ijms-logo

Journal Browser

Journal Browser

Molecular Signals and Genetic Regulations of Neurological Disorders

A special issue of International Journal of Molecular Sciences (ISSN 1422-0067). This special issue belongs to the section "Molecular Genetics and Genomics".

Deadline for manuscript submissions: closed (31 August 2022) | Viewed by 18624

Special Issue Editors


E-Mail Website
Guest Editor
1. UOC Geriatra - Disturbi Cognitivi e Demenze, Dipartimento di Cure Primarie. AUSL Modena, 41012 Modena, Italy
2. Department of Geriatrics, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica Sacro Cuore, 00168 Rome, Italy
Interests: modifiable lifestyle factors and plasma biomarkers of Alzheimer’s disease
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues,

Intra- and extracellular molecular signaling play a major role in modulating genetic and epigenetic regulation of cellular growth, differentiation mechanisms, and vice versa. Moreover, the proteins that make up an organism (proteome) are constantly changing and are intricately linked to neurological diseases. A growing body of research data strongly suggests that imbalance in the abovementioned mechanism and genetic abnormalities are at the basis of the development of several neurological disorders. Thus, the scope of this Special Issue is to cover molecular signaling and genetic regulation of neurological disorders, particularly regarding the development and progression of the most prevalent neurological disorders, which are the causes of cognitive impairment, movement disorders, cerebrovascular diseases, and brain cancer. We are also interested in discussing neurological disorders such as Down Syndrome, since it shows some molecular similarity with Alzheimer’s disease, and repeat expansion diseases.

Dr. Emanuele Rocco Villani
Prof. Dr. Emanuele Marzetti
Guest Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. International Journal of Molecular Sciences is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. There is an Article Processing Charge (APC) for publication in this open access journal. For details about the APC please see here. Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • mTOR pathways
  • Alzheimer’s disease, aging, biomarkers, cognitive decline, cytokines, metabolomics, neuroinflammation
  • Parkinson’s disease
  • Cerebrovascular diseases, CADASIL, HTR1A
  • Mitochondrial dysfunction, mitochondrial quality control, mitochondrial-lysosomal axis, mitophagy, mtDNA
  • proteomics
  • exosomes
  • extracellular vesicles
  • Chromosome 21 trisomy, Down’s syndrome, DYRK1-A

Published Papers (8 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Editorial

Jump to: Research

4 pages, 645 KiB  
Editorial
Molecular Signals and Genetic Regulations of Neurological Disorders
by Emanuele Rocco Villani and Emanuele Marzetti
Int. J. Mol. Sci. 2023, 24(6), 5902; https://doi.org/10.3390/ijms24065902 - 21 Mar 2023
Cited by 1 | Viewed by 901
Abstract
Neurological disorders are a large and heterogeneous field of research that can be tackled through a variety of approaches, ranging from epidemiology to molecular biology, through clinical, biostatistical, and laboratory experiments [...] Full article
(This article belongs to the Special Issue Molecular Signals and Genetic Regulations of Neurological Disorders)
Show Figures

Figure 1

Research

Jump to: Editorial

17 pages, 4936 KiB  
Article
Data-Driven Approaches Used for Compound Library Design for the Treatment of Parkinson’s Disease
by Oscar Barrera-Vazquez, Jose Alberto Santiago-de-la-Cruz, Nadia Alejandra Rivero-Segura, Edgar Antonio Estrella-Parra, Genaro Salvador Morales-Paoli, Edgar Flores-Soto and Juan Carlos Gomez-Verjan
Int. J. Mol. Sci. 2023, 24(2), 1134; https://doi.org/10.3390/ijms24021134 - 06 Jan 2023
Cited by 1 | Viewed by 1889
Abstract
Parkinson’s disease (PD) is the second most common neurodegenerative disease in older individuals worldwide. Pharmacological treatment for such a disease consists of drugs such as monoamine oxidase B (MAO-B) inhibitors to increase dopamine concentration in the brain. However, such drugs have adverse reactions [...] Read more.
Parkinson’s disease (PD) is the second most common neurodegenerative disease in older individuals worldwide. Pharmacological treatment for such a disease consists of drugs such as monoamine oxidase B (MAO-B) inhibitors to increase dopamine concentration in the brain. However, such drugs have adverse reactions that limit their use for extended periods; thus, the design of less toxic and more efficient compounds may be explored. In this context, cheminformatics and computational chemistry have recently contributed to developing new drugs and the search for new therapeutic targets. Therefore, through a data-driven approach, we used cheminformatic tools to find and optimize novel compounds with pharmacological activity against MAO-B for treating PD. First, we retrieved from the literature 3316 original articles published between 2015–2021 that experimentally tested 215 natural compounds against PD. From such compounds, we built a pharmacological network that showed rosmarinic acid, chrysin, naringenin, and cordycepin as the most connected nodes of the network. From such compounds, we performed fingerprinting analysis and developed evolutionary libraries to obtain novel derived structures. We filtered these compounds through a docking test against MAO-B and obtained five derived compounds with higher affinity and lead likeness potential. Then we evaluated its antioxidant and pharmacokinetic potential through a docking analysis (NADPH oxidase and CYP450) and physiologically-based pharmacokinetic (PBPK modeling). Interestingly, only one compound showed dual activity (antioxidant and MAO-B inhibitors) and pharmacokinetic potential to be considered a possible candidate for PD treatment and further experimental analysis. Full article
(This article belongs to the Special Issue Molecular Signals and Genetic Regulations of Neurological Disorders)
Show Figures

