ijms-logo

Journal Browser

Journal Browser

Identifying the Molecular Mechanisms of Psychiatric Disorders to Define New Therapeutic Targets

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

Deadline for manuscript submissions: closed (30 November 2022) | Viewed by 51451

Special Issue Editors


E-Mail Website
Guest Editor
Sagol Department of Neurobiology, The Integrated Brain and Behavior Research Center, University of Haifa, Haifa 3498838, Israel
Interests: angelman syndrome; neurodevelopmental disorders; cellular neurophysiology; bioinformatics; transcranial direct current stimulation (tDCS)
Special Issues, Collections and Topics in MDPI journals

E-Mail Website
Guest Editor
Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, The National Institute for Biotechnology in the Negev, Beer-Sheva 84105, Israel
Interests: cognition; plasticity; psychiatry; molecular biology; electrophysiology; Alzheimer's; neurodegenerative diseases
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues,

Psychiatric disorders constitute a broad group of diseases, including anxiety, depression, schizophrenia, bipolar disorder, and autism. Psychiatric disorders have specific behavioral and mental patterns and are characterized by atypical behavior, ideas, perceptions, emotions, and relationships. Currently, therapeutic approaches revolve around imbalance in the function of neurotransmitters such as dopamine, serotonin, GABA, and norepinephrine for most psychiatric disorders. However, although new generations of neurotransmitter-based therapies (including antipsychotics, serotonin reuptake inhibitors, and serotonin-norepinephrine reuptake inhibitors) are available, the efficacy of these medications is limited. In addition, relapse rates in psychiatric disorders are relatively high, indicating the potential involvement of other pathological mechanisms. Indeed, recent genetics and molecular biology studies have shown that the pathogenesis of major psychiatric illnesses involves hundreds of genes and numerous signaling pathways. This Special Issue aims to present new data regarding cellular and molecular mechanisms contributing to such disorders to nominate new candidate targets for future drug development.

Keywords

  • anxiety
  • depression
  • schizophrenia
  • bipolar disorders
  • autism spectrum disorders
  • signaling pathways

Published Papers (15 papers)

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

Research

Jump to: Review

19 pages, 3088 KiB  
Article
Sex-Dependent Effects of Chronic Restraint Stress on Mood-Related Behaviours and Neurochemistry in Mice
by Mauritz Frederick Herselman, Liying Lin, Shayan Luo, Akihiro Yamanaka, Xin-Fu Zhou and Larisa Bobrovskaya
Int. J. Mol. Sci. 2023, 24(12), 10353; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms241210353 - 19 Jun 2023
Cited by 3 | Viewed by 1300
Abstract
Anxiety and depressive disorders are closely associated; however, the pathophysiology of these disorders remains poorly understood. Further exploration of the mechanisms involved in anxiety and depression such as the stress response may provide new knowledge that will contribute to our understanding of these [...] Read more.
Anxiety and depressive disorders are closely associated; however, the pathophysiology of these disorders remains poorly understood. Further exploration of the mechanisms involved in anxiety and depression such as the stress response may provide new knowledge that will contribute to our understanding of these disorders. Fifty-eight 8–12-week-old C57BL6 mice were separated into experimental groups by sex as follows: male controls (n = 14), male restraint stress (n = 14), female controls (n = 15) and female restraint stress (n = 15). These mice were taken through a 4-week randomised chronic restraint stress protocol, and their behaviour, as well as tryptophan metabolism and synaptic proteins, were measured in the prefrontal cortex and hippocampus. Adrenal catecholamine regulation was also measured. The female mice showed greater anxiety-like behaviour than their male counterparts. Tryptophan metabolism was unaffected by stress, but some basal sex characteristics were noted. Synaptic proteins were reduced in the hippocampus in stressed females but increased in the prefrontal cortex of all female mice. These changes were not found in any males. Finally, the stressed female mice showed increased catecholamine biosynthesis capability, but this effect was not found in males. Future studies in animal models should consider these sex differences when evaluating mechanisms related to chronic stress and depression. Full article
Show Figures

