Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Brain ; 147(9): 3032-3047, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-38940350

ABSTRACT

In frontotemporal lobar degeneration (FTLD), pathological protein aggregation in specific brain regions is associated with declines in human-specialized social-emotional and language functions. In most patients, disease protein aggregates contain either TDP-43 (FTLD-TDP) or tau (FTLD-tau). Here, we explored whether FTLD-associated regional degeneration patterns relate to regional gene expression of human accelerated regions (HARs), conserved sequences that have undergone positive selection during recent human evolution. To this end, we used structural neuroimaging from patients with FTLD and human brain regional transcriptomic data from controls to identify genes expressed in FTLD-targeted brain regions. We then integrated primate comparative genomic data to test our hypothesis that FTLD targets brain regions linked to expression levels of recently evolved genes. In addition, we asked whether genes whose expression correlates with FTLD atrophy are enriched for genes that undergo cryptic splicing when TDP-43 function is impaired. We found that FTLD-TDP and FTLD-tau subtypes target brain regions with overlapping and distinct gene expression correlates, highlighting many genes linked to neuromodulatory functions. FTLD atrophy-correlated genes were strongly enriched for HARs. Atrophy-correlated genes in FTLD-TDP showed greater overlap with TDP-43 cryptic splicing genes and genes with more numerous TDP-43 binding sites compared with atrophy-correlated genes in FTLD-tau. Cryptic splicing genes were enriched for HAR genes, and vice versa, but this effect was due to the confounding influence of gene length. Analyses performed at the individual-patient level revealed that the expression of HAR genes and cryptically spliced genes within putative regions of disease onset differed across FTLD-TDP subtypes. Overall, our findings suggest that FTLD targets brain regions that have undergone recent evolutionary specialization and provide intriguing potential leads regarding the transcriptomic basis for selective vulnerability in distinct FTLD molecular-anatomical subtypes.


Subject(s)
Brain , Frontotemporal Lobar Degeneration , Humans , Frontotemporal Lobar Degeneration/genetics , Frontotemporal Lobar Degeneration/metabolism , Brain/metabolism , Brain/pathology , Male , Female , Aged , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Middle Aged , tau Proteins/genetics , tau Proteins/metabolism , Atrophy/genetics , Animals , Evolution, Molecular , Gene Expression/genetics
2.
Hum Brain Mapp ; 44(18): 6364-6374, 2023 Dec 15.
Article in English | MEDLINE | ID: mdl-37846762

ABSTRACT

Alzheimer's disease (AD) is one of the most prevalent forms of dementia in older individuals. Convergent evidence suggests structural connectome abnormalities in specific brain regions are linked to AD progression. The biological basis underpinnings of these connectome changes, however, have remained elusive. We utilized an individual regional mean connectivity strength (RMCS) derived from a regional radiomics similarity network to capture altered morphological connectivity in 1654 participants (605 normal controls, 766 mild cognitive impairment [MCI], and 283 AD). Then, we also explored the biological basis behind these morphological changes through gene enrichment analysis and cell-specific analysis. We found that RMCS probes of the hippocampus and medial temporal lobe were significantly altered in AD and MCI, with these differences being spatially related to the expression of AD-risk genes. In addition, gene enrichment analysis revealed that the modulation of chemical synaptic transmission is the most relevant biological process associated with the altered RMCS in AD. Notably, neuronal cells were found to be the most pertinent cells in the altered RMCS. Our findings shed light on understanding the biological basis of structural connectome changes in AD, which may ultimately lead to more effective diagnostic and therapeutic strategies for this devastating disease.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Connectome , Humans , Aged , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Magnetic Resonance Imaging , Brain/diagnostic imaging , Brain/metabolism , Temporal Lobe/diagnostic imaging , Temporal Lobe/metabolism , Cognitive Dysfunction/diagnostic imaging , Transcription, Genetic
3.
Hum Brain Mapp ; 44(9): 3467-3480, 2023 06 15.
Article in English | MEDLINE | ID: mdl-36988434