Figure 1

18 pages, 4072 KiB  
Article
12/15-Lipoxygenase Regulation of Diabetic Cognitive Dysfunction Is Determined by Interfering with Inflammation and Cell Apoptosis
by Qi Chen, Qixue Zheng, Yang Yang, Ying Luo, Hong Wang, Huan Li, Lu Yang, Congli Hu, Jiahua Zhang, Yuke Li, Hui Xia, Zhihao Chen, Jie Ma, Xiaoyan Tian and Junqing Yang
Int. J. Mol. Sci. 2022, 23(16), 8997; https://doi.org/10.3390/ijms23168997 - 12 Aug 2022
Cited by 7 | Viewed by 1944
Abstract
This study aimed to discuss the role of 12/15-lipoxygenase (12/15-LOX) regulation involved in diabetes cognitive dysfunction. First, Mini Mental State Examination (MMSE) test was used to evaluate cognitive ability in diabetic patients and normal controls. The plasma test showed that the plasma level [...] Read more.
This study aimed to discuss the role of 12/15-lipoxygenase (12/15-LOX) regulation involved in diabetes cognitive dysfunction. First, Mini Mental State Examination (MMSE) test was used to evaluate cognitive ability in diabetic patients and normal controls. The plasma test showed that the plasma level of 12/15-LOX in patients with MMSE scores below 27 was significantly increased compared with that of the normal group. Second, 12/15-LOX inhibitor was administered to diabetic rats. Behavioral tests, biochemistry, enzyme-linked immunosorbent assays, and Western blotting were used in this study. We found that the levels of fasting and random blood glucose increased rapidly in diabetic rats, the levels of triglycerides and total cholesterol in the diabetic group increased, and insulin levels decreased significantly. In the Morris water maze test, the escape latency was prolonged, and the crossing times decreased in the diabetic group. Under the microscope, the apoptosis of hippocampal neurons in diabetic rats increased significantly. The levels of TNF-α, IL-6 and 12-hydroxyindoleic acid (12(S)-HETE) significantly increased, and the protein expression of 12/15-LOX, p38 MAPK, Aβ1-42, caspase-3, caspase-9 and cPLA2 increased, while that of Bcl-2 decreased. However, the use of 12/15-LOX inhibitor reversed these results. Third, 12/15-LOX shRNA and p38MAPK inhibitor were administered to HT22 cells in high-glucose medium. The results of the cell experiment were consistent with those of the animal experiment. Our results indicated that the 12/15-LOX pathway participates in diabetic brain damage by activating p38MAPK to promote inflammation and neuronal apoptosis, and intervention 12/15-LOX can improve diabetic cognitive dysfunction. Full article
(This article belongs to the Special Issue Molecular Signals and Genetic Regulations of Neurological Disorders)
Show Figures

