang HT, Ma CG. Transcriptome analysis of fasudil treatment in the APPswe/PSEN1dE9 transgenic (APP/PS1) mice model of Alzheimer's disease. Sci Rep 2022; 12:6625. [PMID: 35459923 PMCID: PMC9033779 DOI: 10.1038/s41598-022-10554-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 03/30/2022] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease (AD) is the most common cause of progressive dementia. In the present study, we showed hippocampal tissue transcriptome analysis in APPswe/PSEN1dE9 (APP/PS1, AD model) mice treated with fasudil (ADF) and compared with AD mice treated with saline (ADNS) and wild type mice (WT). The competing endogenous RNA (ceRNA) network was constructed and validated the differential expression of mRNA, lncRNA, miRNA, and circRNA. Our study showed differentially expressed mRNAs (DEMs) between WT and ADNS, while enriched in cell growth and death and nervous system pathways. DEMs between ADNS-ADF were enriched in the nervous system, glycosaminoglycan biosynthesis-keratan sulfate (KS) and Quorum sensing pathways. We validated four genes with RT-PCR, whereas enrichment of Acyl-CoA Synthetase Long Chain Family Member 4 (Acsl4, ENSMUST00000112903) in Quorum sensing pathways, and BTG anti-proliferation factor 1 (Btg1, ENSMUST00000038377) in RNA degradation pathways were conducted. Expression of these two genes were higher in ADNS, but were significantly reduced in ADF. Histone H4 transcription factor (Hinfp, ENSMUST00000216508) orchestrate G1/S transition of mitotic cell cycle and co-expressed with mmu-miR-26a-2-3p-mediated ceRNA and mmu-miR-3065-5p-mediated ceRNA; Wnt family member 4 (Wnt4, ENSMUST00000045747) was enriched in mTOR, Hippo and Wnt signaling pathway. Expression of these two genes were significantly lower in ADNS, and fasudil treatment reverse it. The present studies demonstrated four genes: Acsl4, Btg1, Hinfp, Wnt4 could be potential biomarkers of AD and the targets of fasudil treatment. These results will pave a novel direction for future clinic studies for AD and fasudil treatment.
Collapse
Affiliation(s)
- Hailong Yan
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, 037009, China
| | - Yuqing Yan
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, 037009, China. .,The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation, Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong, 030619, China.
| | - Ye Gao
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, 037009, China
| | - Nianping Zhang
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, 037009, China
| | - Gajendra Kumar
- Department of Neuroscience, City University of Hong Kong, Tat Chee Avenue, Kowloon Tong, Hong Kong
| | - Qingli Fang
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, 037009, China
| | - Ziqing Li
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, 037009, China
| | - Jiehui Li
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, 037009, China
| | - Yuna Zhang
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, 037009, China
| | - Lijuan Song
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation, Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong, 030619, China
| | - Jiawei Wang
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, 037009, China
| | - Jingxian Sun
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, 037009, China
| | - Han-Ting Zhang
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao, 266073, China.
| | - Cun-Gen Ma
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, 037009, China. .,The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation, Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong, 030619, China.
| |
Collapse
|
36
|
Liu S, Fan M, Zheng Q, Hao S, Yang L, Xia Q, Qi C, Ge J. MicroRNAs in Alzheimer's disease: Potential diagnostic markers and therapeutic targets. Biomed Pharmacother 2022; 148:112681. [PMID: 35177290 DOI: 10.1016/j.biopha.2022.112681] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 01/26/2022] [Accepted: 01/31/2022] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease, with cognitive decline as the primary clinical feature. According to epidemiological statistics, 50 million people worldwide are currently affected by Alzheimer's disease. Although new drugs such as aducanumab have been approved for use in the treatment of AD, none of them have reversed the progression of AD. MicroRNAs (miRNAs) are small molecule RNAs that exert their biological functions by regulating the expression of intracellular proteins, and differential abundance and varieties are found between the central and peripheral tissues of AD patients and healthy controls. This article will summarise the changes of miRNAs in the AD process, and the potential role of diagnostic markers and therapeutic targets in AD will be explored.
Collapse
Affiliation(s)
- Sen Liu
- School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei 230032, China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China; Anhui Provincial Laboratory of Inflammatory and Immunity Disease, Anhui Institute of Innovative Drugs, Hefei, China
| | - Min Fan
- School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei 230032, China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China; Anhui Provincial Laboratory of Inflammatory and Immunity Disease, Anhui Institute of Innovative Drugs, Hefei, China
| | - Qiang Zheng
- School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei 230032, China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China; Anhui Provincial Laboratory of Inflammatory and Immunity Disease, Anhui Institute of Innovative Drugs, Hefei, China
| | - Shengwei Hao
- School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei 230032, China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China; Anhui Provincial Laboratory of Inflammatory and Immunity Disease, Anhui Institute of Innovative Drugs, Hefei, China
| | - Longjun Yang
- Chaohu Clinical Medical College, Anhui Medical University, Hefei, China
| | - Qingrong Xia
- Department of Pharmacy, Hefei Fourth People's Hospital, Hefei, China
| | - Congcong Qi
- Department of Laboratory Animal Science, Fudan University, Shanghai, China.
