Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 8.056
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 177(4): 1050-1066.e14, 2019 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-30982596

RESUMEN

Calcium imaging using two-photon scanning microscopy has become an essential tool in neuroscience. However, in its typical implementation, the tradeoffs between fields of view, acquisition speeds, and depth restrictions in scattering brain tissue pose severe limitations. Here, using an integrated systems-wide optimization approach combined with multiple technical innovations, we introduce a new design paradigm for optical microscopy based on maximizing biological information while maintaining the fidelity of obtained neuron signals. Our modular design utilizes hybrid multi-photon acquisition and allows volumetric recording of neuroactivity at single-cell resolution within up to 1 × 1 × 1.22 mm volumes at up to 17 Hz in awake behaving mice. We establish the capabilities and potential of the different configurations of our imaging system at depth and across brain regions by applying it to in vivo recording of up to 12,000 neurons in mouse auditory cortex, posterior parietal cortex, and hippocampus.


Asunto(s)
Microscopía/métodos , Imagen Molecular/métodos , Neuroimagen/métodos , Animales , Encéfalo/fisiología , Calcio/metabolismo , Femenino , Hipocampo/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/fisiología , Análisis de la Célula Individual/métodos
2.
Cell ; 171(7): 1649-1662.e10, 2017 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-29198526

RESUMEN

Animals generate complex patterns of behavior across development that may be shared or unique to individuals. Here, we examine the contributions of developmental programs and individual variation to behavior by monitoring single Caenorhabditis elegans nematodes over their complete developmental trajectories and quantifying their behavior at high spatiotemporal resolution. These measurements reveal reproducible trajectories of spontaneous foraging behaviors that are stereotyped within and between developmental stages. Dopamine, serotonin, the neuropeptide receptor NPR-1, and the TGF-ß peptide DAF-7 each have stage-specific effects on behavioral trajectories, implying the existence of a modular temporal program controlled by neuromodulators. In addition, a fraction of individuals within isogenic populations raised in controlled environments have consistent, non-genetic behavioral biases that persist across development. Several neuromodulatory systems increase or decrease the degree of non-genetic individuality to shape sustained patterns of behavior across the population.


Asunto(s)
Variación Biológica Individual , Caenorhabditis elegans/crecimiento & desarrollo , Caenorhabditis elegans/fisiología , Neuropéptidos/metabolismo , Animales , Conducta Animal , Dopamina/metabolismo , Regulación de la Expresión Génica , Larva/fisiología , Neuroimagen/instrumentación , Neuroimagen/métodos , Neuropéptidos/genética , Receptores de Serotonina/genética , Receptores de Serotonina/metabolismo
3.
Cell ; 165(7): 1803-1817, 2016 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-27180908

RESUMEN

A scalable and high-throughput method to identify precise subcellular localization of endogenous proteins is essential for integrative understanding of a cell at the molecular level. Here, we developed a simple and generalizable technique to image endogenous proteins with high specificity, resolution, and contrast in single cells in mammalian brain tissue. The technique, single-cell labeling of endogenous proteins by clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9-mediated homology-directed repair (SLENDR), uses in vivo genome editing to insert a sequence encoding an epitope tag or a fluorescent protein to a gene of interest by CRISPR-Cas9-mediated homology-directed repair (HDR). Single-cell, HDR-mediated genome editing was achieved by delivering the editing machinery to dividing neuronal progenitors through in utero electroporation. We demonstrate that SLENDR allows rapid determination of the localization and dynamics of many endogenous proteins in various cell types, regions, and ages in the brain. Thus, SLENDR provides a high-throughput platform to map the subcellular localization of endogenous proteins with the resolution of micro- to nanometers in the brain.