Figure 1

27 pages, 5396 KiB  
Article
Maternal Prenatal Inflammation Increases Brain Damage Susceptibility of Lipopolysaccharide in Adult Rat Offspring via COX-2/PGD-2/DPs Pathway Activation
by Jiahua Zhang, Peishuang Yao, Wenli Han, Ying Luo, Yuke Li, Yang Yang, Hui Xia, Zhihao Chen, Qi Chen, Hong Wang, Lu Yang, Huan Li, Congli Hu, Haifeng Huang, Zhe Peng, Xiaodan Tan, Miaomiao Li and Junqing Yang
Int. J. Mol. Sci. 2022, 23(11), 6142; https://doi.org/10.3390/ijms23116142 - 30 May 2022
Cited by 1 | Viewed by 2018
Abstract
A growing body of research suggests that inflammatory insult contributes to the etiology of central nervous system diseases, such as depression, Alzheimer’s disease, and so forth. However, the effect of prenatal systemic inflammation exposure on offspring brain development and cerebral susceptibility to inflammatory [...] Read more.
A growing body of research suggests that inflammatory insult contributes to the etiology of central nervous system diseases, such as depression, Alzheimer’s disease, and so forth. However, the effect of prenatal systemic inflammation exposure on offspring brain development and cerebral susceptibility to inflammatory insult remains unknown. In this study, we utilized the prenatal inflammatory insult model in vivo and the neuronal damage model in vitro. The results obtained show that prenatal maternal inflammation exacerbates LPS-induced memory impairment, neuronal necrosis, brain inflammatory response, and significantly increases protein expressions of COX-2, DP2, APP, and Aβ, while obviously decreasing that of DP1 and the exploratory behaviors of offspring rats. Meloxicam significantly inhibited memory impairment, neuronal necrosis, oxidative stress, and inflammatory response, and down-regulated the expressions of APP, Aβ, COX-2, and DP2, whereas significantly increased exploring behaviors and the expression of DP1 in vivo. Collectively, these findings suggested that maternal inflammation could cause offspring suffering from inflammatory and behavioral disorders and increase the susceptibility of offspring to cerebral pathological factors, accompanied by COX-2/PGD-2/DPs pathway activation, which could be ameliorated significantly by COX-2 inhibitor meloxicam treatment. Full article
(This article belongs to the Special Issue Molecular Signals and Genetic Regulations of Neurological Disorders)
Show Figures

Figure 1

12 pages, 987 KiB  
Communication
Rev-erbα Knockout Reduces Ethanol Consumption and Preference in Male and Female Mice
by Yasmine Al-Sabagh, Hayley Hope Allyssa Thorpe, Bryan William Jenkins, Shahnaza Hamidullah, Malik Asfandyaar Talhat, Cara Beth Suggett, Cristine Joelle Reitz, Mina Rasouli, Tami Avril Martino and Jibran Younis Khokhar
Int. J. Mol. Sci. 2022, 23(9), 5197; https://doi.org/10.3390/ijms23095197 - 06 May 2022
Cited by 1 | Viewed by 2240
Abstract
Alcohol use is a contributor in the premature deaths of approximately 3 million people annually. Among the risk factors for alcohol misuse is circadian rhythm disruption; however, this connection remains poorly understood. Inhibition of the circadian nuclear receptor REV-ERBα is known to disrupt [...] Read more.
Alcohol use is a contributor in the premature deaths of approximately 3 million people annually. Among the risk factors for alcohol misuse is circadian rhythm disruption; however, this connection remains poorly understood. Inhibition of the circadian nuclear receptor REV-ERBα is known to disrupt molecular feedback loops integral to daily oscillations, and impact diurnal fluctuations in the expression of proteins required for reward-related neurotransmission. However, the role of REV-ERBα in alcohol and substance use-related phenotypes is unknown. Herein, we used a Rev-erbα knockout mouse line and ethanol two-bottle choice preference testing to show that disruption of Rev-erbα reduces ethanol preference in male and female mice. Rev-erbα null mice showed the lowest ethanol preference in a two-bottle choice test across all genotypes, whereas there were no ethanol preference differences between heterozygotes and wildtypes. In a separate experiment, alcohol-consuming wildtype C57Bl/6N mice were administered the REV-ERBα/β inhibitor SR8278 (25 mg/kg or 50 mg/kg) for 7 days and alcohol preference was evaluated daily. No differences in alcohol preference were observed between the treatment and vehicle groups. Our data provides evidence that genetic variation in REV-ERBα may contribute to differences in alcohol drinking. Full article
(This article belongs to the Special Issue Molecular Signals and Genetic Regulations of Neurological Disorders)
Show Figures