| | - Jinfang Ge
- School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei 230032, China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China; Anhui Provincial Laboratory of Inflammatory and Immunity Disease, Anhui Institute of Innovative Drugs, Hefei, China.
| |
Collapse
|
37
|
Zhou S, Chen R, She Y, Liu X, Zhao H, Li C, Jia Y. A new perspective on depression and neuroinflammation: Non-coding RNA. J Psychiatr Res 2022; 148:293-306. [PMID: 35193033 DOI: 10.1016/j.jpsychires.2022.02.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 01/30/2022] [Accepted: 02/14/2022] [Indexed: 12/19/2022]
Abstract
The high incidence and relapse rate of depression, as well comorbidity with other diseases, has made depression one of the primary causes of years of life lived with disability. Moreover, the unknown biological mechanism of depression has made treatment difficult. Neuroinflammation is important in the pathogenesis of depression. Neuroinflammation may affect depression by regulating the production of immune factors, immune cell activation, neuron generation, synaptic plasticity, and neurotransmission. Non-coding RNAs (ncRNAs) may be a breakthrough link between depression and neuroinflammation, as ncRNAs participate in these biological changes. We summarize the functions and mechanisms of ncRNAs in neuroinflammation and depression, and predict ncRNAs that may regulate the occurrence and progression of depression through neuritis. These findings not only broaden our understanding of the genetic regulation of depression and neuroinflammation but also provide a new perspective of the underlying mechanism and aid in the design of novel prevention, diagnosis, and treatment strategies.
Collapse
Affiliation(s)
- Shanyao Zhou
- Guangdong Traditional Medical and Sports Injury Rehabilitation Research Institute, Guangdong Second Provincial General Hospital, 466 Xin Gang Zhong Road, Guangzhou, 510317, China
| | - Rui Chen
- Guangdong Traditional Medical and Sports Injury Rehabilitation Research Institute, Guangdong Second Provincial General Hospital, 466 Xin Gang Zhong Road, Guangzhou, 510317, China.
| | - Yanling She
- Guangdong Traditional Medical and Sports Injury Rehabilitation Research Institute, Guangdong Second Provincial General Hospital, 466 Xin Gang Zhong Road, Guangzhou, 510317, China
| | - Xuanjun Liu
- Department of Psychiatry, First Affiliated Hospital of Jinan University, 613 W. Huangpu Avenue, Guangzhou, 510630, China
| | - Hui Zhao
- Department of Psychiatry, First Affiliated Hospital of Jinan University, 613 W. Huangpu Avenue, Guangzhou, 510630, China
| | - Cheng Li
- Guangdong Traditional Medical and Sports Injury Rehabilitation Research Institute, Guangdong Second Provincial General Hospital, 466 Xin Gang Zhong Road, Guangzhou, 510317, China.
| | - Yanbin Jia
- Department of Psychiatry, First Affiliated Hospital of Jinan University, 613 W. Huangpu Avenue, Guangzhou, 510630, China.
| |
Collapse
|
38
|
LncRNA functional annotation with improved false discovery rate achieved by disease associations. Comput Struct Biotechnol J 2022; 20:322-332. [PMID: 35035785 PMCID: PMC8724965 DOI: 10.1016/j.csbj.2021.12.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 12/09/2021] [Accepted: 12/11/2021] [Indexed: 12/11/2022] Open
Abstract
The long non‐coding RNAs (lncRNAs) play critical roles in various biological processes and are associated with many diseases. Functional annotation of lncRNAs in diseases attracts great attention in understanding their etiology. However, the traditional co-expression-based analysis usually produces a significant number of false positive function assignments. It is thus crucial to develop a new approach to obtain lower false discovery rate for functional annotation of lncRNAs. Here, a novel strategy termed DAnet which combining disease associations with cis-regulatory network between lncRNAs and neighboring protein-coding genes was developed, and the performance of DAnet was systematically compared with that of the traditional differential expression-based approach. Based on a gold standard analysis of the experimentally validated lncRNAs, the proposed strategy was found to perform better in identifying the experimentally validated lncRNAs compared with the other method. Moreover, the majority of biological pathways (40%∼100%) identified by DAnet were reported to be associated with the studied diseases. In sum, the DAnet is expected to be used to identify the function of specific lncRNAs in a particular disease or multiple diseases.