ABSTRACT

Alzheimer's disease (AD) is a common neurodegeneration disease associated with substantial disruptions in the brain network. However, most studies investigated static resting-state functional connections, while the alteration of dynamic functional connectivity in AD remains largely unknown. This study used group independent component analysis and the sliding-window method to estimate the subject-specific dynamic connectivity states in 1704 individuals from three data sets. Informative inherent states were identified by the multivariate pattern classification method, and classifiers were built to distinguish ADs from normal controls (NCs) and to classify mild cognitive impairment (MCI) patients with informative inherent states similar to ADs or not. In addition, MCI subgroups with heterogeneous functional states were examined in the context of different cognition decline trajectories. Five informative states were identified by feature selection, mainly involving functional connectivity belonging to the default mode network and working memory network. The classifiers discriminating AD and NC achieved the mean area under the receiver operating characteristic curve of 0.87 with leave-one-site-out cross-validation. Alterations in connectivity strength, fluctuation, and inter-synchronization were found in AD and MCIs. Moreover, individuals with MCI were clustered into two subgroups, which had different degrees of atrophy and different trajectories of cognition decline progression. The present study uncovered the alteration of dynamic functional connectivity in AD and highlighted that the dynamic states could be powerful features to discriminate patients from NCs. Furthermore, it demonstrated that these states help to identify MCIs with faster cognition decline and might contribute to the early prevention of AD.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Humans , Magnetic Resonance Imaging/methods , Brain/diagnostic imaging , Machine Learning
4.
BMC Med ; 21(1): 250, 2023 07 10.
Article in English | MEDLINE | ID: mdl-37424013

ABSTRACT

BACKGROUND: Inflammation has been implicated in the pathology of schizophrenia and may cause neuronal cell death and dendrite loss. Neuroimaging studies have highlighted longitudinal brain structural changes in patients with schizophrenia, yet it is unclear whether this is related to inflammation. We aim to address this question, by relating brain structural changes with the transcriptional profile of inflammation markers in the early stage of schizophrenia. METHODS: Thirty-eight patients with first-episode schizophrenia and 51 healthy controls were included. High-resolution T1-weighted magnetic resonance imaging (MRI) and clinical assessments were performed at baseline and 2 ~ 6 months follow-up for all subjects. Changes in the brain structure were analyzed using surface-based morphological analysis and correlated with the expression of immune cells-related gene sets of interest reported by previous reviews. Transcriptional data were retrieved from the Allen Human Brain Atlas. Furthermore, we examined the brain structural changes and peripheral inflammation markers in association with behavioral symptoms and cognitive functioning in patients. RESULTS: Patients exhibited accelerated cortical thickness decrease in the left frontal cortices, less decrease or an increase in the superior parietal lobule and right lateral occipital lobe, and increased volume in the bilateral pallidum, compared with controls. Changes in cortical thickness correlated with the transcriptional level of monocyte across cortical regions in patients (r = 0.54, p < 0.01), but not in controls (r = - 0.05, p = 0.76). In addition, cortical thickness change in the left superior parietal lobule positively correlated with changes in digital span-backward test scores in patients. CONCLUSIONS: Patients with schizophrenia exhibit regional-specific cortical thickness changes in the prefrontal and parietooccipital cortices, which is related to their cognitive impairment. Inflammation may be an important factor contributing to cortical thinning in first-episode schizophrenia. Our findings suggest that the immunity-brain-behavior association may play a crucial role in the pathogenesis of schizophrenia.


Subject(s)
Schizophrenia , Humans , Schizophrenia/diagnostic imaging , Schizophrenia/genetics , Magnetic Resonance Imaging/methods , Brain/diagnostic imaging , Brain/pathology , Cognition , Cerebral Cortex/pathology
5.
Cereb Cortex ; 32(5): 1024-1039, 2022 02 19.
Article in English | MEDLINE | ID: mdl-34378030

ABSTRACT

Functional brain networks require dynamic reconfiguration to support flexible cognitive function. However, the developmental principles shaping brain network dynamics remain poorly understood. Here, we report the longitudinal development of large-scale brain network dynamics during childhood and adolescence, and its connection with gene expression profiles. Using a multilayer network model, we show the temporally varying modular architecture of child brain networks, with higher network switching primarily in the association cortex and lower switching in the primary regions. This topographical profile exhibits progressive maturation, which manifests as reduced modular dynamics, particularly in the transmodal (e.g., default-mode and frontoparietal) and sensorimotor regions. These developmental refinements mediate age-related enhancements of global network segregation and are linked with the expression profiles of genes associated with the enrichment of ion transport and nucleobase-containing compound transport. These results highlight a progressive stabilization of brain dynamics, which expand our understanding of the neural mechanisms that underlie cognitive development.


