Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 548.647
Filter
1.
Transl Psychiatry ; 14(1): 231, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38824172

ABSTRACT

Mapping brain-behaviour associations is paramount to understand and treat psychiatric disorders. Standard approaches involve investigating the association between one brain and one behavioural variable (univariate) or multiple variables against one brain/behaviour feature ('single' multivariate). Recently, large multimodal datasets have propelled a new wave of studies that leverage on 'doubly' multivariate approaches capable of parsing the multifaceted nature of both brain and behaviour simultaneously. Within this movement, canonical correlation analysis (CCA) and partial least squares (PLS) emerge as the most popular techniques. Both seek to capture shared information between brain and behaviour in the form of latent variables. We provide an overview of these methods, review the literature in psychiatric disorders, and discuss the main challenges from a predictive modelling perspective. We identified 39 studies across four diagnostic groups: attention deficit and hyperactive disorder (ADHD, k = 4, N = 569), autism spectrum disorders (ASD, k = 6, N = 1731), major depressive disorder (MDD, k = 5, N = 938), psychosis spectrum disorders (PSD, k = 13, N = 1150) and one transdiagnostic group (TD, k = 11, N = 5731). Most studies (67%) used CCA and focused on the association between either brain morphology, resting-state functional connectivity or fractional anisotropy against symptoms and/or cognition. There were three main findings. First, most diagnoses shared a link between clinical/cognitive symptoms and two brain measures, namely frontal morphology/brain activity and white matter association fibres (tracts between cortical areas in the same hemisphere). Second, typically less investigated behavioural variables in multivariate models such as physical health (e.g., BMI, drug use) and clinical history (e.g., childhood trauma) were identified as important features. Finally, most studies were at risk of bias due to low sample size/feature ratio and/or in-sample testing only. We highlight the importance of carefully mitigating these sources of bias with an exemplar application of CCA.


Subject(s)
Brain , Mental Disorders , Humans , Brain/diagnostic imaging , Brain/physiopathology , Mental Disorders/physiopathology , Autism Spectrum Disorder/physiopathology , Depressive Disorder, Major/physiopathology , Canonical Correlation Analysis , Attention Deficit Disorder with Hyperactivity/physiopathology , Least-Squares Analysis
2.
Sci Rep ; 14(1): 12604, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38824230

ABSTRACT

Pulse wave encephalopathy (PWE) is hypothesised to initiate many forms of dementia, motivating its identification and risk assessment. As candidate pulsatility based biomarkers for PWE, pulsatility index and pulsatility damping have been studied and, currently, do not adequately stratify risk due to variability in pulsatility and spatial bias. Here, we propose a locus-independent pulsatility transmission coefficient computed by spatially tracking pulsatility along vessels to characterise the brain pulse dynamics at a whole-organ level. Our preliminary analyses in a cohort of 20 subjects indicate that this measurement agrees with clinical observations relating blood pulsatility with age, heart rate, and sex, making it a suitable candidate to study the risk of PWE. We identified transmission differences between vascular regions perfused by the basilar and internal carotid arteries attributed to the identified dependence on cerebral blood flow, and some participants presented differences between the internal carotid perfused regions that were not related to flow or pulsatility burden, suggesting underlying mechanical differences. Large populational studies would benefit from retrospective pulsatility transmission analyses, providing a new comprehensive arterial description of the hemodynamic state in the brain. We provide a publicly available implementation of our tools to derive this coefficient, built into pre-existing open-source software.


Subject(s)
Cerebrovascular Circulation , Magnetic Resonance Imaging , Pulsatile Flow , Humans , Female , Male , Cerebrovascular Circulation/physiology , Magnetic Resonance Imaging/methods , Aged , Middle Aged , Brain/diagnostic imaging , Brain/physiology , Brain/blood supply , Pulse Wave Analysis/methods , Carotid Artery, Internal/diagnostic imaging , Carotid Artery, Internal/physiology , Basilar Artery/diagnostic imaging , Basilar Artery/physiology , Adult
3.
Vet Res ; 55(1): 71, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38822398