Figure 1

19 pages, 10907 KiB  
Article
Transcriptomic Analyses of Brains of RBM8A Conditional Knockout Mice at Different Developmental Stages Reveal Conserved Signaling Pathways Contributing to Neurodevelopmental Diseases
by Colleen McSweeney, Miranda Chen, Fengping Dong, Aswathy Sebastian, Derrick James Reynolds, Jennifer Mott, Zifei Pei, Jizhong Zou, Yongsheng Shi and Yingwei Mao
Int. J. Mol. Sci. 2023, 24(5), 4600; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms24054600 - 27 Feb 2023
Cited by 1 | Viewed by 1484
Abstract
RNA-binding motif 8A (RBM8A) is a core component of the exon junction complex (EJC) that binds pre-mRNAs and regulates their splicing, transport, translation, and nonsense-mediated decay (NMD). Dysfunction in the core proteins has been linked to several detriments in brain development and neuropsychiatric [...] Read more.
RNA-binding motif 8A (RBM8A) is a core component of the exon junction complex (EJC) that binds pre-mRNAs and regulates their splicing, transport, translation, and nonsense-mediated decay (NMD). Dysfunction in the core proteins has been linked to several detriments in brain development and neuropsychiatric diseases. To understand the functional role of Rbm8a in brain development, we have generated brain-specific Rbm8a knockout mice and used next-generation RNA-sequencing to identify differentially expressed genes (DEGs) in mice with heterozygous, conditional knockout (cKO) of Rbm8a in the brain at postnatal day 17 (P17) and at embryonic day 12. Additionally, we analyzed enriched gene clusters and signaling pathways within the DEGs. At the P17 time point, between the control and cKO mice, about 251 significant DEGs were identified. At E12, only 25 DEGs were identified in the hindbrain samples. Bioinformatics analyses have revealed many signaling pathways related to the central nervous system (CNS). When E12 and P17 results were compared, three DEGs, Spp1, Gpnmb, and Top2a, appeared to peak at different developmental time points in the Rbm8a cKO mice. Enrichment analyses suggested altered activity in pathways affecting cellular proliferation, differentiation, and survival. The results support the hypothesis that loss of Rbm8a causes decreased cellular proliferation, increased apoptosis, and early differentiation of neuronal subtypes, which may lead ultimately to an altered neuronal subtype composition in the brain. Full article
Show Figures

Figure 1

20 pages, 1799 KiB  
Article
MiRNA Differences Related to Treatment-Resistant Schizophrenia
by Daniel Pérez-Rodríguez, Maria Aránzazu Penedo, Tania Rivera-Baltanás, Tonatiuh Peña-Centeno, Susanne Burkhardt, Andre Fischer, José M. Prieto-González, José Manuel Olivares, Hugo López-Fernández and Roberto Carlos Agís-Balboa
Int. J. Mol. Sci. 2023, 24(3), 1891; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms24031891 - 18 Jan 2023
Cited by 3 | Viewed by 2631
Abstract
Schizophrenia (SZ) is a serious mental disorder that is typically treated with antipsychotic medication. Treatment-resistant schizophrenia (TRS) is the condition where symptoms remain after pharmacological intervention, resulting in long-lasting functional and social impairments. As the identification and treatment of a TRS patient requires [...] Read more.
Schizophrenia (SZ) is a serious mental disorder that is typically treated with antipsychotic medication. Treatment-resistant schizophrenia (TRS) is the condition where symptoms remain after pharmacological intervention, resulting in long-lasting functional and social impairments. As the identification and treatment of a TRS patient requires previous failed treatments, early mechanisms of detection are needed in order to quicken the access to effective therapy, as well as improve treatment adherence. In this study, we aim to find a microRNA (miRNA) signature for TRS, as well as to shed some light on the molecular pathways potentially involved in this severe condition. To do this, we compared the blood miRNAs of schizophrenia patients that respond to medication and TRS patients, thus obtaining a 16-miRNA TRS profile. Then, we assessed the ability of this signature to separate responders and TRS patients using hierarchical clustering, observing that most of them are grouped correctly (~70% accuracy). We also conducted a network, pathway analysis, and bibliography search to spot molecular pathways potentially altered in TRS. We found that the response to stress seems to be a key factor in TRS and that proteins p53, SIRT1, MDM2, and TRIM28 could be the potential mediators of such responses. Finally, we suggest a molecular pathway potentially regulated by the miRNAs of the TRS profile. Full article
Show Figures