Asunto(s)
Química Encefálica , Mapeo Encefálico/métodos , Proteínas del Tejido Nervioso/análisis , Encéfalo/embriología , Sistemas CRISPR-Cas , Ingeniería Genética , Neuroimagen/métodos , Neuronas/química , Análisis de la Célula Individual
4.
Cell ; 165(7): 1789-1802, 2016 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-27238021

RESUMEN

Understanding how neural information is processed in physiological and pathological states would benefit from precise detection, localization, and quantification of the activity of all neurons across the entire brain, which has not, to date, been achieved in the mammalian brain. We introduce a pipeline for high-speed acquisition of brain activity at cellular resolution through profiling immediate early gene expression using immunostaining and light-sheet fluorescence imaging, followed by automated mapping and analysis of activity by an open-source software program we term ClearMap. We validate the pipeline first by analysis of brain regions activated in response to haloperidol. Next, we report new cortical regions downstream of whisker-evoked sensory processing during active exploration. Last, we combine activity mapping with axon tracing to uncover new brain regions differentially activated during parenting behavior. This pipeline is widely applicable to different experimental paradigms, including animal species for which transgenic activity reporters are not readily available.


Asunto(s)
Conducta Animal , Inmunohistoquímica , Neuroimagen/métodos , Animales , Antipsicóticos/administración & dosificación , Encéfalo/metabolismo , Conducta Exploratoria , Genes Inmediatos-Precoces , Haloperidol/administración & dosificación , Ratones , Ratones Endogámicos C57BL
5.
Cell ; 157(3): 726-39, 2014 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-24746791

RESUMEN

Systems-level identification and analysis of cellular circuits in the brain will require the development of whole-brain imaging with single-cell resolution. To this end, we performed comprehensive chemical screening to develop a whole-brain clearing and imaging method, termed CUBIC (clear, unobstructed brain imaging cocktails and computational analysis). CUBIC is a simple and efficient method involving the immersion of brain samples in chemical mixtures containing aminoalcohols, which enables rapid whole-brain imaging with single-photon excitation microscopy. CUBIC is applicable to multicolor imaging of fluorescent proteins or immunostained samples in adult brains and is scalable from a primate brain to subcellular structures. We also developed a whole-brain cell-nuclear counterstaining protocol and a computational image analysis pipeline that, together with CUBIC reagents, enable the visualization and quantification of neural activities induced by environmental stimulation. CUBIC enables time-course expression profiling of whole adult brains with single-cell resolution.


Asunto(s)
Neuroimagen/métodos , Animales , Encéfalo/citología , Callithrix , Indicadores y Reactivos/química , Ratones , Microscopía/métodos
6.
Cell ; 157(5): 1230-42, 2014 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-24855954

RESUMEN

The complexity and cellular heterogeneity of neural circuitry presents a major challenge to understanding the role of discrete neural populations in controlling behavior. While neuroanatomical methods enable high-resolution mapping of neural circuitry, these approaches do not allow systematic molecular profiling of neurons based on their connectivity. Here, we report the development of an approach for molecularly profiling projective neurons. We show that ribosomes can be tagged with a camelid nanobody raised against GFP and that this system can be engineered to selectively capture translating mRNAs from neurons retrogradely labeled with GFP. Using this system, we profiled neurons projecting to the nucleus accumbens. We then used an AAV to selectively profile midbrain dopamine neurons projecting to the nucleus accumbens. By comparing the captured mRNAs from each experiment, we identified a number of markers specific to VTA dopaminergic projection neurons. The current method provides a means for profiling neurons based on their projections.


Asunto(s)
Proteínas Fluorescentes Verdes/análisis , Neurobiología/métodos , Neuroimagen/métodos , Neuronas/citología , Ribosomas/química , Animales , Anticuerpos/genética , Proteínas Fluorescentes Verdes/metabolismo , Inmunoprecipitación , Ratones Transgénicos , Núcleo Accumbens/citología , Biosíntesis de Proteínas
7.
Annu Rev Neurosci ; 42: 295-313, 2019 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-31283896

RESUMEN

Light-sheet microscopy is an imaging approach that offers unique advantages for a diverse range of neuroscience applications. Unlike point-scanning techniques such as confocal and two-photon microscopy, light-sheet microscopes illuminate an entire plane of tissue, while imaging this plane onto a camera. Although early implementations of light sheet were optimized for longitudinal imaging of embryonic development in small specimens, emerging implementations are capable of capturing light-sheet images in freely moving, unconstrained specimens and even the intact in vivo mammalian brain. Meanwhile, the unique photobleaching and signal-to-noise benefits afforded by light-sheet microscopy's parallelized detection deliver the ability to perform volumetric imaging at much higher speeds than can be achieved using point scanning. This review describes the basic principles and evolution of light-sheet microscopy, followed by perspectives on emerging applications and opportunities for both imaging large, cleared, and expanded neural tissues and high-speed, functional imaging in vivo.


