1
|
Marino C, Malotaux V, Giudicessi A, Aguillon D, Sepulveda-Falla D, Lopera F, Quiroz YT. Protective genetic variants against Alzheimer's disease. Lancet Neurol 2025; 24:524-534. [PMID: 40409316 DOI: 10.1016/s1474-4422(25)00116-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 01/24/2025] [Accepted: 03/31/2025] [Indexed: 05/25/2025]
Abstract
Genetic studies can offer powerful insights for the development of disease-modifying therapies for Alzheimer's disease. Protective genetic variants that delay the onset of cognitive impairment have been found in people with sporadic Alzheimer's disease and in carriers of mutations that usually cause autosomal-dominant Alzheimer's disease in mid-life. The study of families who carry autosomal dominant mutations provides a unique opportunity to uncover genetic modifiers of disease progression, including rare variants in genes such as APOE and RELN. Understanding how these variants confer protection can help identify the biological pathways that contribute to cognitive resilience, such as the heparan-sulphate proteoglycan-APOE receptor pathway, the TREM-2-driven signalling pathways in the microglia, and phagocytosis. Therapies able to replicate the beneficial effects of these natural defences could provide novel strategies for slowing or preventing the progression of Alzheimer's disease.
Collapse
Affiliation(s)
- Claudia Marino
- The Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA, USA; Department of Neurology, Sealy Institute for Drug Discovery and Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical Branch, Galveston, TX, USA
| | - Vincent Malotaux
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Averi Giudicessi
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Boston University, Department of Psychological and Brain Sciences, Boston, MA, USA
| | - David Aguillon
- The Neuroscience Group of Antioquia, School of Medicine, University of Antioquia, Medellín, Colombia
| | - Diego Sepulveda-Falla
- The Institute of Neuropathology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Francisco Lopera
- The Neuroscience Group of Antioquia, School of Medicine, University of Antioquia, Medellín, Colombia
| | - Yakeel T Quiroz
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Boston University, Department of Psychological and Brain Sciences, Boston, MA, USA; The Neuroscience Group of Antioquia, School of Medicine, University of Antioquia, Medellín, Colombia; Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
2
|
Sandberg A, Puttagunta S, Duval N, Fleming H, Koza L, Hieber K, Holsopple J, Reyna M, Paredes D, Linseman DA. Immunocal ®, a cysteine-rich whey protein, rescues reelin and reduces amyloid plaque burden in a transgenic amyloid-β protein precursor (hAβPP SweInd) mouse model of Alzheimer's disease. J Alzheimers Dis 2025:13872877251338323. [PMID: 40325994 DOI: 10.1177/13872877251338323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2025]
Abstract
BackgroundDeficits in Reelin expression play a significant role in the pathogenesis of various neurological disorders, including schizophrenia and Alzheimer's disease (AD). Notably, Reelin-expressing neurons of the entorhinal cortex layer II are among the first to be affected in AD.ObjectiveStrategies aimed at correcting deficits in Reelin might provide a novel therapeutic approach for AD.MethodsHere, we examined the effects of the whey protein supplement and glutathione (GSH) precursor, Immunocal®, on Reelin expression both in vitro in hippocampal-entorhinal cortex slices from rat brain and in vivo in the hAβPPSweInd (J20) mouse model of AD.ResultsIncubation of brain slices with Immunocal® increased Reelin expression at the mRNA and protein levels. Oral treatment with Immunocal®, given ad libitum in drinking water beginning at 3 months of age, corrected a deficit in cortical GSH levels observed in untreated mice and preserved Reelin expression in the hippocampal-entorhinal cortex sub-region of 5-month-old J20 mice. We also assessed the long-term effects of Immunocal® by treating J20 mice from 3 months old to 12 months old. Long-term Immunocal® treatment preserved brain GSH and rescued Reelin mRNA and protein expression, while significantly reducing amyloid plaque formation in the entorhinal cortex and hippocampus of AD mice.ConclusionsThese findings suggest that Immunocal® promotes Reelin expression in vitro and sustains brain GSH and Reelin expression while diminishing amyloid plaque load in the entorhinal cortex and hippocampus of J20 mice. Thus, Immunocal® offers a promising therapeutic approach to enhance Reelin expression and curtail amyloid deposition in AD.