Figure 1

14 pages, 3975 KiB  
Article
Interleukin-4 Aggravates LPS-Induced Striatal Neurodegeneration In Vivo via Oxidative Stress and Polarization of Microglia/Macrophages
by Jaegeun Jang, Ahreum Hong, Youngcheul Chung and Byungkwan Jin
Int. J. Mol. Sci. 2022, 23(1), 571; https://doi.org/10.3390/ijms23010571 - 05 Jan 2022
Cited by 7 | Viewed by 2973
Abstract
The present study investigated the effects of interleukin (IL)-4 on striatal neurons in lipopolysaccharide (LPS)-injected rat striatum in vivo. Either LPS or PBS as a control was unilaterally injected into the striatum, and brain tissues were processed for immunohistochemical and Nissl staining or [...] Read more.
The present study investigated the effects of interleukin (IL)-4 on striatal neurons in lipopolysaccharide (LPS)-injected rat striatum in vivo. Either LPS or PBS as a control was unilaterally injected into the striatum, and brain tissues were processed for immunohistochemical and Nissl staining or for hydroethidine histochemistry at the indicated time points after LPS injection. Analysis by NeuN and Nissl immunohistochemical staining showed a significant loss of striatal neurons at 1, 3, and 7 days post LPS. In parallel, IL-4 immunoreactivity was upregulated as early as 1 day, reached a peak at 3 days, and was sustained up to 7 days post LPS. Increased levels of IL-4 immunoreactivity were exclusively detected in microglia/macrophages, but not in neurons nor astrocytes. The neutralizing antibody (NA) for IL-4 significantly protects striatal neurons against LPS-induced neurotoxicity in vivo. Accompanying neuroprotection, IL-4NA inhibited activation of microglia/macrophages, production of reactive oxygen species (ROS), ROS-derived oxidative damage and nitrosative stress, and produced polarization of microglia/macrophages shifted from M1 to M2. These results suggest that endogenous IL-4 expressed in LPS-activated microglia/macrophages contributes to striatal neurodegeneration in which oxidative/nitrosative stress and M1/M2 polarization are implicated. Full article
(This article belongs to the Special Issue Molecular Signals and Genetic Regulations of Neurological Disorders)
Show Figures

Figure 1

18 pages, 6618 KiB  
Article
proNGF Involvement in the Adult Neurogenesis Dysfunction in Alzheimer’s Disease
by Bolanle Fatimat Olabiyi, Catherine Fleitas, Bahira Zammou, Isidro Ferrer, Claire Rampon, Joaquim Egea and Carme Espinet
Int. J. Mol. Sci. 2021, 22(19), 10744; https://doi.org/10.3390/ijms221910744 - 04 Oct 2021
Cited by 4 | Viewed by 2252
Abstract
In recent decades, neurogenesis in the adult brain has been well demonstrated in a number of animal species, including humans. Interestingly, work with rodents has shown that adult neurogenesis in the dentate gyrus (DG) of the hippocampus is vital for some cognitive aspects, [...] Read more.
In recent decades, neurogenesis in the adult brain has been well demonstrated in a number of animal species, including humans. Interestingly, work with rodents has shown that adult neurogenesis in the dentate gyrus (DG) of the hippocampus is vital for some cognitive aspects, as increasing neurogenesis improves memory, while its disruption triggers the opposite effect. Adult neurogenesis declines with age and has been suggested to play a role in impaired progressive learning and memory loss seen in Alzheimer’s disease (AD). Therefore, therapeutic strategies designed to boost adult hippocampal neurogenesis may be beneficial for the treatment of AD. The precursor forms of neurotrophins, such as pro-NGF, display remarkable increase during AD in the hippocampus and entorhinal cortex. In contrast to mature NGF, pro-NGF exerts adverse functions in survival, proliferation, and differentiation. Hence, we hypothesized that pro-NGF and its p75 neurotrophin receptor (p75NTR) contribute to disrupting adult hippocampal neurogenesis during AD. To test this hypothesis, in this study, we took advantage of the availability of mouse models of AD (APP/PS1), which display memory impairment, and AD human samples to address the role of pro-NGF/p75NTR signaling in different aspects of adult neurogenesis. First, we observed that DG doublecortin (DCX) + progenitors express p75NTR both, in healthy humans and control animals, although the percentage of DCX+ cells are significantly reduced in AD. Interestingly, the expression of p75NTR in these progenitors is significantly decreased in AD conditions compared to controls. In order to assess the contribution of the pro-NGF/p75NTR pathway to the memory deficits of APP/PS1 mice, we injected pro-NGF neutralizing antibodies (anti-proNGF) into the DG of control and APP/PS1 mice and animals are subjected to a Morris water maze test. Intriguingly, we observed that anti-pro-NGF significantly restored memory performance of APP/PS1 animals and significantly increase the percentage of DCX+ progenitors in the DG region of these animals. In summary, our results suggest that pro-NGF is involved in disrupting spatial memory in AD, at least in part by blocking adult neurogenesis. Moreover, we propose that adult neurogenesis alteration should be taken into consideration for better understanding of AD pathology. Additionally, we provide a new molecular entry point (pro-NGF/p75NTR signaling) as a promising therapeutic target in AD. Full article
(This article belongs to the Special Issue Molecular Signals and Genetic Regulations of Neurological Disorders)
Show Figures