Figure 1

22 pages, 5593 KiB  
Article
The Effects of Prenatal Dexamethasone Exposure on Brain Metabolic Homeostasis in Adulthood: Implications for Depression
by Katarzyna Głombik, Magdalena Kukla-Bartoszek, Katarzyna Curzytek, Jan Detka, Agnieszka Basta-Kaim and Bogusława Budziszewska
Int. J. Mol. Sci. 2023, 24(2), 1156; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms24021156 - 06 Jan 2023
Cited by 3 | Viewed by 1538
Abstract
Since depression produces a long-term negative impact on quality of life, understanding the pathophysiological changes implicated in this disorder is urgent. There is growing evidence that demonstrates a key role for dysfunctional energy metabolism in driving the onset of depression; thus, bioenergetic alterations [...] Read more.
Since depression produces a long-term negative impact on quality of life, understanding the pathophysiological changes implicated in this disorder is urgent. There is growing evidence that demonstrates a key role for dysfunctional energy metabolism in driving the onset of depression; thus, bioenergetic alterations should be extensively studied. Brain metabolism is known to be a glucocorticoid-sensitive process, but the long-lasting consequences in adulthood following high levels of glucocorticoids at the early stages of life are unclear. We examined a possible association between brain energetic changes induced by synthetic glucocorticoid-dexamethasone treatment in the prenatal period and depressive-like behavior. The results show a reduction in the oxidative phosphorylation process, Krebs cycle impairment, and a weakening of the connection between the Krebs cycle and glycolysis in the frontal cortex of animals receiving dexamethasone, which leads to ATP reduction. These changes appear to be mainly due to decreased expression of pyruvate dehydrogenase, impairment of lactate transport to neurons, and pyruvate to the mitochondria. Acute stress in adulthood only slightly modified the observed alterations in the frontal cortex, while in the case of the hippocampus, prenatal exposure to dexamethasone made this structure more sensitive to future adverse factors. Full article
Show Figures

Figure 1

16 pages, 2804 KiB  
Article
Nna1, Essential for Purkinje Cell Survival, Is also Associated with Emotion and Memory
by Li Zhou, Kohtarou Konno, Maya Yamazaki, Manabu Abe, Rie Natsume, Masahiko Watanabe, Hirohide Takebayashi and Kenji Sakimura
Int. J. Mol. Sci. 2022, 23(21), 12961; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms232112961 - 26 Oct 2022
Cited by 2 | Viewed by 1600
Abstract
Nna1/CCP1 is generally known as a causative gene for a spontaneous autosomal recessive mouse mutation, Purkinje cell degeneration (pcd). There is enough evidence that the cytosolic function of the zinc carboxypeptidase (CP) domain at the C-terminus of the Nna1 protein is [...] Read more.
Nna1/CCP1 is generally known as a causative gene for a spontaneous autosomal recessive mouse mutation, Purkinje cell degeneration (pcd). There is enough evidence that the cytosolic function of the zinc carboxypeptidase (CP) domain at the C-terminus of the Nna1 protein is associated with cell death. On the other hand, this molecule’s two nuclear localization signals (NLSs) suggest some other functions exist. We generated exon 3-deficient mice (Nna1N KO), which encode a portion of the N-terminal NLS. Despite the frameshift occurring in these mice, there was an expression of the Nna1 protein lacking the N-terminal side. Surprisingly, the pcd phenotype did not occur in the Nna1N KO mouse. Behavioral analysis revealed that they were less anxious when assessed by the elevated plus maze and the light/dark box tests compared to the control. Furthermore, they showed impairments in context-dependent and sound stimulus-dependent learning. Biochemical analysis of Nna1N KO mice revealed a reduced level of the AMPA-type glutamine receptor GluA2 in the hippocampal synaptosomal fraction. In addition, the motor protein kinesin-1, which transports GluA2 to dendrites, was also decreased. These results indicate that Nna1 is also involved in emotion and memory learning, presumably through the trafficking and expression of synaptic signaling molecules, besides a known role in cell survival. Full article
Show Figures