Collapse
Affiliation(s)
- Alexandra Sandberg
- Department of Biological Sciences, University of Denver, Denver, CO, USA
| | | | - Nathan Duval
- Knoebel Institute for Healthy Aging, University of Denver, Denver, CO, USA
| | - Holly Fleming
- Knoebel Institute for Healthy Aging, University of Denver, Denver, CO, USA
| | - Lilia Koza
- Department of Biological Sciences, University of Denver, Denver, CO, USA
| | - Kade Hieber
- Department of Biological Sciences, University of Denver, Denver, CO, USA
| | - Jessica Holsopple
- Department of Biological Sciences, University of Denver, Denver, CO, USA
| | - Michael Reyna
- Department of Biological Sciences, University of Denver, Denver, CO, USA
| | - Daniel Paredes
- Knoebel Institute for Healthy Aging, University of Denver, Denver, CO, USA
| | - Daniel A Linseman
- Department of Biological Sciences, University of Denver, Denver, CO, USA
- Knoebel Institute for Healthy Aging, University of Denver, Denver, CO, USA
| |
Collapse
|
3
|
Ashitomi H, Nakagawa T, Nakagawa M, Hosoi T. Cullin-RING Ubiquitin Ligases in Neurodevelopment and Neurodevelopmental Disorders. Biomedicines 2025; 13:810. [PMID: 40299365 PMCID: PMC12024872 DOI: 10.3390/biomedicines13040810] [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: 03/06/2025] [Revised: 03/26/2025] [Accepted: 03/27/2025] [Indexed: 04/30/2025] Open
Abstract
Ubiquitination is a dynamic and tightly regulated post-translational modification essential for modulating protein stability, trafficking, and function to preserve cellular homeostasis. This process is orchestrated through a hierarchical enzymatic cascade involving three key enzymes: the E1 ubiquitin-activating enzyme, the E2 ubiquitin-conjugating enzyme, and the E3 ubiquitin ligase. The final step of ubiquitination is catalyzed by the E3 ubiquitin ligase, which facilitates the transfer of ubiquitin from the E2 enzyme to the substrate, thereby dictating which proteins undergo ubiquitination. Emerging evidence underscores the critical roles of ubiquitin ligases in neurodevelopment, regulating fundamental processes such as neuronal polarization, axonal outgrowth, synaptogenesis, and synaptic function. Mutations in genes encoding ubiquitin ligases and the consequent dysregulation of these pathways have been increasingly implicated in a spectrum of neurodevelopmental disorders, including autism spectrum disorder, intellectual disability, and attention-deficit/hyperactivity disorder. This review synthesizes current knowledge on the molecular mechanisms underlying neurodevelopment regulated by Cullin-RING ubiquitin ligases-the largest subclass of ubiquitin ligases-and their involvement in the pathophysiology of neurodevelopmental disorders. A deeper understanding of these mechanisms holds significant promise for informing novel therapeutic strategies, ultimately advancing clinical outcomes for individuals affected by neurodevelopmental disorders.
Collapse
Affiliation(s)
- Honoka Ashitomi
- Department of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Sanyo-Onoda 756-0084, Japan; (H.A.)
| | - Tadashi Nakagawa
- Department of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Sanyo-Onoda 756-0084, Japan; (H.A.)
- Division of Cell Proliferation, United Centers for Advanced Research and Translational Medicine, Graduate School of Medicine, Tohoku University, Sendai 980-8575, Japan
| | - Makiko Nakagawa
- Institute of Gene Research, Yamaguchi University Science Research Center, Ube 755-8505, Japan
- Advanced Technology Institute, Life Science Division, Yamaguchi University, Ube 755-8611, Japan
| | - Toru Hosoi
- Department of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Sanyo-Onoda 756-0084, Japan; (H.A.)
| |
Collapse
|
4
|
Bu F, Zhong J, Guan R. Biomarkers in glioblastoma and degenerative CNS diseases: defining new advances in clinical usefulness and therapeutic molecular target. Front Mol Biosci 2025; 12:1506961. [PMID: 40171042 PMCID: PMC11959069 DOI: 10.3389/fmolb.2025.1506961] [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: 10/06/2024] [Accepted: 02/28/2025] [Indexed: 04/03/2025] Open
Abstract
Background Discovering biomarkers is central to the research and treatment of degenerative central nervous system (CNS) diseases, playing a crucial role in early diagnosis, disease monitoring, and the development of new treatments, particularly for challenging conditions like degenerative CNS diseases and glioblastoma (GBM). Methods This study analyzed gene expression data from a public database, employing differential expression analyses and Gene Co-expression Network Analysis (WGCNA) to identify gene modules associated with degenerative CNS diseases and GBM. Machine learning methods, including Random Forest, Least Absolute Shrinkage and Selection Operator (LASSO), and Support Vector Machine - Recursive Feature Elimination (SVM-RFE), were used for case-control differentiation, complemented by functional enrichment analysis and external validation of key genes. Results Ninety-five commonly altered genes related to degenerative CNS diseases and GBM were identified, with RELN and GSTO2 emerging as significant through machine learning screening. Receiver operating characteristic (ROC) analysis confirmed their diagnostic value, which was further validated externally, indicating their elevated expression in controls. Conclusion The study's integration of WGCNA and machine learning uncovered RELN and GSTO2 as potential biomarkers for degenerative CNS diseases and GBM, suggesting their utility in diagnostics and as therapeutic targets. This contributes new perspectives on the pathogenesis and treatment of these complex conditions.