Collapse
|
39
|
Tai Y, Chen J, Tao Z, Ren J. Non-coding RNAs: New players in mitophagy and neurodegeneration. Neurochem Int 2021; 152:105253. [PMID: 34864089 DOI: 10.1016/j.neuint.2021.105253] [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: 07/02/2021] [Revised: 11/14/2021] [Accepted: 11/29/2021] [Indexed: 10/19/2022]
Abstract
Mitophagy controls mitochondrial quality to maintain cellular homeostasis, while aberrations in this process are responsible for neurodegenerative diseases. Mitophagy is initiated through the recruitment of autophagosomes in a ubiquitin-dependent or ubiquitin-independent manner under different stress conditions. Although the detailed molecular mechanisms of how mitophagy processes influence neurodegeneration remain largely uncharacterized, there is mounting evidence indicating that non-coding RNAs (ncRNAs), a variety of endogenous regulators, including microRNAs and long non-coding RNAs, extensively participate in mitophagy processes and play pivotal roles in the aging process and neurodegenerative diseases. Here, we reviewed the major mitophagy pathways modulated by some classical and newly found ncRNAs and summarized the diverse mechanisms in a regulatory network. We also discussed the generalizability of ncRNAs in the development of common neurodegenerative diseases related to proteotoxicity and the importance of mitophagy in the pathogenesis of these diseases. In summary, we propose that ncRNAs act as linkers between mitophagy and neurodegeneration, showing the potential therapeutic application of mitophagy regulation mediated by ncRNAs in neurodegenerative diseases.
Collapse
Affiliation(s)
- Yusi Tai
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Jing Chen
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Zhouteng Tao
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
| | - Jin Ren
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
40
|
Das T, Das TK, Khodarkovskaya A, Dash S. Non-coding RNAs and their bioengineering applications for neurological diseases. Bioengineered 2021; 12:11675-11698. [PMID: 34756133 PMCID: PMC8810045 DOI: 10.1080/21655979.2021.2003667] [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] [Indexed: 02/07/2023] Open
Abstract
Engineering of cellular biomolecules is an emerging landscape presenting creative therapeutic opportunities. Recently, several strategies such as biomimetic materials, drug-releasing scaffolds, stem cells, and dynamic culture systems have been developed to improve specific biological functions, however, have been confounded with fundamental and technical roadblocks. Rapidly emerging investigations on the bioengineering prospects of mammalian ribonucleic acid (RNA) is expected to result in significant biomedical advances. More specifically, the current trend focuses on devising non-coding (nc) RNAs as therapeutic candidates for complex neurological diseases. Given the pleiotropic and regulatory role, ncRNAs such as microRNAs and long non-coding RNAs are deemed as attractive therapeutic candidates. Currently, the list of non-coding RNAs in mammals is evolving, which presents the plethora of hidden possibilities including their scope in biomedicine. Herein, we critically review on the emerging repertoire of ncRNAs in neurological diseases such as Alzheimer’s disease, Parkinson’s disease, neuroinflammation and drug abuse disorders. Importantly, we present the advances in engineering of ncRNAs to improve their biocompatibility and therapeutic feasibility as well as provide key insights into the applications of bioengineered non-coding RNAs that are investigated for neurological diseases.
Collapse
Affiliation(s)
- Tuhin Das
- Quanta Therapeutics, San Francisco, CA, 94158, USA.,RayBiotech, Inc, 3607 Parkway Lane, Peachtree Corners, GA, 30092, USA
| | - Tushar Kanti Das
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - Anne Khodarkovskaya
- Department of Pathology, Weill Cornell Medicine, Medical College of Cornell University, New York, NY, 10065, USA
| | - Sabyasachi Dash
- Department of Pathology, Weill Cornell Medicine, Medical College of Cornell University, New York, NY, 10065, USA.,School of Biotechnology, Kalinga Institute of Industrial Technology, Bhubaneswar, Odisha, 751024 India
| |
Collapse
|
41
|
Butt MS, Tariq U, Iahtisham-Ul-Haq, Naz A, Rizwan M. Neuroprotective effects of oleuropein: Recent developments and contemporary research. J Food Biochem 2021; 45:e13967. [PMID: 34716610 DOI: 10.1111/jfbc.13967] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 09/28/2021] [Accepted: 09/28/2021] [Indexed: 12/16/2022]
Abstract
Neurological disorders are increasing at a faster pace due to oxidative stress, protein aggregation, excitotoxicity, and neuroinflammation. It is reported that the Mediterranean diet including olives as a major dietary component prevents and ameliorates neurological anomalies. Oleuropein is the major bioactive component in different parts of the Olive (Olea europaea L.) tree. Several mechanisms have been reported for the neuroprotective role of oleuropein including induction of apoptosis and autophagy, enhancing the antioxidant pool of the cerebral region, decreasing the unnecessary release of proinflammatory cytokines and chemokines by deactivating the microglia cells and astrocytes thus preventing the occurrence of neuroinflammation. Regular intake of oleuropein seems to be correlated with decreased risks of neural disorders including Alzheimer's, Parkinson's, strokes, depression, anxiety, epilepsy, and others. This review majorly discusses the chemistry, biosynthesis, and metabolism of oleuropein along with an updated vision of its neuroprotective role in counteracting the acute and chronic neurodegenerative and neuropsychiatric disorders. Moreover, mechanisms by which oleuropein may prevent neurodegeneration are reviewed. PRACTICAL APPLICATION: Neurological disorders are negatively affecting the health and life quality of individuals around the globe. Although various medicinal solutions are available to tackle such ailments, none has proven to fully cure and being deprived of side effects. In this respect, the prevention of such disorders using natural remedies may be an effective strategy to overcome the incidence of the increasing cases. Furthermore, the natural compounds provide a safer alternative to pharmaceutical drugs. Hence, oleuropein from olive tree products is found to be efficacious against neurological disorders. This review provides an updated insight on the positive effects of oleuropein against neurodegenerative and neuropsychiatric disorders. The diet practitioners and nutraceutical companies may benefit from the provided information to design and develop strategies to improve the mental health of suffering individuals.
