Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 517
Filter
1.
Front Immunol ; 15: 1365871, 2024.
Article in English | MEDLINE | ID: mdl-38756771

ABSTRACT

More than 20% of American adults live with a mental disorder, many of whom are treatment resistant or continue to experience symptoms. Other approaches are needed to improve mental health care, including prevention. The role of the microbiome has emerged as a central tenet in mental and physical health and their interconnectedness (well-being). Under normal conditions, a healthy microbiome promotes homeostasis within the host by maintaining intestinal and brain barrier integrity, thereby facilitating host well-being. Owing to the multidirectional crosstalk between the microbiome and neuro-endocrine-immune systems, dysbiosis within the microbiome is a main driver of immune-mediated systemic and neural inflammation that can promote disease progression and is detrimental to well-being broadly and mental health in particular. In predisposed individuals, immune dysregulation can shift to autoimmunity, especially in the presence of physical or psychological triggers. The chronic stress response involves the immune system, which is intimately involved with the gut microbiome, particularly in the process of immune education. This interconnection forms the microbiota-gut-immune-brain axis and promotes mental health or disorders. In this brief review, we aim to highlight the relationships between stress, mental health, and the gut microbiome, along with the ways in which dysbiosis and a dysregulated immune system can shift to an autoimmune response with concomitant neuropsychological consequences in the context of the microbiota-gut-immune-brain axis. Finally, we aim to review evidenced-based prevention strategies and potential therapeutic targets.


Subject(s)
Brain-Gut Axis , Brain , Dysbiosis , Gastrointestinal Microbiome , Mental Disorders , Mental Health , Stress, Psychological , Humans , Gastrointestinal Microbiome/immunology , Brain-Gut Axis/immunology , Stress, Psychological/immunology , Stress, Psychological/microbiology , Dysbiosis/immunology , Mental Disorders/immunology , Mental Disorders/microbiology , Brain/immunology , Animals , Neuroimmunomodulation
2.
Br J Hosp Med (Lond) ; 84(8): 1-6, 2023 Aug 02.
Article in English | MEDLINE | ID: mdl-37646557

ABSTRACT

The three main theories explaining major mental illness, namely mood disorders, psychoses and dementias, have been partially discredited. Alongside this, there are emerging links between perturbations of the immune system and the onset and phenotypic features of these disorders. This article outlines the alternative pathophysiology and suggests potential treatments which could improve disease burden and avoid the need for psychotropic medication, with their associated side effects and relapse following withdrawal.


Subject(s)
Mental Disorders , Mental Disorders/immunology , Mental Disorders/therapy , Humans , Immune System
4.
Article in English | MEDLINE | ID: mdl-34358623

ABSTRACT

Inflammation is involved in the pathogenesis of psychiatric disorders. Many previous studies have defined the important roles of inflammatory factors in the pathogenesis, diagnosis, and treatment outcomes of psychiatric disorders. Macrophage migration inhibitory factor (MIF), a pro-inflammatory factor, has been gradually recognized to be involved in the development of neurological diseases in recent years. Our current review focuses on discussing the potential beneficial and detrimental roles of MIF in psychiatric disorders. We will provide new mechanistic insights for the development of potential diagnostic and therapeutic biomarkers based on MIF for psychiatric diseases.


Subject(s)
Biomarkers , Inflammation , Macrophage Migration-Inhibitory Factors/immunology , Mental Disorders/immunology , Alzheimer Disease/immunology , Animals , Depression/immunology , Humans , Nervous System Diseases , Schizophrenia/immunology
5.
Behav Brain Res ; 418: 113629, 2022 02 10.
Article in English | MEDLINE | ID: mdl-34656692