Subject(s)
Brain , Magnetic Resonance Imaging , Adolescent , Brain Mapping , Cerebral Cortex , Child , Cognition , Humans , Magnetic Resonance Imaging/methods , Neural Pathways
6.
Hum Brain Mapp ; 43(3): 885-901, 2022 02 15.
Article in English | MEDLINE | ID: mdl-34862695

ABSTRACT

Multiscale integration of gene transcriptomic and neuroimaging data is becoming a widely used approach for exploring the molecular underpinnings of large-scale brain organization in health and disease. Proper statistical evaluation of determined associations between imaging-based phenotypic and transcriptomic data is key in these explorations, in particular to establish whether observed associations exceed "chance level" of random, nonspecific effects. Recent approaches have shown the importance of statistical models that can correct for spatial autocorrelation effects in the data to avoid inflation of reported statistics. Here, we discuss the need for examination of a second category of statistical models in transcriptomic-neuroimaging analyses, namely those that can provide "gene specificity." By means of a couple of simple examples of commonly performed transcriptomic-neuroimaging analyses, we illustrate some of the potentials and challenges of transcriptomic-imaging analyses, showing that providing gene specificity on observed transcriptomic-neuroimaging effects is of high importance to avoid reports of nonspecific effects. Through means of simulations we show that the rate of reported nonspecific effects (i.e., effects that cannot be specifically linked to a specific gene or gene-set) can run as high as 60%, with only less than 5% of transcriptomic-neuroimaging associations observed through ordinary linear regression analyses showing both spatial and gene specificity. We provide a discussion, a tutorial, and an easy-to-use toolbox for the different options of null models in transcriptomic-neuroimaging analyses.


Subject(s)
Brain Diseases , Brain , Models, Statistical , Neuroimaging , Transcriptome , Brain/diagnostic imaging , Brain/physiology , Brain Diseases/diagnostic imaging , Brain Diseases/genetics , Connectome , Humans
7.
Neuroimage ; 239: 118274, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34146709

ABSTRACT

The parcellation of the brain's cortical surface into anatomically and/or functionally distinct areas is a topic of ongoing investigation and interest. We provide digital versions of six classical human brain atlases in common MRI space. The cortical atlases represent a range of modalities, including cyto- and myeloarchitecture (Campbell, Smith, Brodmann and Von Economo), myelogenesis (Flechsig), and mappings of symptomatic information in relation to the spatial location of brain lesions (Kleist). Digital reconstructions of these important cortical atlases widen the range of modalities for which cortex-wide imaging atlases are currently available and offer the opportunity to compare and combine microstructural and lesion-based functional atlases with in-vivo imaging-based atlases.


Subject(s)
Atlases as Topic , Cerebral Cortex/anatomy & histology , Connectome , Magnetic Resonance Imaging/methods , Positron-Emission Tomography/methods , Cerebral Cortex/cytology , Cerebral Cortex/diagnostic imaging , Diffusion Tensor Imaging/methods , Humans , Image Processing, Computer-Assisted , Medical Illustration , Software , White Matter/diagnostic imaging
8.
Neuroimage ; 245: 118743, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34800667

ABSTRACT

It has been revealed that intersubject variability (ISV) in intrinsic functional connectivity (FC) is associated with a wide variety of cognitive and behavioral performances. However, the underlying organizational principle of ISV in FC and its related gene transcriptional profiles remain unclear. Using resting-state fMRI data from the Human Connectome Project (299 adult participants) and microarray gene expression data from the Allen Human Brain Atlas, we conducted a transcription-neuroimaging association study to investigate the spatial configurations of ISV in intrinsic FC and their associations with spatial gene transcriptional profiles. We found that the multimodal association cortices showed the greatest ISV in FC, while the unimodal cortices and subcortical areas showed the least ISV. Importantly, partial least squares regression analysis revealed that the transcriptional profiles of genes associated with human accelerated regions (HARs) could explain 31.29% of the variation in the spatial distribution of ISV in FC. The top-related genes in the transcriptional profiles were enriched for the development of the central nervous system, neurogenesis and the cellular components of synapse. Moreover, we observed that the effect of gene expression profile on the heterogeneous distribution of ISV in FC was significantly mediated by the cerebral blood flow configuration. These findings highlighted the spatial arrangement of ISV in FC and their coupling with variations in transcriptional profiles and cerebral blood flow supply.