ABSTRACT

In the wake of the COVID-19 pandemic caused by SARS-CoV-2, questions emerged about the potential effects of Bacillus Calmette-Guérin (BCG) vaccine on the immune response to SARS-CoV-2 infection, including the neurodegenerative diseases it may contribute to. To explore this, an experimental study was carried out in BCG-stimulated and non-stimulated k18-hACE2 mice challenged with SARS-CoV-2. Viral loads in tissues determined by RT-qPCR, histopathology in brain and lungs, immunohistochemical study in brain (IHC) as well as mortality rates, clinical signs and plasma inflammatory and coagulation biomarkers were assessed. Our results showed BCG-SARS-CoV-2 challenged mice presented higher viral loads in the brain and an increased frequency of neuroinvasion, with the greatest differences observed between groups at 3-4 days post-infection (dpi). Histopathological examination showed a higher severity of brain lesions in BCG-SARS-CoV-2 challenged mice, mainly consisting of neuroinflammation, increased glial cell population and neuronal degeneration, from 5 dpi onwards. This group also presented higher interstitial pneumonia and vascular thrombosis in lungs (3-4 dpi), BCG-SARS-CoV-2 mice showed higher values for TNF-α and D-dimer values, while iNOS values were higher in SARS-CoV-2 mice at 3-4 dpi. Results presented in this study indicate that BCG stimulation could have intensified the inflammatory and neurodegenerative lesions promoting virus neuroinvasion and dissemination in this experimental model. Although k18-hACE2 mice show higher hACE2 expression and neurodissemination, this study suggests that, although the benefits of BCG on enhancing heterologous protection against pathogens and tumour cells have been broadly demonstrated, potential adverse outcomes due to the non-specific effects of BCG should be considered.


Subject(s)
BCG Vaccine , Brain , COVID-19 , SARS-CoV-2 , Animals , Mice , BCG Vaccine/administration & dosage , COVID-19/immunology , COVID-19/virology , SARS-CoV-2/physiology , Brain/pathology , Brain/virology , Viral Load , Lung/pathology , Lung/virology , Lung/immunology , Angiotensin-Converting Enzyme 2/metabolism , Mice, Transgenic , Female
4.
Acta Neuropathol Commun ; 12(1): 84, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38822421

ABSTRACT

Alpha-synuclein (αsyn) is an intrinsically disordered protein that aggregates in the brain in several neurodegenerative diseases collectively called synucleinopathies. Phosphorylation of αsyn at serine 129 (PSER129) was considered rare in the healthy human brain but is enriched in pathological αsyn aggregates and is used as a specific marker for disease inclusions. However, recent observations challenge this assumption by demonstrating that PSER129 results from neuronal activity and can be readily detected in the non-diseased mammalian brain. Here, we investigated experimental conditions under which two distinct PSER129 pools, namely endogenous-PSER129 and aggregated-PSER129, could be detected and differentiated in the mammalian brain. Results showed that in the wild-type (WT) mouse brain, perfusion fixation conditions greatly influenced the detection of endogenous-PSER129, with endogenous-PSER129 being nearly undetectable after delayed perfusion fixation (30-min and 1-h postmortem interval). Exposure to anesthetics (e.g., Ketamine or xylazine) before perfusion did not significantly influence endogenous-PSER129 detection or levels. In situ, non-specific phosphatase calf alkaline phosphatase (CIAP) selectively dephosphorylated endogenous-PSER129 while αsyn preformed fibril (PFF)-seeded aggregates and genuine disease aggregates (Lewy pathology and Papp-Lantos bodies in Parkinson's disease and multiple systems atrophy brain, respectively) were resistant to CIAP-mediated dephosphorylation. The phosphatase resistance of aggregates was abolished by sample denaturation, and CIAP-resistant PSER129 was closely associated with proteinase K (PK)-resistant αsyn (i.e., a marker of aggregation). CIAP pretreatment allowed for highly specific detection of seeded αsyn aggregates in a mouse model that accumulates non-aggregated-PSER129. We conclude that αsyn aggregates are impervious to phosphatases, and CIAP pretreatment increases detection specificity for aggregated-PSER129, particularly in well-preserved biological samples (e.g., perfusion fixed or flash-frozen mammalian tissues) where there is a high probability of interference from endogenous-PSER129. Our findings have important implications for the mechanism of PSER129-accumulation in the synucleinopathy brain and provide a simple experimental method to differentiate endogenous-from aggregated PSER129.