ABSTRACT

Mice homozygous for the nude mutation (Foxn1nu) are hairless and exhibit congenital dysgenesis of the thymic epithelium, resulting in a primary immunodeficiency of mature T-cells, and have been used for decades in research with tumour grafts. Early studies have already demonstrated social behaviour impairments and central nervous system (CNS) alterations in these animals, but did not address the complex interplay between CNS, immune system and behavioural alterations. Here we investigate the impact of T-cell immunodeficiency on behaviours relevant to the study of neurodevelopmental and neuropsychiatric disorders. Moreover, we aimed to characterise in a multidisciplinary manner the alterations related to those findings, through evaluation of the excitatory/inhibitory synaptic proteins, cytokines expression and biological spectrum signature of different biomolecules in nude mice CNS. We demonstrate that BALB/c nude mice display sociability impairments, a complex pattern of repetitive behaviours and higher sensitivity to thermal nociception. These animals also have a reduced IFN-γ gene expression in the prefrontal cortex and an absence of T-cells in meningeal tissue, both known modulators of social behaviour. Furthermore, excitatory synaptic protein PSD-95 immunoreactivity was also reduced in the prefrontal cortex, suggesting an intricate involvement of social behaviour related mechanisms. Lastly, employing biospectroscopy analysis, we have demonstrated that BALB/c nude mice have a different CNS spectrochemical signature compared to their heterozygous littermates. Altogether, our results show a comprehensive behavioural analysis of BALB/c nude mice and potential neuroimmunological influences involved with the observed alterations.


Subject(s)
Mental Disorders/immunology , Mutation/genetics , Neurodevelopmental Disorders/immunology , T-Lymphocytes/immunology , Animals , Mice , Mice, Inbred BALB C , Mice, Nude
7.
Nat Commun ; 12(1): 6810, 2021 11 23.
Article in English | MEDLINE | ID: mdl-34815409

ABSTRACT

The prefrontal-hippocampal dysfunction that underlies cognitive deficits in mental disorders emerges during early development. The lateral entorhinal cortex (LEC) is tightly interconnected with both prefrontal cortex (PFC) and hippocampus (HP), yet its contribution to the early dysfunction is fully unknown. Here we show that mice that mimic the dual genetic (G) -environmental (E) etiology (GE mice) of psychiatric risk have poor LEC-dependent recognition memory at pre-juvenile age and abnormal communication within LEC-HP-PFC networks throughout development. These functional and behavioral deficits relate to sparser projections from LEC to CA1 and decreased efficiency of axonal terminals to activate the hippocampal circuits in neonatal GE mice. In contrast, the direct entorhinal drive to PFC is not affected, yet the PFC is indirectly compromised, as target of the under-activated HP. Thus, the entorhinal-hippocampal circuit is already impaired from neonatal age on in GE mice.


Subject(s)
CA1 Region, Hippocampal/physiopathology , Cognitive Dysfunction/physiopathology , Entorhinal Cortex/physiopathology , Mental Disorders/physiopathology , Prefrontal Cortex/physiopathology , Animals , Animals, Newborn , Cognitive Dysfunction/genetics , Cognitive Dysfunction/immunology , Disease Models, Animal , Female , Gene-Environment Interaction , Humans , Male , Mental Disorders/genetics , Mental Disorders/immunology , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Neural Pathways/physiopathology , Optogenetics , Patch-Clamp Techniques , Pregnancy
8.
J Neuroinflammation ; 18(1): 231, 2021 Oct 13.
Article in English | MEDLINE | ID: mdl-34645457

ABSTRACT

It is well accepted that environmental stressors experienced over a one's life, from microbial infections to chemical toxicants to even psychological stressors, ultimately shape central nervous system (CNS) functioning but can also contribute to its eventual breakdown. The severity, timing and type of such environmental "hits", woven together with genetic factors, likely determine what CNS outcomes become apparent. This focused review assesses the current COVID-19 pandemic through the lens of a multi-hit framework and disuses how the SARS-COV-2 virus (causative agent) might impact the brain and potentially interact with other environmental insults. What the long-term consequences of SAR2 COV-2 upon neuronal processes is yet unclear, but emerging evidence is suggesting the possibility of microglial or other inflammatory factors as potentially contributing to neurodegenerative illnesses. Finally, it is critical to consider the impact of the virus in the context of the substantial psychosocial stress that has been associated with the global pandemic. Indeed, the loneliness, fear to the future and loss of social support alone has exerted a massive impact upon individuals, especially the vulnerable very young and the elderly. The substantial upswing in depression, anxiety and eating disorders is evidence of this and in the years to come, this might be matched by a similar spike in dementia, as well as motor and cognitive neurodegenerative diseases.