Collapse
Affiliation(s)
- Fan Bu
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Jifa Zhong
- Heilongjiang University of Chinese Medicine Affiliated Second Hospital, Harbin, China
| | - Ruiqian Guan
- Heilongjiang University of Chinese Medicine Affiliated Second Hospital, Harbin, China
| |
Collapse
|
5
|
Tezcan ME, Ekici F, Ugur C, Can Ü, Karatoprak S, Sağlıyan GA, Uçak EF, Güleç A, Erbasan V, Sen B, Simsek F, Atas AE. Do specific myelin autoantibodies and increased cerebral dopamine neurotrophic factor in the context of inflammation predict the diagnosis of attention deficit hyperactivity disorder in medication-free children? Brain Behav Immun 2025; 124:125-136. [PMID: 39617068 DOI: 10.1016/j.bbi.2024.11.026] [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: 07/18/2024] [Revised: 10/15/2024] [Accepted: 11/22/2024] [Indexed: 01/20/2025] Open
Abstract
BACKGROUND The aim of this study was to investigate the serum levels of anti-myelin basic protein (anti-MBP), anti-myelin oligodentrocyte glycoprotein (anti-MOG), myelin-associated glycoprotein (MAG), high-sensitivity C-reactive protein (hs-CRP), cerebral dopamine neurotrophic factor (CDNF), cerebellin-1, and reelin and their relationships with clinical severity and irritability behaviours in children with attention deficit (AD) hyperactivity disorder (ADHD) and typically developing (TD) healthy controls. METHODS In this study, 141 children with ADHD between the ages of 8 and 14 years who were medication-free and 135 TD healthy controls were included. The serum levels of anti-MBP, anti-MOG, MAG, CDNF, hs-CRP, cerebellin, and reelin were measured using enzyme-linked immunosorbent assay kits. The Turgay Diagnostic and Statistical Manual of Mental Disorders, Fourth Edition (DSM-IV)-based Screening and Evaluation Scale for Attention Deficit and Disruptive Behavior Disorders-Parent Form (TDSM-IV-O) and the affective reactivity index (ARI) scale were used to assess clinical severity and irritability behaviours in the children. RESULTS The MAG, CDNF, hs-CRP, reelin, and cerebellin levels were significantly higher in the ADHD group than in the control group, but no significant differences in anti-MBP and anti-MOG levels were found between the groups. Compared with the controls, the patients with ADHD showed significantly higher scores on the ARI self- and parent-report scales. The reelin, hs-CRP, and MAG levels were significantly associated with the TDSM-IV-O AD scores, AD and oppositional defiant (OD) disorder scores and hyperactivity, and OD and conduct disorder scores, respectively. Hs-CRP was significantly associated with anti-MBP and cerebellin levels. In an analysis of covariance, the results were unchanged even after controlling for potential confounders such as age, body mass index, and sex. CONCLUSION This study demonstrates that MAG, CDNF, hs-CRP, reelin, and cerebellin levels may play a potential role in the pathogenesis of ADHD.
Collapse
Affiliation(s)
- Mustafa Esad Tezcan
- Department of Child and Adolescent Psychiatry, Konya City Hospital, Karatay-Konya 42020, Turkey.
| | - Fatih Ekici
- Department of Psychiatry, Konya City Hospital, Karatay-Konya 42020, Turkey.
| | - Cüneyt Ugur
- Department of Pediatrics, Konya City Health Application and Research, University of Health Sciences Turkey, Karatay-Konya 42020, Turkey.
| | - Ümmügülsüm Can
- Department of Medical Biochemistry, Konya City Health Application and Research, University of Health Sciences Turkey, Karatay-Konya 42020, Turkey.
| | - Serdar Karatoprak
- Department of Child and Adolescent Psychiatry, Konya City Hospital, Karatay-Konya 42020, Turkey.
| | | | - Ekrem Furkan Uçak
- Department of Psychiatry, Konya City Hospital, Karatay-Konya 42020, Turkey.
| | - Ahmet Güleç
- Department of Child and Adolescent Psychiatry, Balıkesir City Hospital, Altıeylül, Balıkesir, Turkey.
| | - Vefa Erbasan
- Department of Psychiatry, İzmir City Hospital, Bayraklı, 35540 Izmir, Turkey.
| | - Barıs Sen
- Department of Psychiatry, Manavgat State Hospital, Manavgat-Antalya, Turkey.
| | - Fulya Simsek
- Department of Child and Adolescent Psychiatry, Konya City Hospital, Karatay-Konya 42020, Turkey.
| | - Abdullah Enes Atas
- Department of Radiology, Konya City Hospital, Karatay-Konya 42020, Turkey.
| |
Collapse
|
6
|
Cai H, Lee SM, Choi Y, Lee B, Im SJ, Kim DH, Choi HJ, Kim JH, Kim Y, Shin BA, Jeon S. Memory Decline and Aberration of Synaptic Proteins in X-Linked Moesin Knockout Male Mice. Psychiatry Investig 2025; 22:10-25. [PMID: 39885788 PMCID: PMC11788833 DOI: 10.30773/pi.2024.0186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 09/23/2024] [Accepted: 10/02/2024] [Indexed: 02/01/2025] Open
Abstract
OBJECTIVE This study aims to investigate may moesin deficiency resulted in neurodevelopmental abnormalities caused by negative impact on synaptic signaling ultimately leading to synaptic structure and plasticity. METHODS Behavioral assessments measured neurodevelopment (surface righting, negative geotaxis, cliff avoidance), anxiety (open field test, elevated plus maze test), and memory (passive avoidance test, Y-maze test) in moesin-knockout mice (KO) compared to wild-type mice (WT). Whole exome sequencing (WES) of brain (KO vs. WT) and analysis of synaptic proteins were performed to determine the disruption of signal pathways downstream of moesin. Risperidone, a therapeutic agent, was utilized to reverse the neurodevelopmental aberrance in moesin KO. RESULTS Moesin-KO pups exhibited decrease in the surface righting ability on postnatal day 7 (p<0.05) and increase in time spent in the closed arms (p<0.01), showing increased anxiety-like behavior. WES revealed mutations in pathway aberration in neuron projection, actin filament-based processes, and neuronal migration in KO. Decreased cell viability (p<0.001) and expression of soluble NSF adapter protein 25 (SNAP25) (p<0.001) and postsynaptic density protein 95 (PSD95) (p<0.01) was observed in days in vitro 7 neurons. Downregulation of synaptic proteins, and altered phosphorylation levels of Synapsin I, mammalian uncoordinated 18 (MUNC18), extracellular signal-regulated kinase (ERK), and cAMP response element-binding protein (CREB) was observed in KO cortex and hippocampus. Risperidone reversed the memory impairment in the passive avoidance test and the spontaneous alternation percentage in the Y maze test. Risperidone also restored the reduced expression of PSD95 (p<0.01) and the phosphorylation of Synapsin at Ser605 (p<0.05) and Ser549 (p<0.001) in the cortex of moesin-KO. CONCLUSION Moesin deficiency leads to neurodevelopmental delay and memory decline, which may be caused through altered regulation in synaptic proteins and function.