Figure 1

14 pages, 988 KiB  
Article
Hyperbaric Oxygen Therapy Alleviates Social Behavior Dysfunction and Neuroinflammation in a Mouse Model for Autism Spectrum Disorders
by Inbar Fischer, Sophie Shohat, Gilad Levy, Ela Bar, Sari Schokoroy Trangle, Shai Efrati and Boaz Barak
Int. J. Mol. Sci. 2022, 23(19), 11077; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms231911077 - 21 Sep 2022
Cited by 7 | Viewed by 9689
Abstract
Autism spectrum disorder (ASD) is a multifactorial neurodevelopmental disorder (NDD) characterized by impaired social communication and repetitive behavior, among other symptoms. ASD is highly heritable, with SHANK3 being one of the high-risk genes for ASD. In recent years, knowledge has been growing regarding [...] Read more.
Autism spectrum disorder (ASD) is a multifactorial neurodevelopmental disorder (NDD) characterized by impaired social communication and repetitive behavior, among other symptoms. ASD is highly heritable, with SHANK3 being one of the high-risk genes for ASD. In recent years, knowledge has been growing regarding the neuroplasticity effect induced by hyperbaric oxygen therapy (HBOT) and its potential use for ASD. Here, we characterized the effect of HBOT on a mouse model for ASD with the human genetic condition of InsG3680 mutation in the Shank3 gene. As compared to placebo, HBOT improved social behavior and reduced neuroinflammation in the cortex of the InsG3680(+/+) mice. Specifically, HBOT induced upregulation of Insulin-like growth factor 1 (Igf1) expression levels and reduced the number of Iba1-positive cells in the mouse model for ASD compared to placebo control. Together, our research suggests that HBOT has the potential to improve the clinical outcome of ASD by ameliorating some of the core pathophysiological processes responsible for the development of the disorder. Full article
Show Figures

Figure 1

18 pages, 2307 KiB  
Article
An Association Study of DNA Methylation and Gene Expression in Angelman Syndrome: A Bioinformatics Approach
by Julia Panov and Hanoch Kaphzan
Int. J. Mol. Sci. 2022, 23(16), 9139; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms23169139 - 15 Aug 2022
Viewed by 1950
Abstract
Angelman syndrome (AS) is a neurodevelopmental disorder caused by the loss of function of the E3-ligase UBE3A. Despite multiple studies, AS pathophysiology is still obscure and has mostly been explored in rodent models of the disease. In recent years, a growing body of [...] Read more.
Angelman syndrome (AS) is a neurodevelopmental disorder caused by the loss of function of the E3-ligase UBE3A. Despite multiple studies, AS pathophysiology is still obscure and has mostly been explored in rodent models of the disease. In recent years, a growing body of studies has utilized omics datasets in the attempt to focus research regarding the pathophysiology of AS. Here, for the first time, we utilized a multi-omics approach at the epigenomic level and the transcriptome level, for human-derived neurons. Using publicly available datasets for DNA methylation and gene expression, we found genome regions in proximity to gene promoters and intersecting with gene-body regions that were differentially methylated and differentially expressed in AS. We found that overall, the genome in AS postmortem brain tissue was hypo-methylated compared to healthy controls. We also found more upregulated genes than downregulated genes in AS. Many of these dysregulated genes in neurons obtained from AS patients are known to be critical for neuronal development and synaptic functioning. Taken together, our results suggest a list of dysregulated genes that may be involved in AS development and its pathological features. Moreover, these genes might also have a role in neurodevelopmental disorders similar to AS. Full article
Show Figures