Collapse
Affiliation(s)
- Masood Sadiq Butt
- Faculty of Food, Nutrition and Home Sciences, National Institute of Food Science and Technology, University of Agriculture, Faisalabad, Pakistan
| | - Urwa Tariq
- Faculty of Food, Nutrition and Home Sciences, National Institute of Food Science and Technology, University of Agriculture, Faisalabad, Pakistan
| | - Iahtisham-Ul-Haq
- Faculty of Life Sciences, Department of Food Science and Technology, University of Central Punjab, Lahore, Pakistan
| | - Ambreen Naz
- Department of Food Science and Technology, Muhammad Nawaz Shareef University of Agriculture, Multan, Pakistan
| | - Muhammad Rizwan
- Faculty of Life Sciences, Department of Food Science and Technology, University of Central Punjab, Lahore, Pakistan
| |
Collapse
|
42
|
Hu Y, Zhang Y, Ren R, Dammer EB, Xie X, Chen S, Huang Q, Huang W, Zhang R, Chen H, Wang H, Wang G. microRNA-425 loss mediates amyloid plaque microenvironment heterogeneity and promotes neurodegenerative pathologies. Aging Cell 2021; 20:e13454. [PMID: 34510683 PMCID: PMC8520725 DOI: 10.1111/acel.13454] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 04/19/2021] [Accepted: 08/05/2021] [Indexed: 01/09/2023] Open
Abstract
Different cellular and molecular changes underlie the pathogenesis of Alzheimer's disease (AD). Among these, neuron‐specific dysregulation is a necessary event for accumulation of classic pathologies including amyloid plaques. Here, we show that AD‐associated pathophysiology including neuronal cell death, inflammatory signaling, and endolysosomal dysfunction is spatially colocalized to amyloid plaques in regions with abnormal microRNA‐425 (miR‐425) levels and this change leads to focal brain microenvironment heterogeneity, that is, an amyloid plaque‐associated microenvironment (APAM). APAM consists of multiple specific neurodegenerative signature pathologies associated with senile plaques that contribute to the heterogeneity and complexity of AD. Remarkably, miR‐425, a neuronal‐specific regulator decreased in AD brain, maintains a normal spatial transcriptome within brain neurons. We tested the hypothesis that miR‐425 loss correlates with enhanced levels of mRNA targets downstream, supporting APAM and AD progression. A miR‐425‐deficient mouse model has enhanced APP amyloidogenic processing, neuroinflammation, neuron loss, and cognitive impairment. In the APP/PS1 mouse model, intervening with miR‐425 supplementation ameliorated APAM changes and memory deficits. This study reveals a novel mechanism of dysregulation of spatial transcriptomic changes in AD brain, identifying a probable neuronal‐specific microRNA regulator capable of staving off amyloid pathogenesis. Moreover, our findings provide new insights for developing AD treatment strategies with miRNA oligonucleotide(s).