Subject(s)
Brain , Mice, Inbred C57BL , alpha-Synuclein , alpha-Synuclein/metabolism , Animals , Brain/metabolism , Brain/pathology , Mice , Phosphorylation , Humans , Protein Aggregates/physiology , Male , Mice, Transgenic , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/pathology , Alkaline Phosphatase/metabolism , Synucleinopathies/metabolism , Synucleinopathies/pathology , Phosphoric Monoester Hydrolases/metabolism
5.
Acta Neuropathol Commun ; 12(1): 83, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38822428

ABSTRACT

Human brain experimental models recapitulating age- and disease-related characteristics are lacking. There is urgent need for human-specific tools that model the complex molecular and cellular interplay between different cell types to assess underlying disease mechanisms and test therapies. Here we present an adapted ex vivo organotypic slice culture method using human post-mortem brain tissue cultured at an air-liquid interface to also study brain white matter. We assessed whether these human post-mortem brain slices recapitulate the in vivo neuropathology and if they are suitable for pathophysiological, experimental and pre-clinical treatment development purposes, specifically regarding leukodystrophies. Human post-mortem brain tissue and cerebrospinal fluid were obtained from control, psychiatric and leukodystrophy donors. Slices were cultured up to six weeks, in culture medium with or without human cerebrospinal fluid. Human post-mortem organotypic brain slice cultures remained viable for at least six weeks ex vivo and maintained tissue structure and diversity of (neural) cell types. Supplementation with cerebrospinal fluid could improve slice recovery. Patient-derived organotypic slice cultures recapitulated and maintained known in vivo neuropathology. The cultures also showed physiologic multicellular responses to lysolecithin-induced demyelination ex vivo, indicating their suitability to study intrinsic repair mechanisms upon injury. The slice cultures were applicable for various experimental studies, as multi-electrode neuronal recordings. Finally, the cultures showed successful cell-type dependent transduction with gene therapy vectors. These human post-mortem organotypic brain slice cultures represent an adapted ex vivo model suitable for multifaceted studies of brain disease mechanisms, boosting translation from human ex vivo to in vivo. This model also allows for assessing potential treatment options, including gene therapy applications. Human post-mortem brain slice cultures are thus a valuable tool in preclinical research to study the pathomechanisms of a wide variety of brain diseases in living human tissue.


Subject(s)
Brain , Organ Culture Techniques , Humans , Brain/pathology , Brain/metabolism , Male , Female , Aged , Middle Aged , Neurons/metabolism , Neurons/pathology , White Matter/pathology , White Matter/metabolism
6.
Am J Psychiatry ; 181(6): 479-481, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38822583
7.
Am J Psychiatry ; 181(6): 482-492, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38822584

ABSTRACT

Schizophrenia is routinely referred to as a neurodevelopmental disorder, but the role of brain development in a disorder typically diagnosed during early adult life is enigmatic. The authors revisit the neurodevelopmental model of schizophrenia with genomic insights from the most recent schizophrenia clinical genetic association studies, transcriptomic and epigenomic analyses from human postmortem brain studies, and analyses from cellular models that recapitulate neurodevelopment. Emerging insights into schizophrenia genetic risk continue to converge on brain development, particularly stages of early brain development, that may be perturbed to deviate from a typical, normative course, resulting in schizophrenia clinical symptomatology. As the authors explicate, schizophrenia genetic risk is likely dynamic and context dependent, with effects of genetic risk varying spatiotemporally, across the neurodevelopmental continuum. Optimizing therapeutic strategies for the heterogeneous collective of individuals with schizophrenia may likely be guided by leveraging markers of genetic risk and derivative functional insights, well before the emergence of psychosis. Ultimately, rather than a focus on therapeutic intervention during adolescence or adulthood, principles of prediction and prophylaxis in the pre- and perinatal and neonatal stages may best comport with the biology of schizophrenia to address the early-stage perturbations that alter the normative neurodevelopmental trajectory.


Subject(s)
Genetic Predisposition to Disease , Schizophrenia , Humans , Schizophrenia/genetics , Schizophrenia/etiology , Genetic Predisposition to Disease/genetics , Brain/pathology , Neurodevelopmental Disorders/genetics , Neurodevelopmental Disorders/etiology
9.
FASEB J ; 38(11): e23648, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38822661

ABSTRACT

Previous studies on germ-free (GF) animals have described altered anxiety-like and social behaviors together with dysregulations in brain serotonin (5-HT) metabolism. Alterations in circulating 5-HT levels and gut 5-HT metabolism have also been reported in GF mice. In this study, we conducted an integrative analysis of various behaviors as well as markers of 5-HT metabolism in the brain and along the GI tract of GF male mice compared with conventional (CV) ones. We found a strong decrease in locomotor activity, accompanied by some signs of increased anxiety-like behavior in GF mice compared with CV mice. Brain gene expression analysis showed no differences in HTR1A and TPH2 genes. In the gut, we found decreased TPH1 expression in the colon of GF mice, while it was increased in the cecum. HTR1A expression was dramatically decreased in the colon, while HTR4 expression was increased both in the cecum and colon of GF mice compared with CV mice. Finally, SLC6A4 expression was increased in the ileum and colon of GF mice compared with CV mice. Our results add to the evidence that the microbiota is involved in regulation of behavior, although heterogeneity among studies suggests a strong impact of genetic and environmental factors on this microbiota-mediated regulation. While no impact of GF status on brain 5-HT was observed, substantial differences in gut 5-HT metabolism were noted, with tissue-dependent results indicating a varying role of microbiota along the GI tract.