Subject(s)
COVID-19/immunology , Inflammation Mediators/immunology , Mental Disorders/immunology , Neurodegenerative Diseases/immunology , Neuroimmunomodulation/immunology , Animals , Brain/immunology , COVID-19/epidemiology , Humans , Immunotherapy/trends , Mental Disorders/epidemiology , Mental Disorders/therapy , Neurodegenerative Diseases/epidemiology , Neurodegenerative Diseases/therapy , Stress, Psychological/epidemiology , Stress, Psychological/immunology , Stress, Psychological/therapy
9.
Sci Rep ; 11(1): 21088, 2021 10 26.
Article in English | MEDLINE | ID: mdl-34702870

ABSTRACT

Evolutionary trends may underlie some aspects of the risk for common, non-communicable disorders, including psychiatric disease. We analyzed whole exome sequencing data from 80 unique individuals from India coming from families with two or more individuals with severe mental illness. We used Population Branch Statistics (PBS) to identify variants and genes under positive selection and identified 74 genes as candidates for positive selection. Of these, 20 were previously associated with Schizophrenia, Alzheimer's disease and cognitive abilities in genome wide association studies. We then checked whether any of these 74 genes were involved in common biological pathways or related to specific cellular or molecular functions. We found that immune related pathways and functions related to innate immunity such as antigen binding were over-represented. We also evaluated for the presence of Neanderthal introgressed segments in these genes and found Neanderthal introgression in a single gene out of the 74 candidate genes. However, the introgression pattern indicates the region is unlikely to be the source for selection. Our findings hint at how selection pressures in individuals from families with a history of severe mental illness may diverge from the general population. Further, it also provides insights into the genetic architecture of severe mental illness, such as schizophrenia and its link to immune factors.


Subject(s)
Brain , Exome Sequencing , Immunity, Innate/genetics , Mental Disorders , Animals , Brain/growth & development , Brain/immunology , Female , Humans , Male , Mental Disorders/genetics , Mental Disorders/immunology , Neanderthals/genetics , Neanderthals/immunology
10.
Pharmacol Res ; 173: 105909, 2021 11.
Article in English | MEDLINE | ID: mdl-34543739

ABSTRACT

Recently, increasing evidence has shown gut microbiota dysbiosis might be implicated in the physiological mechanisms of neuropsychiatric disorders. Altered microbial community composition, diversity and distribution traits have been reported in neuropsychiatric disorders. However, the exact pathways by which the intestinal microbiota contribute to neuropsychiatric disorders remain largely unknown. Given that the onset and progression of neuropsychiatric disorders are characterized with complicated alterations of neuroendocrine and immunology, both of which can be continually affected by gut microbiota via "microbiome-gut-brain axis". Thus, we assess the complicated crosstalk between neuroendocrine and immunological regulation might underlie the mechanisms of gut microbiota associated with neuropsychiatric disorders. In this review, we summarized clinical and preclinical evidence on the role of the gut microbiota in neuropsychiatry disorders, especially in mood disorders and neurodevelopmental disorders. This review may elaborate the potential mechanisms of gut microbiota implicating in neuroendocrine-immune regulation and provide a comprehensive understanding of physiological mechanisms for neuropsychiatric disorders.