Asunto(s)
Encéfalo/fisiología , Microscopía , Neuroimagen , Neurociencias , Animales , Humanos , Modelos Animales , Neuroimagen/métodos , Neurociencias/métodos , Relación Señal-Ruido
8.
Nat Methods ; 21(5): 804-808, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38191935

RESUMEN

Neuroimaging research requires purpose-built analysis software, which is challenging to install and may produce different results across computing environments. The community-oriented, open-source Neurodesk platform ( https://www.neurodesk.org/ ) harnesses a comprehensive and growing suite of neuroimaging software containers. Neurodesk includes a browser-accessible virtual desktop, command-line interface and computational notebook compatibility, allowing for accessible, flexible, portable and fully reproducible neuroimaging analysis on personal workstations, high-performance computers and the cloud.


Asunto(s)
Neuroimagen , Programas Informáticos , Neuroimagen/métodos , Humanos , Interfaz Usuario-Computador , Reproducibilidad de los Resultados , Encéfalo/diagnóstico por imagen
9.
Nat Methods ; 21(5): 809-813, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38605111

RESUMEN

Neuroscience is advancing standardization and tool development to support rigor and transparency. Consequently, data pipeline complexity has increased, hindering FAIR (findable, accessible, interoperable and reusable) access. brainlife.io was developed to democratize neuroimaging research. The platform provides data standardization, management, visualization and processing and automatically tracks the provenance history of thousands of data objects. Here, brainlife.io is described and evaluated for validity, reliability, reproducibility, replicability and scientific utility using four data modalities and 3,200 participants.


Asunto(s)
Nube Computacional , Neurociencias , Neurociencias/métodos , Humanos , Neuroimagen/métodos , Reproducibilidad de los Resultados , Programas Informáticos , Encéfalo/fisiología , Encéfalo/diagnóstico por imagen
10.
Nat Rev Neurosci ; 23(6): 361-375, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35444305

RESUMEN

Mapping human brain function is a long-standing goal of neuroscience that promises to inform the development of new treatments for brain disorders. Early maps of human brain function were based on locations of brain damage or brain stimulation that caused a functional change. Over time, this approach was largely replaced by technologies such as functional neuroimaging, which identify brain regions in which activity is correlated with behaviours or symptoms. Despite their advantages, these technologies reveal correlations, not causation. This creates challenges for interpreting the data generated from these tools and using them to develop treatments for brain disorders. A return to causal mapping of human brain function based on brain lesions and brain stimulation is underway. New approaches can combine these causal sources of information with modern neuroimaging and electrophysiology techniques to gain new insights into the functions of specific brain areas. In this Review, we provide a definition of causality for translational research, propose a continuum along which to assess the relative strength of causal information from human brain mapping studies and discuss recent advances in causal brain mapping and their relevance for developing treatments.


Asunto(s)
Encefalopatías , Neurociencias , Encéfalo/fisiología , Mapeo Encefálico/métodos , Humanos , Neuroimagen/métodos
11.
Proc Natl Acad Sci U S A ; 121(28): e2317458121, 2024 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-38950362

RESUMEN

Functional changes in the pediatric brain following neural injuries attest to remarkable feats of plasticity. Investigations of the neurobiological mechanisms that underlie this plasticity have largely focused on activation in the penumbra of the lesion or in contralesional, homotopic regions. Here, we adopt a whole-brain approach to evaluate the plasticity of the cortex in patients with large unilateral cortical resections due to drug-resistant childhood epilepsy. We compared the functional connectivity (FC) in patients' preserved hemisphere with the corresponding hemisphere of matched controls as they viewed and listened to a movie excerpt in a functional magnetic resonance imaging (fMRI) scanner. The preserved hemisphere was segmented into 180 and 200 parcels using two different anatomical atlases. We calculated all pairwise multivariate statistical dependencies between parcels, or parcel edges, and between 22 and 7 larger-scale functional networks, or network edges, aggregated from the smaller parcel edges. Both the left and right hemisphere-preserved patient groups had widespread reductions in FC relative to matched controls, particularly for within-network edges. A case series analysis further uncovered subclusters of patients with distinctive edgewise changes relative to controls, illustrating individual postoperative connectivity profiles. The large-scale differences in networks of the preserved hemisphere potentially reflect plasticity in the service of maintained and/or retained cognitive function.