Subject(s)
Behavior, Animal , Germ-Free Life , Serotonin , Animals , Serotonin/metabolism , Mice , Male , Gastrointestinal Microbiome/physiology , Brain/metabolism , Tryptophan Hydroxylase/metabolism , Tryptophan Hydroxylase/genetics , Anxiety/metabolism , Anxiety/microbiology , Serotonin Plasma Membrane Transport Proteins/metabolism , Serotonin Plasma Membrane Transport Proteins/genetics , Mice, Inbred C57BL , Receptor, Serotonin, 5-HT1A/metabolism , Receptor, Serotonin, 5-HT1A/genetics , Colon/metabolism , Colon/microbiology
10.
Opt Lett ; 49(11): 3054-3057, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38824326

ABSTRACT

Photoacoustic imaging (PAI) utilizes the photoacoustic effect to record both vascular and functional characteristics of a biological tissue. Photoacoustic signals have typically low amplitude that cannot be read efficiently by data acquisition systems. This necessitates the use of one or more amplifiers. These amplifiers are somewhat bulky (e.g., the ZFL-500LN+, Mini-Circuits, USA, or 351A-3-50-NI, Analog Modules Inc., USA). Here, we describe the fabrication and development process of a transducer with a built-in low-noise preamplifier that is encased within the transducer housing. This new, to the best of our knowledge, design could be advantageous for applications where a compact transducer + preamplifier is required. We demonstrate the performance of this compact detection unit in a laser scanning photoacoustic microscopy system by imaging a rat ear ex vivo and a rat brain vasculature in vivo.


Subject(s)
Equipment Design , Photoacoustic Techniques , Transducers , Photoacoustic Techniques/instrumentation , Photoacoustic Techniques/methods , Animals , Rats , Miniaturization , Brain/diagnostic imaging , Brain/blood supply , Ear/diagnostic imaging , Ear/blood supply , Amplifiers, Electronic
11.
Alzheimers Res Ther ; 16(1): 120, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38824563

ABSTRACT

BACKGROUND: Transcriptome-wide association study (TWAS) is an influential tool for identifying genes associated with complex diseases whose genetic effects are likely mediated through transcriptome. TWAS utilizes reference genetic and transcriptomic data to estimate effect sizes of genetic variants on gene expression (i.e., effect sizes of a broad sense of expression quantitative trait loci, eQTL). These estimated effect sizes are employed as variant weights in gene-based association tests, facilitating the mapping of risk genes with genome-wide association study (GWAS) data. However, most existing TWAS of Alzheimer's disease (AD) dementia are limited to studying only cis-eQTL proximal to the test gene. To overcome this limitation, we applied the Bayesian Genome-wide TWAS (BGW-TWAS) method to leveraging both cis- and trans- eQTL of brain and blood tissues, in order to enhance mapping risk genes for AD dementia. METHODS: We first applied BGW-TWAS to the Genotype-Tissue Expression (GTEx) V8 dataset to estimate cis- and trans- eQTL effect sizes of the prefrontal cortex, cortex, and whole blood tissues. Estimated eQTL effect sizes were integrated with the summary data of the most recent GWAS of AD dementia to obtain BGW-TWAS (i.e., gene-based association test) p-values of AD dementia per gene per tissue type. Then we used the aggregated Cauchy association test to combine TWAS p-values across three tissues to obtain omnibus TWAS p-values per gene. RESULTS: We identified 85 significant genes in prefrontal cortex, 82 in cortex, and 76 in whole blood that were significantly associated with AD dementia. By combining BGW-TWAS p-values across these three tissues, we obtained 141 significant risk genes including 34 genes primarily due to trans-eQTL and 35 mapped risk genes in GWAS Catalog. With these 141 significant risk genes, we detected functional clusters comprised of both known mapped GWAS risk genes of AD in GWAS Catalog and our identified TWAS risk genes by protein-protein interaction network analysis, as well as several enriched phenotypes related to AD. CONCLUSION: We applied BGW-TWAS and aggregated Cauchy test methods to integrate both cis- and trans- eQTL data of brain and blood tissues with GWAS summary data, identifying 141 TWAS risk genes of AD dementia. These identified risk genes provide novel insights into the underlying biological mechanisms of AD dementia and potential gene targets for therapeutics development.