Subject(s)
Gastrointestinal Microbiome , Mental Disorders/immunology , Mental Disorders/microbiology , Animals , Brain-Gut Axis , Humans
11.
Biochem Pharmacol ; 192: 114711, 2021 10.
Article in English | MEDLINE | ID: mdl-34324871

ABSTRACT

Ample evidence indicates that maternal immune activation (MIA) during gestation is linked to an increased risk for neurodevelopmental and psychiatric disorders, such as autism spectrum disorder (ASD), anxiety and depression, in offspring. However, the underlying mechanism for such a link remains largely elusive. Here, we performed RNA sequencing (RNA-seq) to examine the transcriptional profiles changes in mice in response to MIA and identified that the expression of Scn1a gene, encoding the pore-forming α-subunit of the brain voltage-gated sodium channel type-1 (NaV1.1) primarily in fast-spiking inhibitory interneurons, was significantly decreased in the medial prefrontal cortex (mPFC) of juvenile offspring after MIA. Moreover, diminished excitatory drive onto interneurons causes reduction of spontaneous gamma-aminobutyric acid (GABA)ergic neurotransmission in the mPFC of MIA offspring, leading to hyperactivity in this brain region. Remarkably, treatment with low-dose benzodiazepines clonazepam, an agonist of GABAA receptors, completely prevented the behavioral abnormalities, including stereotypies, social deficits, anxiety- and depression-like behavior, via increasing inhibitory neurotransmission as well as decreasing neural activity in the mPFC of MIA offspring. Our results demonstrate that decreased expression of NaV1.1 in the mPFC leads to abnormalities in maternal inflammation-related behaviors and provides a potential therapeutic strategy for the abnormal behavioral phenotypes observed in the offspring exposed to MIA.


Subject(s)
Clonazepam/therapeutic use , GABA Modulators/therapeutic use , GABAergic Neurons/immunology , Mental Disorders/immunology , Prenatal Exposure Delayed Effects/immunology , Synaptic Transmission/immunology , Animals , Clonazepam/pharmacology , Female , GABA Modulators/pharmacology , GABA-A Receptor Agonists/pharmacology , GABA-A Receptor Agonists/therapeutic use , GABAergic Neurons/chemistry , GABAergic Neurons/drug effects , Male , Mental Disorders/chemically induced , Mental Disorders/prevention & control , Mice , Mice, Inbred C57BL , NAV1.1 Voltage-Gated Sodium Channel/biosynthesis , NAV1.1 Voltage-Gated Sodium Channel/immunology , Poly I-C/toxicity , Prefrontal Cortex/drug effects , Prefrontal Cortex/immunology , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/prevention & control , Receptors, GABA-A/immunology , Synaptic Transmission/drug effects
12.
J Steroid Biochem Mol Biol ; 213: 105952, 2021 10.
Article in English | MEDLINE | ID: mdl-34274458

ABSTRACT

The impacts of glucocorticoids (GCs) are mainly mediated by a nuclear receptor (GR) existing in almost every tissue. The GR regulates a wide range of physiological functions, including inflammation, cell metabolism, and differentiation playing a major role in cellular responses to GCs and stress. Therefore, the dysregulation or disruption of GR can cause deficiencies in the adaptation to stress and the preservation of homeostasis. The number of GR polymorphisms associated with different diseases has been mounting per year. Tackling these clinical complications obliges a comprehensive understanding of the molecular network action of GCs at the level of the GR structure and its signaling pathways. Beyond genetic variation in the GR gene, epigenetic changes can enhance our understanding of causal factors involved in the development of diseases and identifying biomarkers. In this review, we highlight the relationships of GC receptor gene polymorphisms and epigenetics with different diseases.


Subject(s)
Autoimmune Diseases/genetics , Bone Diseases/genetics , Cardiovascular Diseases/genetics , Epigenesis, Genetic , Mental Disorders/genetics , Metabolic Diseases/genetics , Receptors, Glucocorticoid/genetics , Adaptation, Physiological/genetics , Adaptation, Physiological/immunology , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Bone Diseases/immunology , Bone Diseases/pathology , Cardiovascular Diseases/immunology , Cardiovascular Diseases/pathology , DNA Methylation , Glucocorticoids/immunology , Glucocorticoids/metabolism , Homeostasis/genetics , Homeostasis/immunology , Humans , Inflammation , Mental Disorders/immunology , Mental Disorders/pathology , Metabolic Diseases/immunology , Metabolic Diseases/pathology , Polymorphism, Genetic , Receptors, Glucocorticoid/chemistry , Receptors, Glucocorticoid/immunology , Signal Transduction , Stress, Physiological/genetics , Stress, Physiological/immunology
13.
Mol Med Rep ; 24(2)2021 Aug.
Article in English | MEDLINE | ID: mdl-34184073