Collapse
Affiliation(s)
- Hua Cai
- Department of Microbiology and Immunology, Chonnam National University Medical School, Gwangju, Republic of Korea
- Department of Molecular Medicine (BK21plus), Chonnam National University Graduate School, Gwangju, Republic of Korea
| | - Seong Mi Lee
- Department of Neuropsychiatry, Dongguk University School of Medicine, Seoul, Republic of Korea
- Mental Health Research Institute, National Center for Mental Health, Seoul, Republic of Korea
| | - Yura Choi
- Department of Neuropsychiatry, Dongguk University School of Medicine, Seoul, Republic of Korea
| | - Bomlee Lee
- Mental Health Research Institute, National Center for Mental Health, Seoul, Republic of Korea
| | - Soo Jung Im
- Mental Health Research Institute, National Center for Mental Health, Seoul, Republic of Korea
| | - Dong Hyeon Kim
- Mental Health Research Institute, National Center for Mental Health, Seoul, Republic of Korea
| | - Hyung Jun Choi
- Mental Health Research Institute, National Center for Mental Health, Seoul, Republic of Korea
| | - Jin Hee Kim
- Department of Microbiology and Immunology, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Yeni Kim
- Department of Neuropsychiatry, Dongguk University School of Medicine, Seoul, Republic of Korea
- Department of Child and Adolescent Psychiatry, National Center for Mental Health, Seoul, Republic of Korea
- Dongguk University International Hospital, Institute of Clinical Psychopharmacology, Goyang, Republic of Korea
| | - Boo Ahn Shin
- Department of Microbiology and Immunology, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Songhee Jeon
- Department of Biomedical Sciences, Center for Glocal Future Biomedical Scientists at Chonnam National University, Gwangju, Republic of Korea
| |
Collapse
|
7
|
Widaja E, Pawitan JA. Integrating epigenetic modification and stem cell therapy strategies: A novel approach for advancing Alzheimer's disease treatment - A literature review. NARRA J 2024; 4:e935. [PMID: 39816083 PMCID: PMC11731673 DOI: 10.52225/narra.v4i3.202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 11/19/2024] [Indexed: 01/18/2025]
Abstract
Alzheimer's disease (AD) is the most frequent form of dementia and represents an increasing global burden, particularly in countries like Indonesia, where the population has begun to age significantly. Current medications, including cholinesterase inhibitors and NMDA receptor antagonists, have modest effects on clinical symptoms in the early to middle stages, but there is no curative treatment available so far despite progress. Activating or repressing epigenetic modifications, including DNA methylation, histone modification and microRNA regulation, appears to play an important role in AD development. These alterations further enact transcriptional changes relevant to the signature AD pathologies of amyloid-β deposition, tau protein malfunctioning, neuroinflammation, and neuronal death. Here, we discuss the feasibility of targeting these epigenetic alterations as a new treatment strategy due to the reversibility of epigenetics and their ability to correct faulty gene expression. We also review the combined promise of stem cell therapies and epigenetic modulation in neurodegeneration, inflammation and cognitive decline. This combined approach may provide a multifaceted strategy to slow disease progression, replace lost neurons, and restore neural function. Despite challenges, including ethical, financial, and methodological barriers, ongoing research in epigenetic modulation and stem cell therapy holds promise for pioneering therapies in AD.
Collapse
Affiliation(s)
- Edhijanto Widaja
- Master's Program in Biomedical Sciences, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
- Regenerative Medicine and Research Institute of Mandaya Hospital Group, Tangerang, Indonesia
| | - Jeanne A. Pawitan
- Department of Histology, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
- Stem Cell Medical Technology Integrated Service Unit, Dr. Cipto Mangunkusumo Central Hospital, Jakarta, Indonesia
| |
Collapse
|
8
|
Kohno T, Nakagawa I, Taniguchi A, Heng F, Hattori M. Biochemical characterizations of the central fragment of human Reelin and identification of amino acid residues involved in its secretion. J Biochem 2024; 176:385-393. [PMID: 39167799 DOI: 10.1093/jb/mvae058] [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: 04/26/2024] [Revised: 07/17/2024] [Accepted: 08/16/2024] [Indexed: 08/23/2024] Open
Abstract
Secreted protein Reelin is implicated in neuropsychiatric disorders and its supplementation ameliorates neurological symptoms in mouse disease models. Recombinant human Reelin protein may be useful for the treatment of human diseases, but its properties remain uncharacterized. Here, we report that full-length human Reelin was well secreted from transfected cells and was able to induce Dab1 phosphorylation. Unexpectedly, the central fragment of human Reelin was much less secreted than that of mouse Reelin. Three residues in the sixth Reelin repeat contributed to the secretion inefficiency, and their substitutions with mouse residues increased the secretion without affecting its biological activity. Our findings help efficient production of human Reelin protein for the supplementation therapy.