Subject(s)
Alzheimer Disease , Bayes Theorem , Brain , Genetic Predisposition to Disease , Genome-Wide Association Study , Quantitative Trait Loci , Transcriptome , Humans , Alzheimer Disease/genetics , Alzheimer Disease/blood , Genome-Wide Association Study/methods , Brain/metabolism , Genetic Predisposition to Disease/genetics , Quantitative Trait Loci/genetics , Polymorphism, Single Nucleotide , Gene Expression Profiling/methods
12.
J Neuroinflammation ; 21(1): 146, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38824594

ABSTRACT

T cells play an important role in the acquired immune response, with regulatory T cells (Tregs) serving as key players in immune tolerance. Tregs are found in nonlymphoid and damaged tissues and are referred to as "tissue Tregs". They have tissue-specific characteristics and contribute to immunomodulation, homeostasis, and tissue repair through interactions with tissue cells. However, important determinants of Treg tissue specificity, such as antigen specificity, tissue environment, and pathology, remain unclear. In this study, we analyzed Tregs in the central nervous system of mice with ischemic stroke and experimental autoimmune encephalomyelitis (EAE), a mouse model of multiple sclerosis. The gene expression pattern of brain Tregs in the EAE model was more similar to that of ischemic stroke Tregs in the brain than to that of spinal cord Tregs. In addition, most T-cell receptors (TCRs) with high clonality were present in both the brain and spinal cord. Furthermore, Gata3+ and Rorc+ Tregs expressed TCRs recognizing MOG in the spinal cord, suggesting a tissue environment conducive to Rorc expression. Tissue-specific chemokine/chemokine receptor interactions in the spinal cord and brain influenced Treg localization. Finally, spinal cord- or brain-derived Tregs had greater anti-inflammatory capacities in EAE mice, respectively. Taken together, these findings suggest that the tissue environment, rather than pathogenesis or antigen specificity, is the primary determinant of the tissue-specific properties of Tregs. These findings may contribute to the development of novel therapies to suppress inflammation through tissue-specific Treg regulation.


Subject(s)
Brain , Encephalomyelitis, Autoimmune, Experimental , Mice, Inbred C57BL , Spinal Cord , T-Lymphocytes, Regulatory , Animals , T-Lymphocytes, Regulatory/immunology , Mice , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Spinal Cord/immunology , Spinal Cord/pathology , Spinal Cord/metabolism , Brain/immunology , Brain/metabolism , Brain/pathology , Female , Disease Models, Animal
13.
Proc Biol Sci ; 291(2023): 20240138, 2024 May.
Article in English | MEDLINE | ID: mdl-38808448

ABSTRACT

A leading hypothesis for the evolution of large brains in humans and other species is that a feedback loop exists whereby intelligent animals forage more efficiently, which results in increased energy intake that fuels the growth and maintenance of large brains. We test this hypothesis for the first time with high-resolution tracking data from four sympatric, frugivorous rainforest mammal species (42 individuals) and drone-based maps of their predominant feeding trees. We found no evidence that larger-brained primates had more efficient foraging paths than smaller brained procyonids. This refutes a key assumption of the fruit-diet hypothesis for brain evolution, suggesting that other factors such as temporal cognition, extractive foraging or sociality have been more important for brain evolution.


Subject(s)
Brain , Diet , Feeding Behavior , Animals , Brain/physiology , Diet/veterinary , Biological Evolution , Fruit , Rainforest , Primates/physiology
15.
Article in Chinese | MEDLINE | ID: mdl-38811178