Subject(s)
Connectome , Gene Expression Profiling , Magnetic Resonance Imaging , Cerebrovascular Circulation , Humans , Image Processing, Computer-Assisted
9.
Hum Brain Mapp ; 42(15): 4996-5009, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34272784

ABSTRACT

Ultra-high field MRI across the depth of the cortex has the potential to provide anatomically precise biomarkers and mechanistic insights into neurodegenerative disease like Huntington's disease that show layer-selective vulnerability. Here we compare multi-parametric mapping (MPM) measures across cortical depths for a 7T 500 µm whole brain acquisition to (a) layer-specific cell measures from the von Economo histology atlas, (b) layer-specific gene expression, using the Allen Human Brain atlas and (c) white matter connections using high-fidelity diffusion tractography, at a 1.3 mm isotropic voxel resolution, from a 300mT/m Connectom MRI system. We show that R2*, but not R1, across cortical depths is highly correlated with layer-specific cell number and layer-specific gene expression. R1- and R2*-weighted connectivity strength of cortico-striatal and intra-hemispheric cortical white matter connections was highly correlated with grey matter R1 and R2* across cortical depths. Limitations of the layer-specific relationships demonstrated are at least in part related to the high cross-correlations of von Economo atlas cell counts and layer-specific gene expression across cortical layers. These findings demonstrate the potential and limitations of combining 7T MPMs, gene expression and white matter connections to provide an anatomically precise framework for tracking neurodegenerative disease.


Subject(s)
Cerebral Cortex , Diffusion Magnetic Resonance Imaging , Echo-Planar Imaging , Gene Expression/physiology , Myelin Sheath , Nerve Net , White Matter , Adult , Atlases as Topic , Cerebral Cortex/anatomy & histology , Cerebral Cortex/diagnostic imaging , Female , Humans , Male , Nerve Net/anatomy & histology , Nerve Net/diagnostic imaging , Neurodegenerative Diseases/diagnostic imaging , White Matter/anatomy & histology , White Matter/diagnostic imaging , Young Adult
10.
PLoS Comput Biol ; 16(2): e1007526, 2020 02.
Article in English | MEDLINE | ID: mdl-32027645

ABSTRACT

We approach the C. elegans connectome as an information processing network that receives input from about 90 sensory neurons, processes that information through a highly recurrent network of about 80 interneurons, and it produces a coordinated output from about 120 motor neurons that control the nematode's muscles. We focus on the feedforward flow of information from sensory neurons to motor neurons, and apply a recently developed network analysis framework referred to as the "hourglass effect". The analysis reveals that this feedforward flow traverses a small core ("hourglass waist") that consists of 10-15 interneurons. These are mostly the same interneurons that were previously shown (using a different analytical approach) to constitute the "rich-club" of the C. elegans connectome. This result is robust to the methodology that separates the feedforward from the feedback flow of information. The set of core interneurons remains mostly the same when we consider only chemical synapses or the combination of chemical synapses and gap junctions. The hourglass organization of the connectome suggests that C. elegans has some similarities with encoder-decoder artificial neural networks in which the input is first compressed and integrated in a low-dimensional latent space that encodes the given data in a more efficient manner, followed by a decoding network through which intermediate-level sub-functions are combined in different ways to compute the correlated outputs of the network. The core neurons at the hourglass waist represent the information bottleneck of the system, balancing the representation accuracy and compactness (complexity) of the given sensory information.