ABSTRACT

Genetic and nongenetic factors associated with an increased inflammatory response may mediate a link between severe coronavirus disease 2019 (COVID­19) and serious mental illness (SMI). However, systematic assessment of inflammatory response­related factors associated with SMI that could influence COVID­19 outcomes is lacking. In the present review, dietary patterns, smoking and the use of psychotropic medications are discussed as potential extrinsic risk factors and angiotensin­converting enzyme (ACE) insertion/deletion (I/D) gene polymorphisms are considered as potential intrinsic risk factors. A genetics­based prediction model for SMI using ACE­I/D genotyping is also proposed for use in patients experiencing severe COVID­19. Furthermore, the literature suggests that ACE inhibitors may have protective effects against SMI or severe COVID­19, which is often linked to hypertension and other cardiovascular comorbidities. For this reason, we hypothesize that using these medications to treat patients with severe COVID­19 might yield improved outcomes, including in the context of SMI associated with COVID­19.


Subject(s)
COVID-19/immunology , COVID-19/psychology , Mental Disorders/immunology , Mental Disorders/virology , COVID-19/metabolism , Comorbidity , Disease Susceptibility , Humans , Inflammation/immunology , Risk Factors , SARS-CoV-2/isolation & purification
14.
Mol Diagn Ther ; 25(3): 283-299, 2021 05.
Article in English | MEDLINE | ID: mdl-33978935

ABSTRACT

BACKGROUND: Psychiatric disorders have a major impact on the global burden of disease while therapeutic interventions remain insufficient to adequately treat a large number of patients. Regrettably, the efficacy of several psychopharmacological treatment regimens becomes apparent only after 4-6 weeks, and at this point, a significant number of patients present as non-responsive. As such, many patients go weeks/months without appropriate treatment or symptom management. Adequate biomarkers for treatment success and outcome prediction are thus urgently needed. OBJECTIVE: With this systematic review, we provide an overview of the use of peripheral blood mononuclear cells (PBMCs) and their signaling pathways in evaluating and/or predicting the effectiveness of different treatment regimens in the course of psychiatric illnesses. We highlight PBMC characteristics that (i) reflect treatment presence, (ii) allow differentiation of responders from non-responders, and (iii) prove predictive at baseline with regard to treatment outcome for a broad range of psychiatric intervention strategies. REVIEW METHODS: A PubMed database search was performed to extract papers investigating the relation between any type of PBMC characteristic and treatment presence and/or outcome in patients suffering from severe mental illness. Criteria for eligibility were: written in English; psychiatric diagnosis based on DSM-III-R or newer; PBMC isolation via gradient centrifugation; comparison between treated and untreated patients via PBMC features; sample size ≥ n = 5 per experimental group. Papers not researching in vivo treatment effects between patients and healthy controls, non-clinical trials, and non-hypothesis-/data-driven (e.g., -omics designs) approaches were excluded. DATA SYNTHESIS: Twenty-nine original articles were included and qualitatively summarized. Antidepressant and antipsychotic treatments were mostly reflected by intracellular inflammatory markers while intervention with mood stabilizers was evidenced through cell maturation pathways. Lastly, cell viability parameters mirrored predominantly non-pharmacological therapeutic strategies. As for response prediction, PBMC (subtype) counts and telomerase activity seemed most promising for antidepressant treatment outcome determination; full length brain-derived neurotrophic factor (BDNF)/truncated BDNF were shown to be most apt to prognosticate antipsychotic treatment. CONCLUSIONS: We conclude that, although inherent limitations to and heterogeneity in study designs in combination with the scarce number of original studies hamper unambiguous identification, several PBMC characteristics-mostly related to inflammatory pathways and cell viability-indeed show promise towards establishment as clinically relevant treatment biomarkers.