ABSTRACT

Objective: To explore the brain white matter damage in patients with moderate to severe obstructive sleep apnea hypopnea syndrome(OSAHS) using diffusional kurtosis imaging(DKI), and to analyze its relationship with anxiety, depression and cognitive impairment in patients. Methods: This was a retrospective case-control study. Fifty confirmed cases (47 males and 3 females) of moderate to severe OSAHS diagnosed by polysomnography(PSG) from November 2017 to December 2022 were selected as OSAHS group(age range from 22 to 65 years old, with median age of 40 years old), and 32 healthy controls(27 males and 5 females) of non-OSAHS diagnosed by PSG were selected as control group(age range from 19 to 56 years old, with median age of 34 years old). DKI scanning, Beck Anxiety Inventory(BAI), Beck Depression Inventory-Ⅱ(BDI-Ⅱ), and Montreal cognitive assessment(MoCA) scores were performed in all subjects. Differences in kurtosis fractional anisotropy(KFA) of various brain regions were compared between the two groups to identify differential brain regions. Correlations were analyzed between KFA reduction and anxiety, depression, and cognitive impairment in OSAHS patients. To study the correlation between brain injury and anxiety, depressive mood, and cognitive dysfunction, statistical methods such as non-parametric tests for two independent samples, chi-square tests, and partial correlation analysis, were used to analyze the evaluation indicators of the two groups. Results: The KFA values in right external capsule, left anterior corona radiata, right anterior corona radiata, left posterior corona radiata, right posterior corona radiata, left superior corona radiata, right superior corona radiata, left superior longitudinal fasciculus, right superior longitudinal fasciculus, genu of corpus callosum, splenium of corpus callosum, body of corpus callosum, posterior cingulate gyrus of moderate to severe OSAHS group were all lower than those in the control group(t=-2.247, -3.028, -3.955, -4.871, -2.632, -2.594, -2.121, -2.167, -3.129, -2.015, -2.317, -2.313, -2.152,P<0.05). For the moderate to severe OSAHS group, the correlation between AHI and KFA values of right posterior corona radiata, right superior corona radiata, left anterior corona radiata, left posterior corona radiata, left superior corona radiata, left superior longitudinal fasciculus, genu of corpus callosum, body of corpus callosum, splenium of corpus callosum were all negative(r=-0.378, -0.307, -0.337, -0.343, -0.341, -0.613, -0.390, -0.384, -0.396, P<0.05). The correlation between LSO2 and KFA values of right anterior corona radiata, right posterior corona radiata, right superior corona radiata, right superior longitudinal fasciculus, left anterior corona radiata, left posterior corona radiata, left superior corona radiata, left superior longitudinal fasciculus, genu of corpus callosum, body of corpus callosum, splenium of corpus callosum, posterior cingulate gyrus were all positive(r=0.330, 0.338, 0.425, 0.312, 0.433, 0.358, 0.410, 0.459, 0.473, 0.659, 0.489, 0.356, P<0.05). The correlation between BAI scores and KFA values of right external capsule, right anterior corona radiata, left posterior corona radiata, left superior corona radiata, body of corpus callosum, splenium of corpus callosum were all negative(r=-0.306, -0.372, -0.296, -0.346, -0.318, -0.386, P<0.05). The correlation between BDI-Ⅱ scores and KFA values of right superior corona radiata, right superior longitudinal fasciculus, left anterior corona radiata, genu of corpus callosum, body of corpus callosum, splenium of corpus callosum were all negative(r=-0.334, -0.289, -0.309, -0.310, -0.503, -0.469, P<0.05). The correlation between MoCA scores and KFA values of right posterior corona radiata, right superior longitudinal fasciculus, left anterior corona radiata, left superior corona radiata, left superior longitudinal fasciculus, genu of corpus callosum, body of corpus callosum, splenium of corpus callosum were all positive(r=0.368, 0.431, 0.324, 0.410, 0.469, 0.384, 0.369, 0.309, P<0.05). Conclusions: With the aggravation of OSAHS, the damage to some brain regions becomes more pronounced in moderate to severe OSAHS patients. These damage brain functional areas are closely related to the anxiety, depression, and cognitive impairment of patients.


Subject(s)
Anxiety , Cognitive Dysfunction , Depression , Sleep Apnea, Obstructive , Humans , Sleep Apnea, Obstructive/diagnostic imaging , Male , Adult , Female , Middle Aged , Case-Control Studies , Retrospective Studies , Cognitive Dysfunction/etiology , Diffusion Tensor Imaging/methods , White Matter/diagnostic imaging , White Matter/pathology , Polysomnography , Aged , Young Adult , Brain/diagnostic imaging , Brain/pathology , Anisotropy
16.
Reprod Toxicol ; 1232024 Jan.
Article in English | MEDLINE | ID: mdl-38706688

ABSTRACT

Exposure to gestational diabetes mellitus (GDM) during pregnancy has significant consequences for the unborn baby and newborn infant. However, whether and how GDM exposure induces the development of neonatal brain hypoxia/ischemia-sensitive phenotype and the underlying molecular mechanisms remain unclear. In this study, we used a late GDM rat model induced by administration of streptozotocin (STZ) on gestational day 12 and investigated its effects of GDM on neonatal brain development. The pregnant rats exhibited increased blood glucose levels in a dose-dependent manner after STZ administration. STZ-induced maternal hyperglycemia led to reduced blood glucose levels in neonatal offspring, resulting in growth restriction and an increased brain to body weight ratio. Importantly, GDM exposure increased susceptibility to hypoxia/ischemia (HI)-induced brain infarct sizes compared to the controls in both male and female neonatal offspring. Further molecular analysis revealed alterations in the PTEN/AKT/mTOR/autophagy signaling pathway in neonatal male offspring brains, along with increased ROS production and autophagy-related proteins (Atg5 and LC3-II). Treatment with the PTEN inhibitor bisperoxovanadate (BPV) eliminated the differences in HI-induced brain infarct sizes between the GDM-exposed and the control groups. These findings provide novel evidence of the development of a brain hypoxia/ischemia-sensitive phenotype in response to GDM exposure and highlight the role of the PTEN/AKT/mTOR/autophagy signaling pathway in this process.