Subject(s)
Caenorhabditis elegans/physiology , Connectome , Animals , Computational Biology , Gap Junctions/physiology , Interneurons/physiology , Motor Neurons/physiology , Sensory Receptor Cells/physiology , Synapses/physiology
11.
Cereb Cortex ; 30(3): 1357-1365, 2020 03 14.
Article in English | MEDLINE | ID: mdl-31504277

ABSTRACT

Degree centrality is a widely used measure in complex networks. Within the brain, degree relates to other topological features, with high-degree nodes (i.e., hubs) exhibiting high betweenness centrality, participation coefficient, and within-module z-score. However, increasing evidence from neuroanatomical and predictive processing literature suggests that topological properties of a brain network may also be impacted by topography, that is, anatomical (spatial) distribution. More specifically, cortical limbic areas (agranular and dysgranular cortices), which occupy an anatomically central position, have been proposed to be topologically central and well suited to initiate predictions in the cerebral cortex. We estimated anatomical centrality and showed that it positively correlated with betweenness centrality, participation coefficient, and communicability, analogously to degree. In contrast to degree, however, anatomical centrality negatively correlated with within-module z-score. Our data suggest that degree centrality and anatomical centrality reflect distinct contributions to cortical organization. Whereas degree would be more related to the amount of information integration performed by an area, anatomical centrality would be more related to an area's position in the predictive hierarchy. Highly anatomically central areas may function as "high-level connectors," integrating already highly integrated information across modules. These results are consistent with a high-level, domain-general limbic workspace, integrated by highly anatomically central cortical areas.


Subject(s)
Cerebral Cortex/anatomy & histology , Connectome/methods , Adult , Female , Humans , Limbic System/anatomy & histology , Magnetic Resonance Imaging , Male , Neural Pathways/anatomy & histology , Young Adult
12.
Hum Brain Mapp ; 38(5): 2734-2750, 2017 05.
Article in English | MEDLINE | ID: mdl-28256774

ABSTRACT

Recent imaging connectome studies demonstrated that the human functional brain network follows an efficient small-world topology with cohesive functional modules and highly connected hubs. However, the functional motif patterns that represent the underlying information flow remain largely unknown. Here, we investigated motif patterns within directed human functional brain networks, which were derived from resting-state functional magnetic resonance imaging data with controlled confounding hemodynamic latencies. We found several significantly recurring motifs within the network, including the two-node reciprocal motif and five classes of three-node motifs. These recurring motifs were distributed in distinct patterns to support intra- and inter-module functional connectivity, which also promoted integration and segregation in network organization. Moreover, the significant participation of several functional hubs in the recurring motifs exhibited their critical role in global integration. Collectively, our findings highlight the basic architecture governing brain network organization and provide insight into the information flow mechanism underlying intrinsic brain activities. Hum Brain Mapp 38:2734-2750, 2017. © 2017 Wiley Periodicals, Inc.


Subject(s)
Brain Mapping , Brain/physiology , Models, Neurological , Nerve Net/physiology , Adult , Brain/diagnostic imaging , Female , Humans , Image Processing, Computer-Assisted , Magnetic Resonance Imaging , Male , Middle Aged , Nerve Net/diagnostic imaging , Neural Pathways/diagnostic imaging , Neural Pathways/physiology , Oxygen/blood , Reaction Time/physiology , Reproducibility of Results , Young Adult
13.
Adv Sci (Weinh) ; 11(2): e2304397, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37933983

ABSTRACT

Infections caused by Enterobacterales producing New Delhi Metallo-ß-lactamases (NDMs), Zn(II)-dependent enzymes hydrolyzing carbapenems, are difficult to treat. Depriving Zn(II) to inactivate NDMs is an effective solution to reverse carbapenems resistance in NDMs-producing bacteria. However, specific Zn(II) deprivation and better bacterial outer membrane penetrability in vivo are challenges. Herein, authors present a pathogen-primed liposomal antibiotic booster (M-MFL@MB), facilitating drugs transportation into bacteria and removing Zn(II) from NDMs. M-MFL@MB introduces bismuth nanoclusters (BiNCs) as a storage tank of Bi(III) for achieving ROS-initiated Zn(II) removal. Inspired by bacteria-specific maltodextrin transport pathway, meropenem-loaded BiNCs are camouflaged by maltodextrin-cloaked membrane fusion liposome to cross the bacterial envelope barrier via selectively targeting bacteria and directly outer membrane fusion. This fusion disturbs bacterial membrane homeostasis, then triggers intracellular ROS amplification, which activates Bi(III)-mediated Zn(II) replacement and meropenem release, realizing more precise and efficient NDMs producer treatment. Benefiting from specific bacteria-targeting, adequate drugs intracellular accumulation and self-activation Zn(II) replacement, M-MFL@MB rescues all mice infected by NDM producer without systemic side effects. Additionally, M-MFL@MB decreases the bacterial outer membrane vesicles secretion, slowing down NDMs producer's transmission by over 35 times. Taken together, liposomal antibiotic booster as an efficient and safe tool provides new strategy for tackling NDMs producer-induced infections.