Subject(s)
Antidepressive Agents/therapeutic use , Antipsychotic Agents/therapeutic use , Biomarkers/metabolism , Leukocytes, Mononuclear/metabolism , Mental Disorders/drug therapy , Antidepressive Agents/pharmacology , Antipsychotic Agents/pharmacology , Brain-Derived Neurotrophic Factor/metabolism , Clinical Trials as Topic , Gene Expression Regulation/drug effects , Humans , Mental Disorders/immunology , Mental Disorders/metabolism , Telomerase/metabolism , Treatment Outcome
15.
Acta Neuropsychiatr ; 33(5): 273-276, 2021 Oct.
Article in English | MEDLINE | ID: mdl-33998428

ABSTRACT

Patients with mental illness are at an increased risk of COVID-19 infection, morbidity, and mortality, and prioritisation of this group for COVID-19 vaccination programmes has therefore been suggested. Vaccine uptake may, however, be compromised by vaccine hesitancy amongst patients with mental illness, posing a critical public health issue. We conducted two surveys to provide weighted estimates of vaccine willingness amongst patients with mental illness and the general population of Denmark. Vaccine willingness was high in both groups, but slightly lower amongst patients with mental illness (84.8%), compared with the general population (89.5%) (p < .001). Based on these findings, vaccine hesitancy does not appear to be a major barrier for vaccine uptake amongst patients with mental illness in Denmark, but may be so in other countries with lower general vaccine willingness. Replication of the present study in other countries is strongly warranted.


Subject(s)
COVID-19 Vaccines/therapeutic use , COVID-19/psychology , Mental Disorders/immunology , Patient Acceptance of Health Care/psychology , Adult , Aged , COVID-19/diagnosis , COVID-19/epidemiology , COVID-19/immunology , COVID-19 Vaccines/administration & dosage , COVID-19 Vaccines/immunology , COVID-19 Vaccines/supply & distribution , Case-Control Studies , Denmark/epidemiology , Female , Humans , Male , Mental Disorders/mortality , Mental Disorders/virology , Middle Aged , Patient Acceptance of Health Care/statistics & numerical data , SARS-CoV-2/genetics , SARS-CoV-2/isolation & purification , Surveys and Questionnaires
16.
Front Immunol ; 12: 637087, 2021.
Article in English | MEDLINE | ID: mdl-33815389

ABSTRACT

During its 30 years history, the Hygiene Hypothesis has shown itself to be adaptable whenever it has been challenged by new scientific developments and this is a still a continuously ongoing process. In this regard, the mini review aims to discuss some selected new developments in relation to their impact on further fine-tuning and expansion of the Hygiene Hypothesis. This will include the role of recently discovered classes of innate and adaptive immune cells that challenges the old Th1/Th2 paradigm, the applicability of the Hygiene Hypothesis to newly identified allergy/asthma phenotypes with diverse underlying pathomechanistic endotypes, and the increasing knowledge derived from epigenetic studies that leads to better understanding of mechanisms involved in the translation of environmental impacts on biological systems. Further, we discuss in brief the expansion of the Hygiene Hypothesis to other disease areas like psychiatric disorders and cancer and conclude that the continuously developing Hygiene Hypothesis may provide a more generalized explanation for health burden in highly industrialized countries also relation to global changes.