Subject(s)
Autophagy , Brain , Diabetes, Gestational , Hypoxia-Ischemia, Brain , Signal Transduction , Streptozocin , Animals , Female , Male , Pregnancy , Rats , Animals, Newborn , Autophagy/drug effects , Blood Glucose , Brain/metabolism , Brain/drug effects , Brain/pathology , Diabetes, Gestational/chemically induced , Diabetes, Gestational/metabolism , Hypoxia-Ischemia, Brain/metabolism , Prenatal Exposure Delayed Effects , Proto-Oncogene Proteins c-akt/metabolism , PTEN Phosphohydrolase/metabolism , Rats, Sprague-Dawley , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism
17.
Acta Neuropathol ; 147(1): 92, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38801558

ABSTRACT

The SARS-CoV-2 virus that led to COVID-19 is associated with significant and long-lasting neurologic symptoms in many patients, with an increased mortality risk for people with Alzheimer's disease (AD) and/or Down syndrome (DS). However, few studies have evaluated the neuropathological and inflammatory sequelae in postmortem brain tissue obtained from AD and people with DS with severe SARS-CoV-2 infections. We examined tau, beta-amyloid (Aß), inflammatory markers and SARS-CoV-2 nucleoprotein in DS, AD, and healthy non-demented controls with COVID-19 and compared with non-infected brain tissue from each disease group (total n = 24). A nested ANOVA was used to determine regional effects of the COVID-19 infection on arborization of astrocytes (Sholl analysis) and percent-stained area of Iba-1 and TMEM 119. SARS-CoV-2 antibodies labeled neurons and glial cells in the frontal cortex of all subjects with COVID-19, and in the hippocampus of two of the three DS COVID-19 cases. SARS-CoV-2-related alterations were observed in peri-vascular astrocytes and microglial cells in the gray matter of the frontal cortex, hippocampus, and para-hippocampal gyrus. Bright field microscopy revealed scattered intracellular and diffuse extracellular Aß deposits in the hippocampus of controls with confirmed SARS-CoV-2 infections. Overall, the present preliminary findings suggest that SARS-CoV-2 infections induce abnormal inflammatory responses in Down syndrome.


Subject(s)
Alzheimer Disease , Brain , COVID-19 , Down Syndrome , Humans , Down Syndrome/pathology , Down Syndrome/metabolism , Down Syndrome/complications , Alzheimer Disease/pathology , Alzheimer Disease/virology , Alzheimer Disease/metabolism , COVID-19/pathology , COVID-19/complications , Male , Female , Aged , Middle Aged , Brain/pathology , Brain/virology , Aged, 80 and over , Astrocytes/pathology , Astrocytes/virology , Astrocytes/metabolism , Amyloid beta-Peptides/metabolism , SARS-CoV-2/pathogenicity , Microglia/pathology , Microglia/metabolism , Adult , tau Proteins/metabolism
18.
Transl Psychiatry ; 14(1): 211, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38802372

ABSTRACT

Lamotrigine is an effective mood stabiliser, largely used for the management and prevention of depression in bipolar disorder. The neuropsychological mechanisms by which lamotrigine acts to relieve symptoms as well as its neural effects on emotional processing remain unclear. The primary objective of this current study was to investigate the impact of an acute dose of lamotrigine on the neural response to a well-characterised fMRI task probing implicit emotional processing relevant to negative bias. 31 healthy participants were administered either a single dose of lamotrigine (300 mg, n = 14) or placebo (n = 17) in a randomized, double-blind design. Inside the 3 T MRI scanner, participants completed a covert emotional faces gender discrimination task. Brain activations showing significant group differences were identified using voxel-wise general linear model (GLM) nonparametric permutation testing, with threshold free cluster enhancement (TFCE) and a family wise error (FWE)-corrected cluster significance threshold of p < 0.05. Participants receiving lamotrigine were more accurate at identifying the gender of fearful (but not happy or angry) faces. A network of regions associated with emotional processing, including amygdala, insula, and the anterior cingulate cortex (ACC), was significantly less activated in the lamotrigine group compared to the placebo group across emotional facial expressions. A single dose of lamotrigine reduced activation in limbic areas in response to faces with both positive and negative expressions, suggesting a valence-independent effect. However, at a behavioural level lamotrigine appeared to reduce the distracting effect of fear on face discrimination. Such effects may be relevant to the mood stabilisation effects of lamotrigine.