Subject(s)
Anti-Bacterial Agents , Carbapenems , Mice , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Carbapenems/therapeutic use , Carbapenems/pharmacology , Meropenem/pharmacology , Escherichia coli , Liposomes , Reactive Oxygen Species , Microbial Sensitivity Tests
14.
Psychoradiology ; 4: kkae005, 2024.
Article in English | MEDLINE | ID: mdl-38694267

ABSTRACT

Background: Schizophrenia is a polygenic disorder associated with changes in brain structure and function. Integrating macroscale brain features with microscale genetic data may provide a more complete overview of the disease etiology and may serve as potential diagnostic markers for schizophrenia. Objective: We aim to systematically evaluate the impact of multi-scale neuroimaging and transcriptomic data fusion in schizophrenia classification models. Methods: We collected brain imaging data and blood RNA sequencing data from 43 patients with schizophrenia and 60 age- and gender-matched healthy controls, and we extracted multi-omics features of macroscale brain morphology, brain structural and functional connectivity, and gene transcription of schizophrenia risk genes. Multi-scale data fusion was performed using a machine learning integration framework, together with several conventional machine learning methods and neural networks for patient classification. Results: We found that multi-omics data fusion in conventional machine learning models achieved the highest accuracy (AUC ~0.76-0.92) in contrast to the single-modality models, with AUC improvements of 8.88 to 22.64%. Similar findings were observed for the neural network, showing an increase of 16.57% for the multimodal classification model (accuracy 71.43%) compared to the single-modal average. In addition, we identified several brain regions in the left posterior cingulate and right frontal pole that made a major contribution to disease classification. Conclusion: We provide empirical evidence for the increased accuracy achieved by imaging genetic data integration in schizophrenia classification. Multi-scale data fusion holds promise for enhancing diagnostic precision, facilitating early detection and personalizing treatment regimens in schizophrenia.

15.
Sci Adv ; 10(41): eado8837, 2024 Oct 11.
Article in English | MEDLINE | ID: mdl-39392880

ABSTRACT

The intricate spatial configurations of brain networks offer essential insights into understanding the specific patterns of brain abnormalities and the underlying biological mechanisms associated with Alzheimer's disease (AD), normal aging, and other neurodegenerative disorders. This study investigated alterations in the topographical structure of the brain related to aging and neurodegenerative diseases by analyzing brain gradients derived from structural MRI data across multiple cohorts (n = 7323). The analysis identified distinct gradient patterns in AD, aging, and other neurodegenerative conditions. Gene enrichment analysis indicated that inorganic ion transmembrane transport was the most significant term in normal aging, while chemical synaptic transmission is a common enrichment term across various neurodegenerative diseases. Moreover, the findings show that each disorder exhibits unique dysfunctional neurophysiological characteristics. These insights are pivotal for elucidating the distinct biological mechanisms underlying AD, thereby enhancing our understanding of its unique clinical phenotypes in contrast to normal aging and other neurodegenerative disorders.


Subject(s)
Alzheimer Disease , Brain , Connectome , Magnetic Resonance Imaging , Alzheimer Disease/pathology , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/metabolism , Humans , Brain/diagnostic imaging , Brain/pathology , Brain/metabolism , Aging/pathology , Aged , Female , Male , Middle Aged
16.
Article in English | MEDLINE | ID: mdl-37286292