Collapse
Affiliation(s)
- Yong‐Bo Hu
- Department of Neurology and Neuroscience Institute Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine Shanghai China
- Department of Pharmacology and Chemical Biology Shanghai Jiao Tong University School of Medicine Shanghai China
- Department of Neurology,Shanghai East Hospital School of Medicine,Tongji University Shanghai China
| | - Yong‐Fang Zhang
- Department of Pharmacology and Chemical Biology Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Ru‐Jing Ren
- Department of Neurology and Neuroscience Institute Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Eric B. Dammer
- Department of Biochemistry and Center for Neurodegenerative Disease Emory University School of Medicine Atlanta Georgia USA
| | - Xin‐Yi Xie
- Department of Neurology and Neuroscience Institute Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Shi‐Wu Chen
- Department of Neurology and Neuroscience Institute Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Qiang Huang
- Department of Neurology and Neuroscience Institute Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Wan‐Ying Huang
- Department of Pharmacology and Chemical Biology Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Rui Zhang
- Department of Pharmacology and Chemical Biology Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Hong‐Zhuan Chen
- Institute of Interdisciplinary Science Shuguang Hospital Shanghai University of Traditional Chinese Medicine Shanghai China
| | - Hao Wang
- Department of Pharmacology and Chemical Biology Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Gang Wang
- Department of Neurology and Neuroscience Institute Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine Shanghai China
| |
Collapse
|
43
|
Dong Z, Gu H, Guo Q, Liang S, Xue J, Yao F, Liu X, Li F, Liu H, Sun L, Zhao K. Profiling of Serum Exosome MiRNA Reveals the Potential of a MiRNA Panel as Diagnostic Biomarker for Alzheimer's Disease. Mol Neurobiol 2021; 58:3084-3094. [PMID: 33629272 DOI: 10.1007/s12035-021-02323-y] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 02/05/2021] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease in the older adults. Although much effort has been made in the analyses of diagnostic biomarkers, such as amyloid-β, tau, and neurofilament light chain, identifying peripheral blood-based biomarkers is in extremely urgent need for their minimal invasiveness and more convenience. Here we characterized the miRNA profile by RNA sequencing in human serum exosomes from AD patients and healthy controls (HC) to investigate its potential for AD diagnosis. Subsequently, Gene Ontology analysis and pathway analysis were performed for the targeted genes from the differentially expressed miRNAs. These basic functions were differentially enriched, including cell adhesion, regulation of transcription, and the ubiquitin system. Functional network analysis highlighted the pathways of proteoglycans in cancer, viral carcinogenesis, signaling pathways regulating pluripotency of stem cells, and cellular senescence in AD. A total of 24 miRNAs showed significantly differential expression between AD and HC with more than ± 2.0-fold change at p value < 0.05 and at least 50 reads for each sample. Logistic regression analysis established a model for AD prediction by serum exosomal miR-30b-5p, miR-22-3p, and miR-378a-3p. Sequencing results were validated using quantitative reverse transcription PCR. The data showed that miR-30b-5p, miR-22-3p, and miR-378a-3p were significantly deregulated in AD, with area under the curve (AUC) of 0.668, 0.637, and 0.718, respectively. The combination of the three miRs gained a better diagnostic capability with AUC of 0.880. This finding revealed a miR panel as potential biomarker in the peripheral blood to distinguish AD from HC.
Collapse
Affiliation(s)
- Zhiwu Dong
- Department of Laboratory Medicine, Jinshan Branch of Shanghai Sixth People's Hospital Affiliated to Shanghai Jiaotong University, 147 Jiankang Road, Jinshan District, Shanghai, 201599, People's Republic of China.
| | - Hongjun Gu
- Shanghai Jinshan Zhongren Aged Care Hospital, Shanghai, 201501, China
| | - Qiang Guo
- Department of Ultrasound Medicine, Jinshan Branch of Shanghai Sixth People's Hospital Affiliated to Shanghai Jiaotong University, Shanghai, 201599, China
| | - Shuang Liang
- Department of Laboratory Medicine, Jinshan Branch of Shanghai Sixth People's Hospital Affiliated to Shanghai Jiaotong University, 147 Jiankang Road, Jinshan District, Shanghai, 201599, People's Republic of China
| | - Jian Xue
- Shanghai Jinshan Zhongren Aged Care Hospital, Shanghai, 201501, China
| | - Feng Yao
- Shanghai Jinshan Zhongren Aged Care Hospital, Shanghai, 201501, China
| | - Xianglu Liu
- Department of Laboratory Medicine, Jinshan Branch of Shanghai Sixth People's Hospital Affiliated to Shanghai Jiaotong University, 147 Jiankang Road, Jinshan District, Shanghai, 201599, People's Republic of China
| | - Feifei Li
- Department of Laboratory Medicine, Jinshan Branch of Shanghai Sixth People's Hospital Affiliated to Shanghai Jiaotong University, 147 Jiankang Road, Jinshan District, Shanghai, 201599, People's Republic of China
| | - Huiling Liu
- Department of Laboratory Medicine, Jinshan Branch of Shanghai Sixth People's Hospital Affiliated to Shanghai Jiaotong University, 147 Jiankang Road, Jinshan District, Shanghai, 201599, People's Republic of China
| | - Li Sun
- Department of Laboratory Medicine, Jinshan Branch of Shanghai Sixth People's Hospital Affiliated to Shanghai Jiaotong University, 147 Jiankang Road, Jinshan District, Shanghai, 201599, People's Republic of China
| | - Kewen Zhao
- Department of Pathophysiology, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, People's Republic of China
| |
Collapse
|
44
|
Gu F, Ji D, Ni H, Chen D. SRY-Box 21 Antisense RNA 1 Knockdown Diminishes Amyloid Beta 25-35-Induced Neuronal Damage by miR-132/PI3K/AKT Pathway. Neurochem Res 2021; 46:2376-2386. [PMID: 34146195 DOI: 10.1007/s11064-021-03373-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 06/04/2021] [Accepted: 06/07/2021] [Indexed: 12/12/2022]