Subject(s)
Emotions , Facial Expression , Healthy Volunteers , Lamotrigine , Magnetic Resonance Imaging , Triazines , Humans , Lamotrigine/pharmacology , Lamotrigine/administration & dosage , Male , Female , Adult , Double-Blind Method , Emotions/drug effects , Triazines/pharmacology , Triazines/administration & dosage , Young Adult , Brain/drug effects , Brain/diagnostic imaging , Facial Recognition/drug effects , Gyrus Cinguli/drug effects , Gyrus Cinguli/diagnostic imaging , Amygdala/drug effects , Amygdala/diagnostic imaging , Antimanic Agents/pharmacology , Antimanic Agents/administration & dosage
19.
Transl Psychiatry ; 14(1): 229, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38816410

ABSTRACT

Depression is a prevalent mental disorder with a complex biological mechanism. Following the rapid development of systems biology technology, a growing number of studies have applied proteomics and metabolomics to explore the molecular profiles of depression. However, a standardized resource facilitating the identification and annotation of the available knowledge from these scattered studies associated with depression is currently lacking. This study presents ProMENDA, an upgraded resource that provides a platform for manual annotation of candidate proteins and metabolites linked to depression. Following the establishment of the protein dataset and the update of the metabolite dataset, the ProMENDA database was developed as a major extension of its initial release. A multi-faceted annotation scheme was employed to provide comprehensive knowledge of the molecules and studies. A new web interface was also developed to improve the user experience. The ProMENDA database now contains 43,366 molecular entries, comprising 20,847 protein entries and 22,519 metabolite entries, which were manually curated from 1370 human, rat, mouse, and non-human primate studies. This represents a significant increase (more than 7-fold) in molecular entries compared to the initial release. To demonstrate the usage of ProMENDA, a case study identifying consistently reported proteins and metabolites in the brains of animal models of depression was presented. Overall, ProMENDA is a comprehensive resource that offers a panoramic view of proteomic and metabolomic knowledge in depression. ProMENDA is freely available at https://menda.cqmu.edu.cn .


Subject(s)
Depression , Metabolomics , Proteomics , Animals , Humans , Rats , Mice , Depression/metabolism , Brain/metabolism , Disease Models, Animal , Databases, Factual
20.
Commun Biol ; 7(1): 668, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38816577

ABSTRACT

Parkinson's disease is managed using levodopa; however, as Parkinson's disease progresses, patients require increased doses of levodopa, which can cause undesirable side effects. Additionally, the oral bioavailability of levodopa decreases in Parkinson's disease patients due to the increased metabolism of levodopa to dopamine by gut bacteria, Enterococcus faecalis, resulting in decreased neuronal uptake and dopamine formation. Parkinson's disease patients have varying levels of these bacteria. Thus, decreasing bacterial metabolism is a promising therapeutic approach to enhance the bioavailability of levodopa in the brain. In this work, we show that Mito-ortho-HNK, formed by modification of a naturally occurring molecule, honokiol, conjugated to a triphenylphosphonium moiety, mitigates the metabolism of levodopa-alone or combined with carbidopa-to dopamine. Mito-ortho-HNK suppresses the growth of E. faecalis, decreases dopamine levels in the gut, and increases dopamine levels in the brain. Mitigating the gut bacterial metabolism of levodopa as shown here could enhance its efficacy.


Subject(s)
Brain , Dopamine , Enterococcus faecalis , Gastrointestinal Microbiome , Levodopa , Parkinson Disease , Levodopa/metabolism , Levodopa/administration & dosage , Gastrointestinal Microbiome/drug effects , Dopamine/metabolism , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Parkinson Disease/microbiology , Brain/metabolism , Brain/drug effects , Animals , Enterococcus faecalis/metabolism , Enterococcus faecalis/drug effects , Male , Antiparkinson Agents/metabolism , Antiparkinson Agents/administration & dosage , Antiparkinson Agents/pharmacology , Carbidopa , Humans , Biphenyl Compounds/metabolism , Mice , Organophosphorus Compounds/metabolism , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL
...