Figure 1

17 pages, 1672 KiB  
Article
Social Fear Affects Limbic System Neuronal Activity and Gene Expression
by Catharina S. Hamann, Julian Bankmann, Hanna Mora Maza, Johannes Kornhuber, Iulia Zoicas and Angelika Schmitt-Böhrer
Int. J. Mol. Sci. 2022, 23(15), 8228; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms23158228 - 26 Jul 2022
Cited by 3 | Viewed by 2315
Abstract
Social anxiety disorder (SAD) is a highly prevalent and comorbid anxiety disorder with rather unclear underlying mechanisms. Here, we aimed to characterize neurobiological changes occurring in mice expressing symptoms of social fear and to identify possible therapeutic targets for SAD. Social fear was [...] Read more.
Social anxiety disorder (SAD) is a highly prevalent and comorbid anxiety disorder with rather unclear underlying mechanisms. Here, we aimed to characterize neurobiological changes occurring in mice expressing symptoms of social fear and to identify possible therapeutic targets for SAD. Social fear was induced via social fear conditioning (SFC), a validated animal model of SAD. We assessed the expression levels of the immediate early genes (IEGs) cFos, Fosl2 and Arc as markers of neuronal activity and the expression levels of several genes of the GABAergic, serotoninergic, oxytocinergic, vasopressinergic and neuropeptide Y (NPY)-ergic systems in brain regions involved in social behavior or fear-related behavior in SFC+ and SFC− mice 2 h after exposure to a conspecific. SFC+ mice showed a decreased number and density of cFos-positive cells and decreased expression levels of IEGs in the dorsal hippocampus. SFC+ mice also showed alterations in the expression of NPY and serotonin system-related genes in the paraventricular nucleus of the hypothalamus, basolateral amygdala, septum and dorsal raphe nucleus, but not in the dorsal hippocampus. Our results describe neuronal alterations occurring during the expression of social fear and identify the NPY and serotonergic systems as possible targets in the treatment of SAD. Full article
Show Figures

Figure 1

24 pages, 3748 KiB  
Article
Heat-Stress Preconditioning Attenuates Behavioral Responses to Psychological Stress: The Role of HSP-70 in Modulating Stress Responses
by Tal Belity, Michal Horowitz, Jay R. Hoffman, Yoram Epstein, Yaron Bruchim, Doron Todder and Hagit Cohen
Int. J. Mol. Sci. 2022, 23(8), 4129; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms23084129 - 08 Apr 2022
Cited by 4 | Viewed by 1972
Abstract
Exposure to high ambient temperature is a stressor that influences both biological and behavioral functions and has been previously shown to have an extensive impact on brain structure and function. Physiological, cellular and behavioral responses to heat-stress (HS) (40–41 °C, 2 h) were [...] Read more.
Exposure to high ambient temperature is a stressor that influences both biological and behavioral functions and has been previously shown to have an extensive impact on brain structure and function. Physiological, cellular and behavioral responses to heat-stress (HS) (40–41 °C, 2 h) were evaluated in adult male Sprague-Dawley rats. The effect of HS exposure before predator-scent stress (PSS) exposure (i.e., HS preconditioning) was examined. Finally, a possible mechanism of HS-preconditioning to PSS was investigated. Immunohistochemical analyses of chosen cellular markers were performed in the hippocampus and in the hypothalamic paraventricular nucleus (PVN). Plasma corticosterone levels were evaluated, and the behavioral assessment included the elevated plus-maze (EPM) and the acoustic startle response (ASR) paradigms. Endogenous levels of heat shock protein (HSP)-70 were manipulated using an amino acid (L-glutamine) and a pharmacological agent (Doxazosin). A single exposure to an acute HS resulted in decreased body mass (BM), increased body temperature and increased corticosterone levels. Additionally, extensive cellular, but not behavioral changes were noted. HS-preconditioning provided behavioral resiliency to anxiety-like behavior associated with PSS, possibly through the induction of HSP-70. Targeting of HSP-70 is an attractive strategy for stress-related psychopathology treatment. Full article
Show Figures