Subject(s)
Asthma/immunology , Hygiene Hypothesis , Hypersensitivity/immunology , Mental Disorders/immunology , Animals , Developed Countries , Environmental Exposure , Global Burden of Disease , Humans , Th1-Th2 Balance
17.
J Integr Neurosci ; 20(1): 185-196, 2021 Mar 30.
Article in English | MEDLINE | ID: mdl-33834706

ABSTRACT

Mast cells are the major effectors in allergic reactions through degranulation and release of inflammatory, vasoactive and nociceptive mediators associated with the pathogenesis of a variety of inflammatory disorders. Mast cells are strategically positioned as gatekeepers at host/environment interfaces, like the skin, airways, gastrointestinal and urogenital tracts, and their presence also in the brain allows them to act not only as sentinels of invading microorganisms but also as targets to respond to different allergens, pathogens and other dangerous agents that can be ingested, inhaled or encountered after the breakdown of the epithelial barrier. Mast cells can respond to any change in the environment by communicating with the different cells involved in the immune response and giving rise to an amplification signal network through feedback loops. They secrete both preformed mediators within minutes of stimulation and de novo synthesized molecules acting as effectors in the relationship between nervous, vascular and immune systems. For this peculiarity, mast cells are master regulators and key players of the immune system and important sources of essential and beneficial mediators with crucial roles in regulating various physiological processes.


Subject(s)
Brain , Enteric Nervous System , Gastrointestinal Diseases , Gastrointestinal Microbiome , Inflammation , Mast Cells , Mental Disorders , Nervous System Diseases , Animals , Brain/immunology , Brain/metabolism , Enteric Nervous System/immunology , Enteric Nervous System/metabolism , Enteric Nervous System/physiology , Gastrointestinal Diseases/immunology , Gastrointestinal Diseases/metabolism , Gastrointestinal Microbiome/immunology , Humans , Inflammation/immunology , Inflammation/metabolism , Mast Cells/immunology , Mast Cells/metabolism , Mental Disorders/immunology , Mental Disorders/metabolism , Nervous System Diseases/immunology , Nervous System Diseases/metabolism
18.
Biochem Pharmacol ; 188: 114532, 2021 06.
Article in English | MEDLINE | ID: mdl-33773976

ABSTRACT

Understanding the pathophysiological mechanisms of neuropsychiatric disorders has been a challenging quest for neurobiologists. Recent years have witnessed enormous technological advances in the field of neuroimmunology, blurring boundaries between the central nervous system and the periphery. Consequently, the discipline has expanded to cover interactions between the nervous and immune systems in health and diseases. The complex interplay between the peripheral and central immune pathways in neuropsychiatric disorders has recently been documented in various studies, but the genetic determinants remain elusive. Recent transcriptome studies have identified dysregulated genes involved in peripheral immune cell activation, blood-brain barrier integrity, glial cell activation, and synaptic plasticity in major depressive disorder, bipolar disorder, autism spectrum disorder, and schizophrenia. Herein, the key transcriptomic techniques applied in investigating differentially expressed genes and pathways responsible for altered brain-immune interactions in neuropsychiatric disorders are discussed. The application of transcriptomics that can aid in identifying molecular targets in various neuropsychiatric disorders is highlighted.


Subject(s)
Brain/immunology , Mental Disorders/immunology , Molecular Targeted Therapy/methods , Neuroimmunomodulation/physiology , Transcriptome/immunology , Brain/drug effects , Humans , Mental Disorders/genetics , Microglia/drug effects , Microglia/immunology , Neuroimmunomodulation/drug effects , Neuronal Plasticity/drug effects , Neuronal Plasticity/immunology , Transcriptome/drug effects
19.
Transl Psychiatry ; 11(1): 160, 2021 03 15.
Article in English | MEDLINE | ID: mdl-33723208