Asunto(s)
Imagen por Resonancia Magnética , Neuroimagen , Humanos , Niño , Imagen por Resonancia Magnética/métodos , Femenino , Masculino , Adolescente , Neuroimagen/métodos , Epilepsia/cirugía , Epilepsia/fisiopatología , Epilepsia/diagnóstico por imagen , Corteza Cerebral/diagnóstico por imagen , Corteza Cerebral/fisiopatología , Corteza Cerebral/cirugía , Plasticidad Neuronal/fisiología , Epilepsia Refractaria/cirugía , Epilepsia Refractaria/diagnóstico por imagen , Epilepsia Refractaria/fisiopatología , Mapeo Encefálico/métodos , Lateralidad Funcional/fisiología
12.
Brief Bioinform ; 25(2)2024 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-38348747

RESUMEN

Integrating and analyzing multiple omics data sets, including genomics, proteomics and radiomics, can significantly advance researchers' comprehensive understanding of Alzheimer's disease (AD). However, current methodologies primarily focus on the main effects of genetic variation and protein, overlooking non-additive effects such as genotype-protein interaction (GPI) and correlation patterns in brain imaging genetics studies. Importantly, these non-additive effects could contribute to intermediate imaging phenotypes, finally leading to disease occurrence. In general, the interaction between genetic variations and proteins, and their correlations are two distinct biological effects, and thus disentangling the two effects for heritable imaging phenotypes is of great interest and need. Unfortunately, this issue has been largely unexploited. In this paper, to fill this gap, we propose $\textbf{M}$ulti-$\textbf{T}$ask $\textbf{G}$enotype-$\textbf{P}$rotein $\textbf{I}$nteraction and $\textbf{C}$orrelation disentangling method ($\textbf{MT-GPIC}$) to identify GPI and extract correlation patterns between them. To ensure stability and interpretability, we use novel and off-the-shelf penalties to identify meaningful genetic risk factors, as well as exploit the interconnectedness of different brain regions. Additionally, since computing GPI poses a high computational burden, we develop a fast optimization strategy for solving MT-GPIC, which is guaranteed to converge. Experimental results on the Alzheimer's Disease Neuroimaging Initiative data set show that MT-GPIC achieves higher correlation coefficients and classification accuracy than state-of-the-art methods. Moreover, our approach could effectively identify interpretable phenotype-related GPI and correlation patterns in high-dimensional omics data sets. These findings not only enhance the diagnostic accuracy but also contribute valuable insights into the underlying pathogenic mechanisms of AD.


Asunto(s)
Enfermedad de Alzheimer , Humanos , Enfermedad de Alzheimer/diagnóstico por imagen , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Multiómica , Genotipo , Neuroimagen/métodos , Fenotipo , Encéfalo/diagnóstico por imagen , Encéfalo/patología
13.
Proc Natl Acad Sci U S A ; 120(9): e2216399120, 2023 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-36802420

RESUMEN

Every year, millions of brain MRI scans are acquired in hospitals, which is a figure considerably larger than the size of any research dataset. Therefore, the ability to analyze such scans could transform neuroimaging research. Yet, their potential remains untapped since no automated algorithm is robust enough to cope with the high variability in clinical acquisitions (MR contrasts, resolutions, orientations, artifacts, and subject populations). Here, we present SynthSeg+, an AI segmentation suite that enables robust analysis of heterogeneous clinical datasets. In addition to whole-brain segmentation, SynthSeg+ also performs cortical parcellation, intracranial volume estimation, and automated detection of faulty segmentations (mainly caused by scans of very low quality). We demonstrate SynthSeg+ in seven experiments, including an aging study on 14,000 scans, where it accurately replicates atrophy patterns observed on data of much higher quality. SynthSeg+ is publicly released as a ready-to-use tool to unlock the potential of quantitative morphometry.