Figure 1

Review

Jump to: Research

13 pages, 969 KiB  
Review
White Matter Microstructure Associated with the Antidepressant Effects of Deep Brain Stimulation in Treatment-Resistant Depression: A Review of Diffusion Tensor Imaging Studies
by Giulia Cattarinussi, Hossein Sanjari Moghaddam, Mohammad Hadi Aarabi, Letizia Squarcina, Fabio Sambataro, Paolo Brambilla and Giuseppe Delvecchio
Int. J. Mol. Sci. 2022, 23(23), 15379; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms232315379 - 06 Dec 2022
Cited by 3 | Viewed by 1533
Abstract
Treatment-resistant depression (TRD) is a severe disorder characterized by high relapse rates and decreased quality of life. An effective strategy in the management of TRD is deep brain stimulation (DBS), a technique consisting of the implantation of electrodes that receive a stimulation via [...] Read more.
Treatment-resistant depression (TRD) is a severe disorder characterized by high relapse rates and decreased quality of life. An effective strategy in the management of TRD is deep brain stimulation (DBS), a technique consisting of the implantation of electrodes that receive a stimulation via a pacemaker-like stimulator into specific brain areas, detected through neuroimaging investigations, which include the subgenual cingulate cortex (sgCC), basal ganglia, and forebrain bundles. In this context, to improve our understanding of the mechanism underlying the antidepressant effects of DBS in TRD, we collected the results of diffusion tensor imaging (DTI) studies exploring how WM microstructure is associated with the therapeutic effects of DBS in TRD. A search on PubMed, Web of Science, and Scopus identified 11 investigations assessing WM microstructure in responders and non-responders to DBS. Altered WM microstructure, particularly in the sgCC, medial forebrain bundle, cingulum bundle, forceps minor, and uncinate fasciculus, was associated with the antidepressant effect of DBS in TRD. Overall, the results show that DBS targeting selective brain regions, including the sgCC, forebrain bundle, cingulum bundle, rectus gyrus, anterior limb of the internal capsule, forceps minor, and uncinate fasciculus, seem to be effective for the treatment of TRD. Full article
Show Figures

Figure 1

29 pages, 2359 KiB  
Review
Therapeutic Implications of microRNAs in Depressive Disorders: A Review
by Mubashir Hassan, Aqsa Amir, Saba Shahzadi and Andrzej Kloczkowski
Int. J. Mol. Sci. 2022, 23(21), 13530; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms232113530 - 04 Nov 2022
Cited by 4 | Viewed by 2312
Abstract
MicroRNAs are hidden players in complex psychophysical phenomena such as depression and anxiety related disorders though the activation and deactivation of multiple proteins in signaling cascades. Depression is classified as a mood disorder and described as feelings of sadness, loss, or anger that [...] Read more.
MicroRNAs are hidden players in complex psychophysical phenomena such as depression and anxiety related disorders though the activation and deactivation of multiple proteins in signaling cascades. Depression is classified as a mood disorder and described as feelings of sadness, loss, or anger that interfere with a person’s everyday activities. In this review, we have focused on exploration of the significant role of miRNAs in depression by affecting associated target proteins (cellular and synaptic) and their signaling pathways which can be controlled by the attachment of miRNAs at transcriptional and translational levels. Moreover, miRNAs have potential role as biomarkers and may help to cure depression through involvement and interactions with multiple pharmacological and physiological therapies. Taken together, miRNAs might be considered as promising novel therapy targets themselves and may interfere with currently available antidepressant treatments. Full article
Show Figures