Collapse
Affiliation(s)
- Takao Kohno
- Department of Biomedical Science, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi 467-8603, Japan
| | - Ikuma Nakagawa
- Department of Biomedical Science, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi 467-8603, Japan
| | - Airi Taniguchi
- Department of Biomedical Science, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi 467-8603, Japan
| | - Fang Heng
- Department of Pharmaceutical Analyses, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin 150040, China
| | - Mitsuharu Hattori
- Department of Biomedical Science, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi 467-8603, Japan
| |
Collapse
|
9
|
Katsuyama Y, Hattori M. REELIN ameliorates Alzheimer's disease, but how? Neurosci Res 2024; 208:8-14. [PMID: 39094979 DOI: 10.1016/j.neures.2024.07.004] [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: 07/19/2024] [Accepted: 07/30/2024] [Indexed: 08/04/2024]
Abstract
Alzheimer's disease (AD) is the most prevalent type of dementia; therefore, there is a high demand for therapeutic medication targeting it. In this context, extensive research has been conducted to identify molecular targets for drugs. AD manifests through two primary pathological signs: senile plaques and neurofibrillary tangles, caused by accumulations of amyloid-beta (Aβ) and phosphorylated tau, respectively. Thus, studies concerning the molecular mechanisms underlying AD etiology have primarily focused on Aβ generation and tau phosphorylation, with the anticipation of uncovering a signaling pathway impacting these molecular processes. Over the past two decades, studies using not only experimental model systems but also examining human brains have accumulated fragmentary evidences suggesting that REELIN signaling pathway is deeply involved in AD. Here, we explore REELIN signaling pathway and its involvement in memory function within the brain and review studies investigating molecular connections between REELIN signaling pathway and AD etiology. This review aims to understand how the manipulation (activation) of this pathway might ameliorate the disease's etiology.
Collapse
Affiliation(s)
- Yu Katsuyama
- Division of Neuroanatomy, Department of Anatomy, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan.
| | - Mitsuharu Hattori
- Department of Biomedical Science, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Aichi 467-8603, Japan
| |
Collapse
|
10
|
Ortí-Casañ N, Bethea JR, Eisel ULM. Mapping Alzheimer's disease: exploring cellular vulnerability and resilience. Signal Transduct Target Ther 2024; 9:299. [PMID: 39468007 PMCID: PMC11519876 DOI: 10.1038/s41392-024-02014-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 10/04/2024] [Accepted: 10/08/2024] [Indexed: 10/30/2024] Open
Affiliation(s)
- Natalia Ortí-Casañ
- Department of Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, The Netherlands
| | - John R Bethea
- Department of Anatomy and Cell Biology, The George Washington University School of Medicine and Health Sciences, Ross Hall, Washington, DC, USA
| | - Ulrich L M Eisel
- Department of Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
11
|
Hosseini K, Cediel-Ulloa A, AL-Sabri MH, Forsby A, Fredriksson R. Assessing the Neurodevelopmental Impact of Fluoxetine, Citalopram, and Paroxetine on Neural Stem Cell-Derived Neurons. Pharmaceuticals (Basel) 2024; 17:1392. [PMID: 39459031 PMCID: PMC11510426 DOI: 10.3390/ph17101392] [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: 09/06/2024] [Revised: 10/14/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Many pregnant women globally suffer from depression and are routinely prescribed selective serotonin reuptake inhibitors (SSRIs). These drugs function by blocking the re-uptake of serotonin by the serotonin transporter (SERT) into neurons, resulting in its accumulation in the presynaptic cleft. Despite a large amount of research suggesting a potential link to neurodevelopmental disorders in children whose mothers took these drugs during pregnancy, their possible adverse effects are still debated, and results are contradictory. On the other hand, there is an immediate need for improved cell-based models for developmental neurotoxicity studies (DNT) to minimize the use of animals in research. METHODS In this study, we aimed to assess the effects of clinically relevant concentrations of paroxetine (PAR), fluoxetine (FLX), and citalopram (CIT)-on maturing neurons derived from human neural stem cells using multiple endpoints. RESULTS Although none of the tested concentrations of FLX, CIT, or PAR significantly affected cell viability, FLX (10 µM) exhibited the highest reduction in viability compared to the other drugs. Regarding neurite outgrowth, CIT did not have a significant effect. However, FLX (10 µM) significantly reduced both mean neurite outgrowth and mean processes, PAR significantly reduced mean processes, and showed a trend of dysregulation of multiple genes associated with neuronal development at therapeutic-relevant serum concentrations. CONCLUSIONS Transcriptomic data and uptake experiments found no SERT activity in the system, suggesting that the adverse effects of FLX and PAR are independent of SERT.
Collapse
Affiliation(s)
- Kimia Hosseini
- Department of Pharmaceutical Bioscience, Uppsala University, 751 24 Uppsala, Sweden (R.F.)
| | - Andrea Cediel-Ulloa
- Department of Organismal Biology, Uppsala University, 752 36 Uppsala, Sweden
| | - Mohamed H. AL-Sabri
- Department of Pharmaceutical Bioscience, Uppsala University, 751 24 Uppsala, Sweden (R.F.)
- Department of Surgical Science, Functional Pharmacology and Neuroscience, Uppsala University, 751 24 Uppsala, Sweden
| | - Anna Forsby
- Department of Biochemistry and Biophysics, Stockholm University, 106 91 Stockholm, Sweden
| | - Robert Fredriksson
- Department of Pharmaceutical Bioscience, Uppsala University, 751 24 Uppsala, Sweden (R.F.)