Abstract
Our study aimed to explore the function and mechanism of action of long noncoding RNA (lncRNA) SRY-Box 21 antisense RNA 1 (SOX21-AS1) in amyloid beta25-35 (Aβ25-35)-induced neuronal damage. To induce neuronal damage, neuronal cells and differentiated IMR-32 neuroblastoma cells were challenged by Aβ25-35. SOX21-AS1 and miR-132 quantities were detected by quantitative reverse transcription polymerase chain reaction. Cell damage was evaluated by detecting the changes of cell viability, apoptosis, and oxidative stress. Cell viability was measured using cell counting kit-8. Cell apoptosis was evaluated by flow cytometry and caspase-3 activity. The oxidative stress was analyzed by reactive oxygen species level. The expression of proteins associated with the phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) pathway was examined by western blot. SOX21-AS1 abundance was up-regulated in Aβ25-35-challenged neuronal cells. Silencing of SOX21-AS1 attenuated Aβ25-35-induced viability reduction and promotion of apoptosis and oxidative stress, suggesting that silencing of SOX21-AS1 repressed Aβ25-35-induced neuronal damage. miR-132 quantity was reduced in Aβ25-35-challenged neuronal cells, and negatively controlled by SOX21-AS1. miR-132 knockdown abolished the effect of SOX21-AS1 silencing on Aβ25-35-induced neuronal damage, indicating that SOX21-AS1 controls Aβ25-35-induced neuronal damage via regulating miR-132. The PI3K/AKT signaling was repressed in Aβ25-35-challenged cells, but this effect was counteracted upon overexpression of miR-132. In conclusion, SOX21-AS1 knockdown mitigated Aβ25-35-dependent neuronal cell damage by promoting miR-132/PI3K/AKT pathway.
Collapse
Affiliation(s)
- Fengming Gu
- Department of Intensive Care Unit, The Affiliated Huai'an Hospital of Xuzhou Medical University, The Second People's Hospital of Huai'an, Huai'an, 223002, China
| | - Daofei Ji
- Department of Neurosurgery, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221006, China
| | - Hongzao Ni
- Department of Neurosurgery, The Affiliated Huai'an Hospital of Xuzhou Medical University, The Second People's Hospital of Huai'an, Huai'an, 223002, China
| | - Depeng Chen
- Department of Intensive Care Unit, People's Hospital of Huai'an Hongze District, 102 Dongfeng Road, Huai'an, 223100, China.
| |
Collapse
|
45
|
Aliperti V, Skonieczna J, Cerase A. Long Non-Coding RNA (lncRNA) Roles in Cell Biology, Neurodevelopment and Neurological Disorders. Noncoding RNA 2021; 7:36. [PMID: 34204536 PMCID: PMC8293397 DOI: 10.3390/ncrna7020036] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 06/14/2021] [Accepted: 06/15/2021] [Indexed: 02/08/2023] Open
Abstract
Development is a complex process regulated both by genetic and epigenetic and environmental clues. Recently, long non-coding RNAs (lncRNAs) have emerged as key regulators of gene expression in several tissues including the brain. Altered expression of lncRNAs has been linked to several neurodegenerative, neurodevelopmental and mental disorders. The identification and characterization of lncRNAs that are deregulated or mutated in neurodevelopmental and mental health diseases are fundamental to understanding the complex transcriptional processes in brain function. Crucially, lncRNAs can be exploited as a novel target for treating neurological disorders. In our review, we first summarize the recent advances in our understanding of lncRNA functions in the context of cell biology and then discussing their association with selected neuronal development and neurological disorders.
Collapse
Affiliation(s)
- Vincenza Aliperti
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy
| | - Justyna Skonieczna
- Centre for Genomics and Child Health, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK;
| | - Andrea Cerase
- Centre for Genomics and Child Health, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK;
| |
Collapse
|
46
|
Kong F, Lv Z, Wang L, Zhang K, Cai Y, Ding Q, Sun Z, Zhen H, Jiao F, Ma Q, Nie C, Yang Y. RNA-sequencing of peripheral blood circular RNAs in Parkinson disease. Medicine (Baltimore) 2021; 100:e25888. [PMID: 34114985 PMCID: PMC8202568 DOI: 10.1097/md.0000000000025888] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 04/20/2021] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Circular RNAs (circRNAs) play an important role in many neurological diseases and can serve as biomarkers for these diseases. However, the information about circRNAs in Parkinson disease (PD) remained limited. In this study, we aimed to determine the circRNAs expression profile in PD patients and discuss the significance of circRNAs in the diagnosis of PD. METHODS AND RESULTS Using RNA-sequencing in peripheral blood RNAs, we showed that a significant number of mRNAs or circRNAs were differentially expressed between PD patients and normal controls (NCs), which included 273 up-regulated and 493 down-regulated mRNAs, and 129 up-regulated and 282 down-regulated circRNAs, respectively. Functional analysis was performed using the Kyoto Encyclopedia of Gene and Genomes (KEGG) pathway analysis, and the results showed that the second most enriched KEGG pathway was PD. These data suggest that the levels of mRNAs and circRNAs in peripheral blood could be potentially used as biomarkers for PD. In addition, we correlated mRNAs and circRNAs by constructing a competing endogenous RNA (ceRNA) network in PD. The resulted-in ceRNA network included 10 differentially expressed mRNAs from PD pathway, 13 predicted miRNAs, and 10 differentially expressed circRNAs. CONCLUSION Collectively, we first characterized the expression profiles of circRNAs and mRNAs in peripheral blood from PD patients and proposed their possible characters in the pathogenesis of PD. These results provided valuable insights into the clues underlying the pathogenesis of PD.