Figure 1

18 pages, 2029 KiB  
Review
Glymphatic System Dysfunction and Sleep Disturbance May Contribute to the Pathogenesis and Progression of Parkinson’s Disease
by Andie Massey, Matthew K. Boag, Annie Magnier, Dharah P. C. F. Bispo, Tien K. Khoo and Dean L. Pountney
Int. J. Mol. Sci. 2022, 23(21), 12928; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms232112928 - 26 Oct 2022
Cited by 27 | Viewed by 5372
Abstract
Parkinson’s disease (PD) is a multisystem alpha-synucleinopathic neurodegenerative disease and the most prevalent neurodegenerative disorder after Alzheimer’s disease with a high incidence rate in the elderly population. PD is highly multifactorial in etiology and has complex and wide-ranging pathogenic mechanisms. Environmental exposures and [...] Read more.
Parkinson’s disease (PD) is a multisystem alpha-synucleinopathic neurodegenerative disease and the most prevalent neurodegenerative disorder after Alzheimer’s disease with a high incidence rate in the elderly population. PD is highly multifactorial in etiology and has complex and wide-ranging pathogenic mechanisms. Environmental exposures and genetic predisposition are prominent risk factors. However, current evidence suggests that an intimate link may exist between the risk factor of sleep disturbance and PD pathogenesis. PD is characterized by the pathological hallmarks of alpha-synuclein aggregations and dopaminergic neuron degeneration in the substantia nigra. The loss of dopamine-producing neurons results in both motor and non-motor symptoms, most commonly, bradykinesia, tremor, rigidity, psychiatric disorders, sleep disorders and gastrointestinal problems. Factors that may exacerbate alpha-synuclein accumulation and dopamine neuron loss include neuroinflammation and glymphatic system impairment. Extracellular alpha-synuclein can induce an inflammatory response which can lead to neural cell death and inhibition of neurogenesis. The glymphatic system functions most optimally to remove extracellular brain solutes during sleep and therefore sleep disruption may be a crucial progression factor as well as a risk factor. This literature review interprets and analyses data from experimental and epidemiological studies to determine the recent advances in establishing a relationship between glymphatic system dysfunction, sleep disturbance, and PD pathogenesis and progression. This review addresses current limitations surrounding the ability to affirm a causal link between improved glymphatic clearance by increased sleep quality in PD prevention and management. Furthermore, this review proposes potential therapeutic approaches that could utilize the protective mechanism of sleep, to promote glymphatic clearance that therefore may reduce disease progression as well as symptom severity in PD patients. Full article
Show Figures

Figure 1

20 pages, 1894 KiB  
Review
New Insights into the Pivotal Role of the Amygdala in Inflammation-Related Depression and Anxiety Disorder
by Ping Hu, Ying Lu, Bing-Xing Pan and Wen-Hua Zhang
Int. J. Mol. Sci. 2022, 23(19), 11076; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms231911076 - 21 Sep 2022
Cited by 27 | Viewed by 7204
Abstract
Depression and anxiety disorders are the two most prevalent psychiatric diseases that affect hundreds of millions of individuals worldwide. Understanding the etiology and related mechanisms is of great importance and might yield new therapeutic strategies to treat these diseases effectively. During the past [...] Read more.
Depression and anxiety disorders are the two most prevalent psychiatric diseases that affect hundreds of millions of individuals worldwide. Understanding the etiology and related mechanisms is of great importance and might yield new therapeutic strategies to treat these diseases effectively. During the past decades, a growing number of studies have pointed out the importance of the stress-induced inflammatory response in the amygdala, a kernel region for processing emotional stimuli, as a potentially critical contributor to the pathophysiology of depression and anxiety disorders. In this review, we first summarized the recent progress from both animal and human studies toward understanding the causal link between stress-induced inflammation and depression and anxiety disorders, with particular emphasis on findings showing the effect of inflammation on the functional changes in neurons in the amygdala, at levels ranging from molecular signaling, cellular function, synaptic plasticity, and the neural circuit to behavior, as well as their contributions to the pathology of inflammation-related depression and anxiety disorders. Finally, we concluded by discussing some of the difficulties surrounding the current research and propose some issues worth future study in this field. Full article
Show Figures