| |
Collapse
|
12
|
Al-Beltagi M, Saeed NK, Bediwy AS, Bediwy EA, Elbeltagi R. Decoding the genetic landscape of autism: A comprehensive review. World J Clin Pediatr 2024; 13:98468. [PMID: 39350903 PMCID: PMC11438927 DOI: 10.5409/wjcp.v13.i3.98468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 07/29/2024] [Accepted: 08/01/2024] [Indexed: 08/30/2024] Open
Abstract
BACKGROUND Autism spectrum disorder (ASD) is a complex neurodevelopmental condition characterized by heterogeneous symptoms and genetic underpinnings. Recent advancements in genetic and epigenetic research have provided insights into the intricate mechanisms contributing to ASD, influencing both diagnosis and therapeutic strategies. AIM To explore the genetic architecture of ASD, elucidate mechanistic insights into genetic mutations, and examine gene-environment interactions. METHODS A comprehensive systematic review was conducted, integrating findings from studies on genetic variations, epigenetic mechanisms (such as DNA methylation and histone modifications), and emerging technologies [including Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)-Cas9 and single-cell RNA sequencing]. Relevant articles were identified through systematic searches of databases such as PubMed and Google Scholar. RESULTS Genetic studies have identified numerous risk genes and mutations associated with ASD, yet many cases remain unexplained by known factors, suggesting undiscovered genetic components. Mechanistic insights into how these genetic mutations impact neural development and brain connectivity are still evolving. Epigenetic modifications, particularly DNA methylation and non-coding RNAs, also play significant roles in ASD pathogenesis. Emerging technologies like CRISPR-Cas9 and advanced bioinformatics are advancing our understanding by enabling precise genetic editing and analysis of complex genomic data. CONCLUSION Continued research into the genetic and epigenetic underpinnings of ASD is crucial for developing personalized and effective treatments. Collaborative efforts integrating multidisciplinary expertise and international collaborations are essential to address the complexity of ASD and translate genetic discoveries into clinical practice. Addressing unresolved questions and ethical considerations surrounding genetic research will pave the way for improved diagnostic tools and targeted therapies, ultimately enhancing outcomes for individuals affected by ASD.
Collapse
Affiliation(s)
- Mohammed Al-Beltagi
- Department of Pediatric, Faculty of Medicine, Tanta University, Alghrabia, Tanta 31511, Egypt
- Department of Pediatric, University Medical Center, King Abdulla Medical City, Arabian Gulf University, Manama 26671, Bahrain
| | - Nermin Kamal Saeed
- Medical Microbiology Section, Department of Pathology, Salmaniya Medical Complex, Ministry of Health, Kingdom of Bahrain, Manama 12, Bahrain
- Medical Microbiology Section, Department of Pathology, Irish Royal College of Surgeon, Muharraq, Busaiteen 15503, Bahrain
| | - Adel Salah Bediwy
- Department of Pulmonology, Faculty of Medicine, Tanta University, Alghrabia, Tanta 31527, Egypt
- Department of Pulmonology, University Medical Center, King Abdulla Medical City, Arabian Gulf University, Manama 26671, Bahrain
| | - Eman A Bediwy
- Internal Medicine, Faculty of Medicine, Tanta University, Algharbia, Tanta 31527, Egypt
| | - Reem Elbeltagi
- Department of Medicine, The Royal College of Surgeons in Ireland-Bahrain, Muharraq, Busiateen 15503, Bahrain
| |
Collapse
|
13
|
Park HR, Cai M, Yang EJ. Herbal Formula Extract Ameliorates Anxiety and Cognitive Impairment via Regulation of the Reelin/Dab-1 Pathway in a Murine Model of Post-Traumatic Stress Disorder. Pharmaceutics 2024; 16:1150. [PMID: 39339187 PMCID: PMC11434737 DOI: 10.3390/pharmaceutics16091150] [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: 07/31/2024] [Revised: 08/27/2024] [Accepted: 08/27/2024] [Indexed: 09/30/2024] Open
Abstract
We investigated the effects of epigenetic modifications on post-traumatic stress disorder (PTSD) using a novel combination of herbal medicines from Panax ginseng, Astragalus membranaceus, Atractylodes macrocephala, and Glycyrrhiza uralensis. The herbal formula extract (HFE) (250 mg/kg) was administered orally once daily for 14 days to determine its effects on PTSD in mice by combining prolonged stress and foot shock. The open field and Y-maze tests determined the effect of HFE on PTSD-induced anxiety and cognition. Hippocampal neuronal plastic changes and molecular mechanism were verified. Treatment with HFE decreased anxiety-like behavior and enhanced cognition. Moreover, it reduced the number of PTSD-related hilar ectopic granule cells in the dentate gyrus (DG). PTSD mice showed reduced neuronal plasticity of doublecortin+ cells in the DG, which was restored by HFE treatment. HFE reversed PTSD-induced inhibition of the Reelin/Dab1 pathway, a critical signaling cascade involved in brain development, and regulated Reelin methylation. Furthermore, DNA methylation, methyl-CpG binding protein 2, and DNA methyltransferase 1, which were elevated in the hippocampus of PTSD mice, were restored following HFE treatment. HFE increased the expression of synaptic plasticity-related factors in the hippocampus of PTSD mice. Our findings suggest that HFE can facilitate PTSD treatment by alleviating behavioral abnormalities through the restoration of hippocampal dysfunction via regulation of the Reelin/Dab-1 pathway and DNA methylation in the hippocampus.
Collapse
Affiliation(s)
| | | | - Eun Jin Yang
- Department of KM Science Research, Korea Institute of Oriental Medicine (KIOM), Daejeon 34054, Republic of Korea; (H.R.P.); (M.C.)