Collapse
Affiliation(s)
- Fancong Kong
- Department of Neurology, The Affiliated Hospital of Jining Medical University
- Clinical Medical College
| | - Zhanyun Lv
- Department of Neurology, The Affiliated Hospital of Jining Medical University
| | - Lifang Wang
- BGI-Shenzhen
- China National GeneBank
- Shenzhen Key Laboratory of Neurogenomics, BGI-Shenzhen, Shenzhen
| | - Kui Zhang
- Department of Neurology, The Affiliated Hospital of Jining Medical University
| | - Ying Cai
- Department of Neurology, The Affiliated Hospital of Jining Medical University
- Clinical Medical College
| | - Qingqing Ding
- Department of Neurology, The Affiliated Hospital of Jining Medical University
- Clinical Medical College
| | - Zuzhen Sun
- Department of Neurology, The Affiliated Hospital of Jining Medical University
| | - Hefu Zhen
- BGI-Shenzhen
- China National GeneBank
- Shenzhen Key Laboratory of Neurogenomics, BGI-Shenzhen, Shenzhen
| | - Fengjuan Jiao
- Shandong Key Laboratory of Behavioral Medicine, School of Mental Health
- Shandong Collaborative Innovation Center for Diagnosis, Treatment & Behavioral Interventions of Mental Disorders, Institute of Mental Health, Jining Medical University, Jining, China
| | - Qianqian Ma
- Department of Neurology, The Affiliated Hospital of Jining Medical University
| | - Chao Nie
- BGI-Shenzhen
- China National GeneBank
- Shenzhen Key Laboratory of Neurogenomics, BGI-Shenzhen, Shenzhen
| | - Yan Yang
- Department of Neurology, The Affiliated Hospital of Jining Medical University
- Clinical Medical College
| |
Collapse
|
47
|
Zhang L, Fang Y, Zhao X, Zheng Y, Ma Y, Li S, Huang Z, Li L. miR-204 silencing reduces mitochondrial autophagy and ROS production in a murine AD model via the TRPML1-activated STAT3 pathway. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 24:822-831. [PMID: 34026326 PMCID: PMC8121631 DOI: 10.1016/j.omtn.2021.02.010] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 02/05/2021] [Indexed: 12/28/2022]
Abstract
Mitochondrial dysfunction is an early feature of Alzheimer's disease (AD), whereby accumulation of damaged mitochondria in conjunction with impaired mitophagy contributes to neurodegeneration. Various non-transcribed microRNAs (miRNAs) are involved in this process. In the present study, we aimed to decipher the participation of miR-204 in a murine AD model. Primary hippocampal neurons were isolated from mice and treated with β-amyloid 1-42 (Aβ1-42) to establish a cell model of AD. Dichloro-dihydro-fluorescein diacetate and dihydrorhodamine 123 staining assays were performed to measure total reactive oxygen species (ROS) and mitochondrial ROS production in neurons, and MitoSOX staining was done to analyze mitochondrial ROS production in hippocampus. Furthermore, mitochondrial autophagy was observed in hippocampus from amyloid precursor protein/pesenilin-1 AD modeled mice, and their cognitive function was assessed by Morris water maze. Mitochondrial damage, ROS production, and mitochondrial autophagy were observed in AD cell model induced by Aβ1-42. In AD, signal transducer and activator of transcription 3 (STAT3) and transient receptor potential mucolipin-1 (TRPML1) expression was downregulated, although miR-204 expression was upregulated. TRPML1 overexpression, downregulation of miR-204, or STAT3 pathway activation reduced the Aβ1-42-induced mitochondrial damage, along with ROS production and mitochondrial autophagy in vivo and in vitro. Silencing of miR-204 could upregulate TRPML1 expression, thus suppressing ROS production and mitochondrial autophagy in AD through STAT3 pathway.