ABSTRACT

BACKGROUND: Psychiatric conditions show overlap in their symptoms, genetics, and involvement in brain areas and circuits. Structural alterations in the brain have been found to run in parallel with expression profiles of risk genes at the level of the brain transcriptome, which may point toward a potential transdiagnostic vulnerability of the brain to disease processes. METHODS: We characterized the transcriptomic vulnerability of the cortex across 4 major psychiatric disorders based on collated data from patients with psychiatric disorders (n = 390) and matched control participants (n = 293). We compared normative expression profiles of risk genes linked to schizophrenia, bipolar disorder, autism spectrum disorder, and major depressive disorder to examine cross-disorder overlap in spatial expression profiles across the cortex and their concordance with a magnetic resonance imaging-derived cross-disorder profile of structural brain alterations. RESULTS: We showed high expression of psychiatric risk genes converging on multimodal cortical regions of the limbic, ventral attention, and default mode networks versus primary somatosensory networks. Risk genes were found to be enriched among genes associated with the magnetic resonance imaging cross-disorder profile, suggestive of a common link between brain anatomy and the transcriptome in psychiatric conditions. Characterization of this cross-disorder structural alteration map further shows enrichment for gene markers of astrocytes, microglia, and supragranular cortical layers. CONCLUSIONS: Our findings suggest that normative expression profiles of disorder risk genes confer a shared and spatially patterned vulnerability of the cortex across multiple psychiatric conditions. Transdiagnostic overlap in transcriptomic risk suggests a common pathway to brain dysfunction across psychiatric disorders.


Subject(s)
Autism Spectrum Disorder , Bipolar Disorder , Depressive Disorder, Major , Schizophrenia , Humans , Depressive Disorder, Major/genetics , Bipolar Disorder/genetics , Schizophrenia/diagnostic imaging , Schizophrenia/genetics , Schizophrenia/metabolism , Transcriptome , Autism Spectrum Disorder/diagnostic imaging , Autism Spectrum Disorder/genetics , Brain/pathology , Neuroimaging
17.
Adv Healthc Mater ; 12(25): e2300449, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37431870

ABSTRACT

Clinical treatment of multidrug resistant (MDR) pathogens-induced infection is emerging as a growing challenge in global public health due to the limited selection of clinically available antibiotics. Nanozymes as artificial enzymes that mimicked natural enzyme-like activities, are received great attention for combating MDR pathogens. However, the relatively deficient catalytic activity in the infectious microenvironment and inability to precisely targeting pathogen restrains their clinical anti-MDR applications. Here, pathogen-targeting bimetallic BiPt nanozymes for nanocatalytic therapy against MDR pathogen are reported. Benefiting from electronic coordination effect, BiPt nanozymes exhibit dual-enzymatic activities, including peroxidase-mimic and oxidase-mimic activities. Moreover, the catalytic efficiency can be efficiently increased 300-fold by ultrasound under inflammatory microenvironment. Notably, BiPt nanozyme is further cloaked with a platelet-bacteria hybrid membrane (BiPt@HMVs), thus presenting excellent homing effect to infectious sites and precise homologous targeting to pathogen. By integrating accurate targeting with highly efficient catalytic, BiPt@HMVs can eliminate carbapenem-resistant Enterobacterales and methicillin-resistant Staphylococcus aureus in osteomyelitis rats model, muscle-infected mice model, and pneumonia mice model. The work provides an alternative strategy based on nanozymes for clinically addressing MDR bacteria-induced infections.

18.
Biol Psychiatry ; 94(2): 174-183, 2023 07 15.
Article in English | MEDLINE | ID: mdl-36803976

ABSTRACT

BACKGROUND: Schizophrenia (SCZ) and bipolar disorder (BD) are severe psychiatric conditions that can involve symptoms of psychosis and cognitive dysfunction. The 2 conditions share symptomatology and genetic etiology and are regularly hypothesized to share underlying neuropathology. Here, we examined how genetic liability to SCZ and BD shapes normative variations in brain connectivity. METHODS: We examined the effect of the combined genetic liability for SCZ and BD on brain connectivity from two perspectives. First, we examined the association between polygenic scores for SCZ and BD for 19,778 healthy subjects from the UK Biobank and individual variation in brain structural connectivity reconstructed by means of diffusion weighted imaging data. Second, we conducted genome-wide association studies using genotypic and imaging data from the UK Biobank, taking SCZ-/BD-involved brain circuits as phenotypes of interest. RESULTS: Our findings showed brain circuits of superior parietal and posterior cingulate regions to be associated with polygenic liability for SCZ and BD, circuitry that overlaps with brain networks involved in disease conditions (r = 0.239, p < .001). Genome-wide association study analysis showed 9 significant genomic loci associated with SCZ-involved circuits and 14 loci associated with BD-involved circuits. Genes related to SCZ-/BD-involved circuits were significantly enriched in gene sets previously reported in genome-wide association studies for SCZ and BD. CONCLUSIONS: Our findings suggest that polygenic liability of SCZ and BD is associated with normative individual variation in brain circuitry.