Figure 1

14 pages, 909 KiB  
Review
Diverse Roles of the Exon Junction Complex Factors in the Cell Cycle, Cancer, and Neurodevelopmental Disorders-Potential for Therapeutic Targeting
by Hannah Martin, Julian Rupkey, Shravan Asthana, Joy Yoon, Shray Patel, Jennifer Mott, Zifei Pei and Yingwei Mao
Int. J. Mol. Sci. 2022, 23(18), 10375; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms231810375 - 08 Sep 2022
Cited by 2 | Viewed by 2105
Abstract
The exon junction complex (EJC) plays a crucial role in regulating gene expression at the levels of alternative splicing, translation, mRNA localization, and nonsense-mediated decay (NMD). The EJC is comprised of three core proteins: RNA-binding motif 8A (RBM8A), Mago homolog (MAGOH), eukaryotic initiation [...] Read more.
The exon junction complex (EJC) plays a crucial role in regulating gene expression at the levels of alternative splicing, translation, mRNA localization, and nonsense-mediated decay (NMD). The EJC is comprised of three core proteins: RNA-binding motif 8A (RBM8A), Mago homolog (MAGOH), eukaryotic initiation factor 4A3 (eIF4A3), and a peripheral EJC factor, metastatic lymph node 51 (MLN51), in addition to other peripheral factors whose structural integration is activity-dependent. The physiological and mechanistic roles of the EJC in contribution to molecular, cellular, and organismal level function continue to be explored for potential insights into genetic or pathological dysfunction. The EJC’s specific role in the cell cycle and its implications in cancer and neurodevelopmental disorders prompt enhanced investigation of the EJC as a potential target for these diseases. In this review, we highlight the current understanding of the EJC’s position in the cell cycle, its relation to cancer and developmental diseases, and potential avenues for therapeutic targeting. Full article
Show Figures

Figure 1

18 pages, 765 KiB  
Review
Beyond NMDA Receptors: Homeostasis at the Glutamate Tripartite Synapse and Its Contributions to Cognitive Dysfunction in Schizophrenia
by Hagar Bauminger and Inna Gaisler-Salomon
Int. J. Mol. Sci. 2022, 23(15), 8617; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms23158617 - 03 Aug 2022
Cited by 4 | Viewed by 4683
Abstract
Cognitive deficits are core symptoms of schizophrenia but remain poorly addressed by dopamine-based antipsychotic medications. Glutamate abnormalities are implicated in schizophrenia-related cognitive deficits. While the role of the NMDA receptor has been extensively studied, less attention was given to other components that control [...] Read more.
Cognitive deficits are core symptoms of schizophrenia but remain poorly addressed by dopamine-based antipsychotic medications. Glutamate abnormalities are implicated in schizophrenia-related cognitive deficits. While the role of the NMDA receptor has been extensively studied, less attention was given to other components that control glutamate homeostasis. Glutamate dynamics at the tripartite synapse include presynaptic and postsynaptic components and are tightly regulated by neuron–astrocyte crosstalk. Here, we delineate the role of glutamate homeostasis at the tripartite synapse in schizophrenia-related cognitive dysfunction. We focus on cognitive domains that can be readily measured in humans and rodents, i.e., working memory, recognition memory, cognitive flexibility, and response inhibition. We describe tasks used to measure cognitive function in these domains in humans and rodents, and the relevance of glutamate alterations in these domains. Next, we delve into glutamate tripartite synaptic components and summarize findings that implicate the relevance of these components to specific cognitive domains. These collective findings indicate that neuron–astrocyte crosstalk at the tripartite synapse is essential for cognition, and that pre- and postsynaptic components play a critical role in maintaining glutamate homeostasis and cognitive well-being. The contribution of these components to cognitive function should be considered in order to better understand the role played by glutamate signaling in cognition and develop efficient pharmacological treatment avenues for schizophrenia treatment-resistant symptoms. Full article
Show Figures

Figure 1

Back to TopTop