| |
Collapse
|
14
|
Fuentealba LM, Pizarro H, Marzolo MP. OCRL1 Deficiency Affects the Intracellular Traffic of ApoER2 and Impairs Reelin-Induced Responses. Biomolecules 2024; 14:799. [PMID: 39062513 PMCID: PMC11274606 DOI: 10.3390/biom14070799] [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: 05/29/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/28/2024] Open
Abstract
Lowe Syndrome (LS) is a rare X-linked disorder characterized by renal dysfunction, cataracts, and several central nervous system (CNS) anomalies. The mechanisms underlying the neurological dysfunction in LS remain unclear, albeit they share some phenotypic characteristics similar to the deficiency or dysfunction of the Reelin signaling, a relevant pathway with roles in CNS development and neuronal functions. In this study, we investigated the role of OCRL1, an inositol polyphosphate 5-phosphatase encoded by the OCRL gene, mutated in LS, focusing on its impact on endosomal trafficking and receptor recycling in human neuronal cells. Specifically, we tested the effects of OCRL1 deficiency in the trafficking and signaling of ApoER2/LRP8, a receptor for the ligand Reelin. We found that loss of OCRL1 impairs ApoER2 intracellular trafficking, leading to reduced receptor expression and decreased levels at the plasma membrane. Additionally, human neurons deficient in OCRL1 showed impairments in ApoER2/Reelin-induced responses. Our findings highlight the critical role of OCRL1 in regulating ApoER2 endosomal recycling and its impact on the ApoER2/Reelin signaling pathway, providing insights into potential mechanisms underlying the neurological manifestations of LS.
Collapse
Affiliation(s)
| | | | - María-Paz Marzolo
- Laboratorio de Tráfico Intracelular y Señalización, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 7810128, Chile; (L.M.F.); (H.P.)
| |
Collapse
|
15
|
Park HR, Cai M. Antiseizure effects of Lilii Bulbus on pentylenetetrazol kindling-induced seizures in mice: Involvement of Reelin, Netrin-1, and semaphorin. Biomed Pharmacother 2024; 173:116385. [PMID: 38460369 DOI: 10.1016/j.biopha.2024.116385] [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: 12/19/2023] [Revised: 02/23/2024] [Accepted: 03/06/2024] [Indexed: 03/11/2024] Open
Abstract
Lilii Bulbus (Lilium lancifolium Thunberg) has a proneurogenic effect on the hippocampus. However, its effects on epilepsy and associated pathological features remain unknown. In this study, we investigated the antiseizure effects of a water extract of Lilii Bulbus (WELB) in mouse model of pentylenetetrazol (PTZ)-induced seizure. Mice were injected with PTZ once every 48 h until full kindling was achieved. WELB (100 and 500 mg/kg) was orally administered once daily before PTZ administration and during the kindling process. We found that WELB treatment protected against PTZ-induced low seizure thresholds and high seizure severity. Further, WELB-treated mice showed attenuated PTZ kindling-induced anxiety and memory impairment. Immunostaining and immunoblots showed that hyperactivation and ectopic migration of dentate granule cells (DGCs) were significantly reduced by WELB treatment in PTZ kindling-induced seizure mice. Staining for mossy fiber sprouting (MFS) using Timm staining and ZnT3 showed that WELB treatment significantly decreased PTZ kindling-induced MFS. Furthermore, the increased or decreased expression of proteins related to ectopic DGCs (Reelin and Dab-1), MFS (Netrin-1, Sema3A, and Sema3F), and their downstream effectors (ERK, AKT, and CREB) in the hippocampus of PTZ kindling mice was significantly restored by WELB treatment. Overall, our findings suggest that WELB is a potential antiseizure drug that acts by reducing ectopic DGCs and MFS and modulating epileptogenesis-related signaling in the hippocampus.
Collapse
Affiliation(s)
- Hee Ra Park
- Department of KM Medicine Science Research Division, Korea Institute of Oriental Medicine (KIOM), Daejeon, Republic of Korea.
| | - Mudan Cai
- Department of KM Medicine Science Research Division, Korea Institute of Oriental Medicine (KIOM), Daejeon, Republic of Korea
| |
Collapse
|
16
|
Calvier L, Alexander A, Marckx AT, Kounnas MZ, Durakoglugil M, Herz J. Safety of Anti-Reelin Therapeutic Approaches for Chronic Inflammatory Diseases. Cells 2024; 13:583. [PMID: 38607022 PMCID: PMC11011630 DOI: 10.3390/cells13070583] [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: 02/23/2024] [Revised: 03/18/2024] [Accepted: 03/25/2024] [Indexed: 04/13/2024] Open
Abstract
Reelin, a large extracellular glycoprotein, plays critical roles in neuronal development and synaptic plasticity in the central nervous system (CNS). Recent studies have revealed non-neuronal functions of plasma Reelin in inflammation by promoting endothelial-leukocyte adhesion through its canonical pathway in endothelial cells (via ApoER2 acting on NF-κB), as well as in vascular tone regulation and thrombosis. In this study, we have investigated the safety and efficacy of selectively depleting plasma Reelin as a potential therapeutic strategy for chronic inflammatory diseases. We found that Reelin expression remains stable throughout adulthood and that peripheral anti-Reelin antibody treatment with CR-50 efficiently depletes plasma Reelin without affecting its levels or functionality within the CNS. Notably, this approach preserves essential neuronal functions and synaptic plasticity. Furthermore, in mice induced with experimental autoimmune encephalomyelitis (EAE), selective modulation of endothelial responses by anti-Reelin antibodies reduces pathological leukocyte infiltration without completely abolishing diapedesis. Finally, long-term Reelin depletion under metabolic stress induced by a Western diet did not negatively impact the heart, kidney, or liver, suggesting a favorable safety profile. These findings underscore the promising role of peripheral anti-Reelin therapeutic strategies for autoimmune diseases and conditions where endothelial function is compromised, offering a novel approach that may avoid the immunosuppressive side effects associated with conventional anti-inflammatory therapies.
Collapse
Affiliation(s)
- Laurent Calvier
- Department of Molecular Genetics, University of Texas (UT) Southwestern Medical Center, Dallas, TX 75390, USA (A.T.M.); (M.D.); (J.H.)