Asunto(s)
Imagen por Resonancia Magnética , Neuroimagen , Imagen por Resonancia Magnética/métodos , Neuroimagen/métodos , Aprendizaje Automático , Encéfalo/diagnóstico por imagen , Algoritmos , Procesamiento de Imagen Asistido por Computador/métodos
14.
Proc Natl Acad Sci U S A ; 120(6): e2211613120, 2023 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-36716365

RESUMEN

Despite the great promise that machine learning has offered in many fields of medicine, it has also raised concerns about potential biases and poor generalization across genders, age distributions, races and ethnicities, hospitals, and data acquisition equipment and protocols. In the current study, and in the context of three brain diseases, we provide evidence which suggests that when properly trained, machine learning models can generalize well across diverse conditions and do not necessarily suffer from bias. Specifically, by using multistudy magnetic resonance imaging consortia for diagnosing Alzheimer's disease, schizophrenia, and autism spectrum disorder, we find that well-trained models have a high area-under-the-curve (AUC) on subjects across different subgroups pertaining to attributes such as gender, age, racial groups and different clinical studies and are unbiased under multiple fairness metrics such as demographic parity difference, equalized odds difference, equal opportunity difference, etc. We find that models that incorporate multisource data from demographic, clinical, genetic factors, and cognitive scores are also unbiased. These models have a better predictive AUC across subgroups than those trained only with imaging features, but there are also situations when these additional features do not help.


Asunto(s)
Enfermedad de Alzheimer , Trastorno del Espectro Autista , Humanos , Masculino , Femenino , Trastorno del Espectro Autista/diagnóstico por imagen , Neuroimagen/métodos , Aprendizaje Automático , Imagen por Resonancia Magnética/métodos , Enfermedad de Alzheimer/diagnóstico por imagen , Enfermedad de Alzheimer/genética , Sesgo
15.
Proc Natl Acad Sci U S A ; 120(50): e2310131120, 2023 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-38048460

RESUMEN

Optical three-dimensional (3D) molecular imaging is highly desirable for providing precise distribution of the target-of-interest in disease models. However, such 3D imaging is still far from wide applications in biomedical research; 3D brain optical molecular imaging, in particular, has rarely been reported. In this report, we designed chemiluminescence probes with high quantum yields, relatively long emission wavelengths, and high signal-to-noise ratios to fulfill the requirements for 3D brain imaging in vivo. With assistance from density-function theory (DFT) computation, we designed ADLumin-Xs by locking up the rotation of the double bond via fusing the furan ring to the phenyl ring. Our results showed that ADLumin-5 had a high quantum yield of chemiluminescence and could bind to amyloid beta (Aß). Remarkably, ADLumin-5's radiance intensity in brain areas could reach 4 × 107 photon/s/cm2/sr, which is probably 100-fold higher than most chemiluminescence probes for in vivo imaging. Because of its strong emission, we demonstrated that ADLumin-5 could be used for in vivo 3D brain imaging in transgenic mouse models of Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer , Ratones , Animales , Enfermedad de Alzheimer/diagnóstico por imagen , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Luminiscencia , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Ratones Transgénicos , Neuroimagen/métodos , Placa Amiloide/metabolismo , Modelos Animales de Enfermedad
16.
J Neurosci ; 44(17)2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38438256