ABSTRACT

Psychiatric symptoms are seen in some COVID-19 patients, as direct or indirect sequelae, but it is unclear whether SARS-CoV-2 infection interacts with underlying neuronal or psychiatric susceptibilities. Such interactions might arise from COVID-19 immune responses, from infection of neurons themselves or may reflect social-psychological causes. To clarify this we sought the key gene expression pathways altered in COVID-19 also affected in bipolar disorder, post-traumatic stress disorder (PTSD) and schizophrenia, since this may identify pathways of interaction that could be treatment targets. We performed large scale comparisons of whole transcriptome data and immune factor transcript data in peripheral blood mononuclear cells (PBMC) from COVID-19 patients and patients with psychiatric disorders. We also analysed genome-wide association study (GWAS) data for symptomatic COVID-19 patients, comparing GWAS and whole-genome sequence data from patients with bipolar disorder, PTSD and schizophrenia patients. These studies revealed altered signalling and ontology pathways shared by COVID-19 patients and the three psychiatric disorders. Finally, co-expression and network analyses identified gene clusters common to the conditions. COVID-19 patients had peripheral blood immune system profiles that overlapped with those of patients with psychiatric conditions. From the pathways identified, PTSD profiles were the most highly correlated with COVID-19, perhaps consistent with stress-immune system interactions seen in PTSD. We also revealed common inflammatory pathways that may exacerbate psychiatric disorders, which may support the usage of anti-inflammatory medications in these patients. It also highlights the potential clinical application of multi-level dataset studies in difficult-to-treat psychiatric disorders in this COVID-19 pandemic.


Subject(s)
Bipolar Disorder/genetics , COVID-19/genetics , Schizophrenia/genetics , Stress Disorders, Post-Traumatic/genetics , Bipolar Disorder/immunology , COVID-19/immunology , Comorbidity , Gene Expression Profiling , Gene Ontology , Gene Regulatory Networks , Genome-Wide Association Study , Genomics , Humans , Immunity/genetics , Inflammation/genetics , Mental Disorders/genetics , Mental Disorders/immunology , SARS-CoV-2 , Schizophrenia/immunology , Signal Transduction/genetics , Stress Disorders, Post-Traumatic/immunology , Whole Genome Sequencing
20.
J Neuroimmune Pharmacol ; 16(2): 213-218, 2021 06.
Article in English | MEDLINE | ID: mdl-33534108

ABSTRACT

The recent outbreak of coronavirus disease 2019 (COVID-19) has gained considerable attention worldwide due to its increased potential to spread and infect the general population. COVID-19 primarily targets the human respiratory epithelium but also has neuro-invasive potential. Indeed, neuropsychiatric manifestations, such as fatigue, febrile seizures, psychiatric symptoms, and delirium, are consistently observed in COVID-19. The neurobiological basis of neuropsychiatric COVID-19 symptoms is not fully understood. However, previous evidence about systemic viral infections pointed to an ongoing neuroinflammatory response to viral antigens and proinflammatory mediators/immune cells from the periphery. Microglia cells mediate the overproduction of inflammatory cytokines, free radicals, and damage signals, culminating with neurotoxic consequences. Semi-synthetic second-generation tetracyclines, including minocycline (MINO) and doxycycline (DOXY), are safe bacteriostatic agents that have remarkable neuroprotective and anti-inflammatory properties. Promising results have been obtained in clinical trials using tetracyclines for major psychiatric disorders, such as schizophrenia and major depression. Tetracyclines can inhibit microglial reactivity and neuroinflammation by inhibiting nuclear factor kappa B (NF-kB) signaling, cyclooxygenase 2, and matrix metalloproteinases (MMPs). This drug class also has a broad profile of activity against bacteria associated with community-based pneumonia, including atypical agents. COVID-19 patients are susceptible to secondary bacterial infections, especially those on invasive ventilation. Therefore, we suggest tetracyclines' repurposing as a potential treatment for COVID-19 neuropsychiatric manifestations. These drugs can represent a valuable multi-modal treatment for COVID-19-associated neuroinflammatory alterations based on their broad antimicrobial profile and neuroinflammation control.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , COVID-19 Drug Treatment , Drug Repositioning/methods , Mental Disorders/drug therapy , Nervous System Diseases/drug therapy , Tetracyclines/administration & dosage , Antiviral Agents/administration & dosage , COVID-19/epidemiology , COVID-19/immunology , Humans , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/immunology , Mental Disorders/epidemiology , Mental Disorders/immunology , Nervous System Diseases/epidemiology , Nervous System Diseases/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...