- Center for Translational Neurodegeneration Research, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Anna Alexander
- Department of Molecular Genetics, University of Texas (UT) Southwestern Medical Center, Dallas, TX 75390, USA (A.T.M.); (M.D.); (J.H.)
- Center for Translational Neurodegeneration Research, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Austin T. Marckx
- Department of Molecular Genetics, University of Texas (UT) Southwestern Medical Center, Dallas, TX 75390, USA (A.T.M.); (M.D.); (J.H.)
- Center for Translational Neurodegeneration Research, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | | | - Murat Durakoglugil
- Department of Molecular Genetics, University of Texas (UT) Southwestern Medical Center, Dallas, TX 75390, USA (A.T.M.); (M.D.); (J.H.)
- Center for Translational Neurodegeneration Research, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Joachim Herz
- Department of Molecular Genetics, University of Texas (UT) Southwestern Medical Center, Dallas, TX 75390, USA (A.T.M.); (M.D.); (J.H.)
- Center for Translational Neurodegeneration Research, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Neurology and Neurotherapeutics, UT Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
17
|
Tolar M, Hey JA, Power A, Abushakra S. The Single Toxin Origin of Alzheimer's Disease and Other Neurodegenerative Disorders Enables Targeted Approach to Treatment and Prevention. Int J Mol Sci 2024; 25:2727. [PMID: 38473975 PMCID: PMC10932387 DOI: 10.3390/ijms25052727] [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: 01/15/2024] [Revised: 02/20/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
New data suggest that the aggregation of misfolded native proteins initiates and drives the pathogenic cascade that leads to Alzheimer's disease (AD) and other age-related neurodegenerative disorders. We propose a unifying single toxin theory of brain neurodegeneration that identifies new targets and approaches to the development of disease-modifying treatments. An extensive body of genetic evidence suggests soluble aggregates of beta-amyloid (Aβ) as the primary neurotoxin in the pathogenesis of AD. New insights from fluid biomarkers, imaging, and clinical studies provide further evidence for the decisive impact of toxic Aβ species in the initiation and progression of AD. Understanding the distinct roles of soluble and insoluble amyloid aggregates on AD pathogenesis has been the key missing piece of the Alzheimer's puzzle. Data from clinical trials with anti-amyloid agents and recent advances in the diagnosis of AD demonstrate that the driving insult in biologically defined AD is the neurotoxicity of soluble Aβ aggregates, called oligomers and protofibrils, rather than the relatively inert insoluble mature fibrils and amyloid plaques. Amyloid oligomers appear to be the primary factor causing the synaptic impairment, neuronal stress, spreading of tau pathology, and eventual cell death that lead to the clinical syndrome of AD dementia. All other biochemical effects and neurodegenerative changes in the brain that are observed in AD are a response to or a downstream effect of this initial toxic insult by oligomers. Other neurodegenerative disorders follow a similar pattern of pathogenesis, in which normal brain proteins with important biological functions become trapped in the aging brain due to impaired clearance and then misfold and aggregate into neurotoxic species that exhibit prion-like behavior. These aggregates then spread through the brain and cause disease-specific neurodegeneration. Targeting the inhibition of this initial step in neurodegeneration by blocking the misfolding and aggregation of healthy proteins has the potential to slow or arrest disease progression, and if treatment is administered early in the course of AD and other neurodegenerative disorders, it may delay or prevent the onset of clinical symptoms.
Collapse
|
18
|
Markiewicz R, Markiewicz-Gospodarek A, Borowski B, Trubalski M, Łoza B. Reelin Signaling and Synaptic Plasticity in Schizophrenia. Brain Sci 2023; 13:1704. [PMID: 38137152 PMCID: PMC10741648 DOI: 10.3390/brainsci13121704] [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: 11/15/2023] [Revised: 12/01/2023] [Accepted: 12/05/2023] [Indexed: 12/24/2023] Open
Abstract
Recent research emphasizes the significance of studying the quality of life of schizophrenia patients, considering the complex nature of the illness. Identifying neuronal markers for early diagnosis and treatment is crucial. Reelin (RELN) stands out among these markers, with genetic studies highlighting its role in mental health. Suppression of RELN expression may contribute to cognitive deficits by limiting dendritic proliferation, affecting neurogenesis, and leading to improper neuronal circuits. Although the physiological function of reelin is not fully understood, it plays a vital role in hippocampal cell stratification and neuroglia formation. This analysis explores reelin's importance in the nervous system, shedding light on its impact on mental disorders such as schizophrenia, paving the way for innovative therapeutic approaches, and at the same time, raises the following conclusions: increased methylation levels of the RELN gene in patients with a diagnosis of schizophrenia results in a multiple decrease in the expression of reelin, and monitoring of this indicator, i.e., methylation levels, can be used to monitor the severity of symptoms in the course of schizophrenia.
Collapse
Affiliation(s)
- Renata Markiewicz
- Occupational Therapy Laboratory, Chair of Nursing Development, Medical University of Lublin, 4 Staszica St., 20-081 Lublin, Poland;
| | | | - Bartosz Borowski
- Students Scientific Association, Department of Normal, Clinical and Imaging Anatomy, Medical University of Lublin, 20-090 Lublin, Poland; (B.B.); (M.T.)
| | - Mateusz Trubalski
- Students Scientific Association, Department of Normal, Clinical and Imaging Anatomy, Medical University of Lublin, 20-090 Lublin, Poland; (B.B.); (M.T.)
| | - Bartosz Łoza
- Department of Psychiatry, Medical University of Warsaw, 02-091 Warsaw, Poland;
| |
Collapse
|