Collapse
Affiliation(s)
- Lu Zhang
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P.R. China
| | - Yu Fang
- ICU, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P.R. China
| | - Xinyu Zhao
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P.R. China
| | - Yake Zheng
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P.R. China
| | - Yunqing Ma
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P.R. China
| | - Shuang Li
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P.R. China
| | - Zhi Huang
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P.R. China
| | - Lihao Li
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P.R. China
| |
Collapse
|
48
|
Jha N, Mangukia N, Patel MP, Bhavsar M, Gadhavi H, Rawal RM, Patel SK. Exploring the MiRnome of Carica papaya: A cross kingdom approach. GENE REPORTS 2021. [DOI: 10.1016/j.genrep.2021.101089] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
49
|
Zhang T, Shen Y, Guo Y, Yao J. Identification of key transcriptome biomarkers based on a vital gene module associated with pathological changes in Alzheimer's disease. Aging (Albany NY) 2021; 13:14940-14967. [PMID: 34031265 PMCID: PMC8221319 DOI: 10.18632/aging.203017] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 04/05/2021] [Indexed: 12/22/2022]
Abstract
Dysregulation of transcriptome expression has been reported to play an increasingly significant role in AD. In this study, we firstly identified a vital gene module associated with the accumulation of β-amyloid (Aβ) and phosphorylated tau (p-tau) using the WGCNA method. The vital module, named target module, was then employed for the identification of key transcriptome biomarkers. For coding RNA, GNA13 and GJA1 were identified as key biomarkers based on ROC analysis. As for non-coding RNA, MEG3, miR-106a-3p, and miR-24-3p were determined as key biomarkers based on analysis of a ceRNA network and ROC analysis. Experimental analyses firstly confirmed that GNA13, GJA1, and ROCK2, a downstream effector of GNA13, were all increased in 5XFAD mice, compared to littermate mice. Moreover, their expression was increased with aging in 5XFAD mice, as Aβ and p-tau pathology developed. Besides, the expression of key ncRNA biomarkers was verified to be decreased in 5XFAD mice. GSEA results indicated that GNA13 and GJA1 were respectively involved in ribosome and spliceosome dysfunction. MEG3, miR-106a-3p, and miR-24-3p were identified to be involved in MAPK pathway and PI3K-Akt pathway based on enrichment analysis. In summary, we identified several key transcriptome biomarkers, which promoted the prediction and diagnosis of AD.
Collapse
Affiliation(s)
- Tong Zhang
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yang Shen
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiqing Guo
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junyan Yao
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
50
|
Cho JD, Kim YA, Rafikian EE, Yang M, Santa-Maria I. Marked Mild Cognitive Deficits in Humanized Mouse Model of Alzheimer's-Type Tau Pathology. Front Behav Neurosci 2021; 15:634157. [PMID: 34093145 PMCID: PMC8175658 DOI: 10.3389/fnbeh.2021.634157] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 04/21/2021] [Indexed: 01/23/2023] Open
Abstract
Hyperphosphorylation and the subsequent aggregation of tau protein into neurofibrillary tangles (NFTs) are well-established neuropathological hallmarks of Alzheimer’s disease (AD) and associated tauopathies. To further examine the impact and progression of human tau pathology in neurodegenerative contexts, the humanized tau (htau) mouse model was originally created. Despite AD-like tau pathological features recapitulated in the htau mouse model, robustness of behavioral phenotypes has not been fully established. With the ultimate goal of evaluating the htau mouse model as a candidate for testing AD therapeutics, we set out to verify, in-house, the presence of robust, replicable cognitive deficits in the htau mice. The present study shows behavioral data collected from a carefully curated battery of learning and memory tests. Here we report a significant short-term spatial memory deficit in aged htau mice, representing a novel finding in this model. However, we did not find salient impairments in long-term learning and memory previously reported in this mouse model. Here, we attempted to understand the discrepancies in the literature by highlighting the necessity of scrutinizing key procedural differences across studies. Reported cognitive deficits in the htau model may depend on task difficulty and other procedural details. While the htau mouse remains a unique and valuable animal model for replicating late onset AD-like human tau pathology, its cognitive deficits are modest under standard testing conditions. The overarching message is that before using any AD mouse model to evaluate treatment efficacies, it is imperative to first characterize and verify the presence of behavioral deficits in-house.
Collapse
Affiliation(s)
- Joshua D Cho
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, United States.,Department of Pathology & Cell Biology, Columbia University, New York, NY, United States
| | - Yoon A Kim
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, United States.,Department of Pathology & Cell Biology, Columbia University, New York, NY, United States
| | - Elizabeth E Rafikian
- The Mouse NeuroBehavior Core, Institute for Genomic Medicine, Columbia University, New York, NY, United States
| | - Mu Yang
- The Mouse NeuroBehavior Core, Institute for Genomic Medicine, Columbia University, New York, NY, United States
| | - Ismael Santa-Maria
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, United States.,Department of Pathology & Cell Biology, Columbia University, New York, NY, United States
| |
Collapse
|