RESUMEN

Recognizing faces regardless of their viewpoint is critical for social interactions. Traditional theories hold that view-selective early visual representations gradually become tolerant to viewpoint changes along the ventral visual hierarchy. Newer theories, based on single-neuron monkey electrophysiological recordings, suggest a three-stage architecture including an intermediate face-selective patch abruptly achieving invariance to mirror-symmetric face views. Human studies combining neuroimaging and multivariate pattern analysis (MVPA) have provided convergent evidence of view selectivity in early visual areas. However, contradictory conclusions have been reached concerning the existence in humans of a mirror-symmetric representation like that observed in macaques. We believe these contradictions arise from low-level stimulus confounds and data analysis choices. To probe for low-level confounds, we analyzed images from two face databases. Analyses of image luminance and contrast revealed biases across face views described by even polynomials-i.e., mirror-symmetric. To explain major trends across neuroimaging studies, we constructed a network model incorporating three constraints: cortical magnification, convergent feedforward projections, and interhemispheric connections. Given the identified low-level biases, we show that a gradual increase of interhemispheric connections across network-layers is sufficient to replicate view-tuning in early processing stages and mirror-symmetry in later stages. Data analysis decisions-pattern dissimilarity measure and data recentering-accounted for the inconsistent observation of mirror-symmetry across prior studies. Pattern analyses of human fMRI data (of either sex) revealed biases compatible with our model. The model provides a unifying explanation of MVPA studies of viewpoint selectivity and suggests observations of mirror-symmetry originate from ineffectively normalized signal imbalances across different face views.


Asunto(s)
Reconocimiento Facial , Humanos , Masculino , Femenino , Reconocimiento Facial/fisiología , Adulto , Neuroimagen/métodos , Estimulación Luminosa/métodos , Modelos Neurológicos , Corteza Visual/fisiología , Corteza Visual/diagnóstico por imagen , Imagen por Resonancia Magnética/métodos , Adulto Joven
17.
Brief Bioinform ; 24(2)2023 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-36882008

RESUMEN

MOTIVATION: With the rapid development of modern technologies, massive data are available for the systematic study of Alzheimer's disease (AD). Though many existing AD studies mainly focus on single-modality omics data, multi-omics datasets can provide a more comprehensive understanding of AD. To bridge this gap, we proposed a novel structural Bayesian factor analysis framework (SBFA) to extract the information shared by multi-omics data through the aggregation of genotyping data, gene expression data, neuroimaging phenotypes and prior biological network knowledge. Our approach can extract common information shared by different modalities and encourage biologically related features to be selected, guiding future AD research in a biologically meaningful way. METHOD: Our SBFA model decomposes the mean parameters of the data into a sparse factor loading matrix and a factor matrix, where the factor matrix represents the common information extracted from multi-omics and imaging data. Our framework is designed to incorporate prior biological network information. Our simulation study demonstrated that our proposed SBFA framework could achieve the best performance compared with the other state-of-the-art factor-analysis-based integrative analysis methods. RESULTS: We apply our proposed SBFA model together with several state-of-the-art factor analysis models to extract the latent common information from genotyping, gene expression and brain imaging data simultaneously from the ADNI biobank database. The latent information is then used to predict the functional activities questionnaire score, an important measurement for diagnosis of AD quantifying subjects' abilities in daily life. Our SBFA model shows the best prediction performance compared with the other factor analysis models. AVAILABILITY: Code are publicly available at https://github.com/JingxuanBao/SBFA. CONTACT: qlong@upenn.edu.


Asunto(s)
Multiómica , Neuroimagen , Teorema de Bayes , Neuroimagen/métodos , Encéfalo/diagnóstico por imagen , Fenotipo
18.
Brief Bioinform ; 24(2)2023 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-36847697

RESUMEN

Brain imaging genomics is an emerging interdisciplinary field, where integrated analysis of multimodal medical image-derived phenotypes (IDPs) and multi-omics data, bridging the gap between macroscopic brain phenotypes and their cellular and molecular characteristics. This approach aims to better interpret the genetic architecture and molecular mechanisms associated with brain structure, function and clinical outcomes. More recently, the availability of large-scale imaging and multi-omics datasets from the human brain has afforded the opportunity to the discovering of common genetic variants contributing to the structural and functional IDPs of the human brain. By integrative analyses with functional multi-omics data from the human brain, a set of critical genes, functional genomic regions and neuronal cell types have been identified as significantly associated with brain IDPs. Here, we review the recent advances in the methods and applications of multi-omics integration in brain imaging analysis. We highlight the importance of functional genomic datasets in understanding the biological functions of the identified genes and cell types that are associated with brain IDPs. Moreover, we summarize well-known neuroimaging genetics datasets and discuss challenges and future directions in this field.