Figure 1

17 pages, 21025 KiB  
Article
The E3 Ubiquitin Ligase NEDD4-1 Mediates Temozolomide-Resistant Glioblastoma through PTEN Attenuation and Redox Imbalance in Nrf2–HO-1 Axis
by Hao-Yu Chuang, Li-Yun Hsu, Chih-Ming Pan, Narpati Wesa Pikatan, Vijesh Kumar Yadav, Iat-Hang Fong, Chao-Hsuan Chen, Chi-Tai Yeh and Shao-Chih Chiu
Int. J. Mol. Sci. 2021, 22(19), 10247; https://doi.org/10.3390/ijms221910247 - 23 Sep 2021
Cited by 18 | Viewed by 3180
Abstract
Background: Glioblastoma (GBM) is the most common primary malignant brain tumor in adults. It is highly resistant to chemotherapy, and tumor recurrence is common. Neuronal precursor cell-expressed developmentally downregulated 4-1 (NEDD4-1) is an E3 ligase that controls embryonic development and animal growth. NEDD4-1 [...] Read more.
Background: Glioblastoma (GBM) is the most common primary malignant brain tumor in adults. It is highly resistant to chemotherapy, and tumor recurrence is common. Neuronal precursor cell-expressed developmentally downregulated 4-1 (NEDD4-1) is an E3 ligase that controls embryonic development and animal growth. NEDD4-1 regulates the tumor suppressor phosphatase and tensin homolog (PTEN), one of the major regulators of the PI3K/AKT/mTOR signaling axis, as well as the response to oxidative stress. Methods: The expression levels of NEDD4-1 in GBM tissues and different cell lines were determined by quantitative real-time polymerase chain reaction and immunohistochemistry. In vitro and in vivo assays were performed to explore the biological effects of NEDD4-1 on GBM cells. Temozolomide (TMZ)-resistant U87MG and U251 cell lines were specifically established to determine NEDD4-1 upregulation and its effects on the tumorigenicity of GBM cells. Subsequently, miRNA expression in TMZ-resistant cell lines was investigated to determine the dysregulated miRNA underlying the overexpression of NEDD4-1. Indole-3-carbinol (I3C) was used to inhibit NEDD4-1 activity, and its effect on chemoresistance to TMZ was verified. Results: NEDD4-1 was significantly overexpressed in the GBM and TMZ-resistant cells and clinical samples. NEDD4-1 was demonstrated to be a key oncoprotein associated with TMZ resistance, inducing oncogenicity and tumorigenesis of TMZ-resistant GBM cells compared with TMZ-responsive cells. Mechanistically, TMZ-resistant cells exhibited dysregulated expression of miR-3129-5p and miR-199b-3p, resulting in the induced NEDD4-1 mRNA-expression level. The upregulation of NEDD4-1 attenuated PTEN expression and promoted the AKT/NRF2/HO-1 oxidative stress signaling axis, which in turn conferred amplified defense against reactive oxygen species (ROS) and eventually higher resistance against TMZ treatment. The combination treatment of I3C, a known inhibitor of NEDD4-1, with TMZ resulted in a synergistic effect and re-sensitized TMZ-resistant tumor cells both in vitro and in vivo. Conclusions: These findings demonstrate the critical role of NEDD4-1 in regulating the redox imbalance in TMZ-resistant GBM cells via the degradation of PTEN and the upregulation of the AKT/NRF2/HO-1 signaling pathway. Targeting this regulatory axis may help eliminate TMZ-resistant glioblastoma. Full article
(This article belongs to the Special Issue Molecular Signals and Genetic Regulations of Neurological Disorders)
Show Figures

Figure 1

Back to TopTop