Subject(s)
Bipolar Disorder , Connectome , Schizophrenia , Humans , Schizophrenia/diagnostic imaging , Schizophrenia/genetics , Bipolar Disorder/genetics , Genome-Wide Association Study , Genetic Predisposition to Disease
19.
eNeuro ; 10(4)2023 04.
Article in English | MEDLINE | ID: mdl-36882310

ABSTRACT

Functional connectivity within resting-state networks (RSN-FC) is vital for cognitive functioning. RSN-FC is heritable and partially translates to the anatomic architecture of white matter, but the genetic component of structural connections of RSNs (RSN-SC) and their potential genetic overlap with RSN-FC remain unknown. Here, we perform genome-wide association studies (N discovery = 24,336; N replication = 3412) and annotation on RSN-SC and RSN-FC. We identify genes for visual network-SC that are involved in axon guidance and synaptic functioning. Genetic variation in RSN-FC impacts biological processes relevant to brain disorders that previously were only phenotypically associated with RSN-FC alterations. Correlations of the genetic components of RSNs are mostly observed within the functional domain, whereas less overlap is observed within the structural domain and between the functional and structural domains. This study advances the understanding of the complex functional organization of the brain and its structural underpinnings from a genetics viewpoint.


Subject(s)
Brain Mapping , Genome-Wide Association Study , Magnetic Resonance Imaging , Brain/diagnostic imaging , Cognition , Nerve Net/diagnostic imaging
20.
EClinicalMedicine ; 65: 102276, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37954904

ABSTRACT

Background: Alzheimer's disease (AD) is a prevalent neurodegenerative disorder that poses a worldwide public health challenge. A neuroimaging biomarker would significantly improve early diagnosis and intervention, ultimately enhancing the quality of life for affected individuals and reducing the burden on healthcare systems. Methods: Cross-sectional and longitudinal data (10,099 participants with 13,380 scans) from 12 independent datasets were used in the present study (this study was performed between September 1, 2021 and February 15, 2023). The Individual Brain-Related Abnormalities In Neurodegeneration (IBRAIN) score was developed via integrated regional- and network-based measures under an ensemble machine learning model based on structural MRI data. We systematically assessed whether IBRAIN could be a neuroimaging biomarker for AD. Findings: IBRAIN accurately differentiated individuals with AD from NCs (AUC = 0.92) and other neurodegenerative diseases, including Frontotemporal dementia (FTD), Parkinson's disease (PD), Vascular dementia (VaD) and Amyotrophic Lateral Sclerosis (ALS) (AUC = 0.92). IBRAIN was significantly correlated to clinical measures and gene expression, enriched in immune process and protein metabolism. The IBRAIN score exhibited a significant ability to reveal the distinct progression of prodromal AD (i.e., Mild cognitive impairment, MCI) (Hazard Ratio (HR) = 6.52 [95% CI: 4.42∼9.62], p < 1 × 10-16), which offers similar powerful performance with Cerebrospinal Fluid (CSF) Aß (HR = 3.78 [95% CI: 2.63∼5.43], p = 2.13 × 10-14) and CSF Tau (HR = 3.77 [95% CI: 2.64∼5.39], p = 9.53 × 10-15) based on the COX and Log-rank test. Notably, the IBRAIN shows comparable sensitivity (beta = -0.70, p < 1 × 10-16) in capturing longitudinal changes in individuals with conversion to AD than CSF Aß (beta = -0.26, p = 4.40 × 10-9) and CSF Tau (beta = 0.12, p = 1.02 × 10-5). Interpretation: Our findings suggested that IBRAIN is a biologically relevant, specific, and sensitive neuroimaging biomarker that can serve as a clinical measure to uncover prodromal AD progression. It has strong potential for application in future clinical practice and treatment trials. Funding: Science and Technology Innovation 2030 Major Projects, the National Natural Science Foundation of China, Beijing Natural Science Funds, the Fundamental Research Funds for the CentralUniversity, and the Startup Funds for Talents at Beijing Normal University.

SELECTION OF CITATIONS
SEARCH DETAIL