Asunto(s)
Encéfalo , Genómica , Humanos , Genómica/métodos , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Fenotipo , Neuroimagen/métodos
19.
Ann Neurol ; 95(6): 1017-1034, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38606939

RESUMEN

Stroke is the chief differential diagnosis in patient presenting to the emergency room with abrupt onset focal neurological deficits. Neuroimaging, including non-contrast computed tomography (CT), magnetic resonance imaging (MRI), vascular and perfusion imaging, is a cornerstone in the diagnosis and treatment decision-making. This review examines the current state of evidence behind the different imaging paradigms for acute ischemic stroke diagnosis and treatment, including current recommendations from the guidelines. Non-contrast CT brain, or in some centers MRI, can help differentiate ischemic stroke and intracerebral hemorrhage (ICH), a pivotal juncture in stroke diagnosis and treatment algorithm, especially for early window thrombolytics. Advanced imaging such as MRI or perfusion imaging can also assist making a diagnosis of ischemic stroke versus mimics such as migraine, Todd's paresis, or functional disorders. Identification of medium-large vessel occlusions with CT or MR angiography triggers consideration of endovascular thrombectomy (EVT), with additional perfusion imaging help identify salvageable brain tissue in patients who are likely to benefit from reperfusion therapies, particularly in the ≥6 h window. We also review recent advances in neuroimaging and ongoing trials in key therapeutic areas and their imaging selection criteria to inform the readers on potential future transitions into use of neuroimaging for stroke diagnosis and treatment decision making. ANN NEUROL 2024;95:1017-1034.


Asunto(s)
Accidente Cerebrovascular Isquémico , Neuroimagen , Humanos , Neuroimagen/métodos , Accidente Cerebrovascular Isquémico/diagnóstico por imagen , Accidente Cerebrovascular Isquémico/terapia
20.
Mol Psychiatry ; 29(5): 1465-1477, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38332374

RESUMEN

Machine learning approaches using structural magnetic resonance imaging (sMRI) can be informative for disease classification, although their ability to predict psychosis is largely unknown. We created a model with individuals at CHR who developed psychosis later (CHR-PS+) from healthy controls (HCs) that can differentiate each other. We also evaluated whether we could distinguish CHR-PS+ individuals from those who did not develop psychosis later (CHR-PS-) and those with uncertain follow-up status (CHR-UNK). T1-weighted structural brain MRI scans from 1165 individuals at CHR (CHR-PS+, n = 144; CHR-PS-, n = 793; and CHR-UNK, n = 228), and 1029 HCs, were obtained from 21 sites. We used ComBat to harmonize measures of subcortical volume, cortical thickness and surface area data and corrected for non-linear effects of age and sex using a general additive model. CHR-PS+ (n = 120) and HC (n = 799) data from 20 sites served as a training dataset, which we used to build a classifier. The remaining samples were used external validation datasets to evaluate classifier performance (test, independent confirmatory, and independent group [CHR-PS- and CHR-UNK] datasets). The accuracy of the classifier on the training and independent confirmatory datasets was 85% and 73% respectively. Regional cortical surface area measures-including those from the right superior frontal, right superior temporal, and bilateral insular cortices strongly contributed to classifying CHR-PS+ from HC. CHR-PS- and CHR-UNK individuals were more likely to be classified as HC compared to CHR-PS+ (classification rate to HC: CHR-PS+, 30%; CHR-PS-, 73%; CHR-UNK, 80%). We used multisite sMRI to train a classifier to predict psychosis onset in CHR individuals, and it showed promise predicting CHR-PS+ in an independent sample. The results suggest that when considering adolescent brain development, baseline MRI scans for CHR individuals may be helpful to identify their prognosis. Future prospective studies are required about whether the classifier could be actually helpful in the clinical settings.


Asunto(s)
Encéfalo , Aprendizaje Automático , Imagen por Resonancia Magnética , Neuroimagen , Trastornos Psicóticos , Humanos , Trastornos Psicóticos/patología , Trastornos Psicóticos/diagnóstico por imagen , Imagen por Resonancia Magnética/métodos , Masculino , Femenino , Encéfalo/patología , Encéfalo/diagnóstico por imagen , Neuroimagen/métodos , Adulto , Adulto Joven , Adolescente , Síntomas Prodrómicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA