Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
Add more filters

Publication year range
1.
Mol Psychiatry ; 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39107583

ABSTRACT

Hemispheric brain asymmetry is a basic organizational principle of the human brain and has been implicated in various psychiatric conditions, including autism spectrum disorder. Brain asymmetry is not a uniquely human feature and is observed in other species such as the mouse. Yet, asymmetry patterns are generally nuanced, and substantial sample sizes are required to detect these patterns. In this pre-registered study, we use a mouse dataset from the Province of Ontario Neurodevelopmental Network, which comprises structural MRI data from over 2000 mice, including genetic models for autism spectrum disorder, to reveal the scope and magnitude of hemispheric asymmetry in the mouse. Our findings demonstrate the presence of robust hemispheric asymmetry in the mouse brain, such as larger right hemispheric volumes towards the anterior pole and larger left hemispheric volumes toward the posterior pole, opposite to what has been shown in humans. This suggests the existence of species-specific traits. Further clustering analysis identified distinct asymmetry patterns in autism spectrum disorder models, a phenomenon that is also seen in atypically developing participants. Our study shows potential for the use of mouse models to understand the biological bases of typical and atypical brain asymmetry but also warrants caution as asymmetry patterns seem to differ between humans and mice.

2.
Cereb Cortex ; 34(1)2024 01 14.
Article in English | MEDLINE | ID: mdl-38100331

ABSTRACT

Imaging awake animals is quickly gaining traction in neuroscience as it offers a means to eliminate the confounding effects of anesthesia, difficulties of inter-species translation (when humans are typically imaged while awake), and the inability to investigate the full range of brain and behavioral states in unconscious animals. In this systematic review, we focus on the development of awake mouse blood oxygen level dependent functional magnetic resonance imaging (fMRI). Mice are widely used in research due to their fast-breeding cycle, genetic malleability, and low cost. Functional MRI yields whole-brain coverage and can be performed on both humans and animal models making it an ideal modality for comparing study findings across species. We provide an analysis of 30 articles (years 2011-2022) identified through a systematic literature search. Our conclusions include that head-posts are favorable, acclimation training for 10-14 d is likely ample under certain conditions, stress has been poorly characterized, and more standardization is needed to accelerate progress. For context, an overview of awake rat fMRI studies is also included. We make recommendations that will benefit a wide range of neuroscience applications.


Subject(s)
Anesthesia , Magnetic Resonance Imaging , Humans , Mice , Rats , Animals , Magnetic Resonance Imaging/methods , Wakefulness , Brain/diagnostic imaging , Brain Mapping
3.
Mol Psychiatry ; 27(2): 865-872, 2022 02.
Article in English | MEDLINE | ID: mdl-34650202

ABSTRACT

The triple-network model of psychopathology is a framework to explain the functional and structural neuroimaging phenotypes of psychiatric and neurological disorders. It describes the interactions within and between three distributed networks: the salience, default-mode, and central executive networks. These have been associated with brain disorder traits in patients. Homologous networks have been proposed in animal models, but their integration into a triple-network organization has not yet been determined. Using resting-state datasets, we demonstrate conserved spatio-temporal properties between triple-network elements in human, macaque, and mouse. The model predictions were also shown to apply in a mouse model for depression. To validate spatial homologies, we developed a data-driven approach to convert mouse brain maps into human standard coordinates. Finally, using high-resolution viral tracers in the mouse, we refined an anatomical model for these networks and validated this using optogenetics in mice and tractography in humans. Unexpectedly, we find serotonin involvement within the salience rather than the default-mode network. Our results support the existence of a triple-network system in the mouse that shares properties with that of humans along several dimensions, including a disease condition. Finally, we demonstrate a method to humanize mouse brain networks that opens doors to fully data-driven trans-species comparisons.


Subject(s)
Magnetic Resonance Imaging , Nerve Net , Animals , Brain , Brain Mapping/methods , Humans , Magnetic Resonance Imaging/methods , Mice , Neural Pathways
4.
Neuroimage ; 225: 117528, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33157264

ABSTRACT

Understanding cortical organization is a fundamental goal of neuroscience that requires comparisons across species and modalities. Large-scale connectivity gradients have recently been introduced as a data-driven representation of the intrinsic organization of the cortex. We studied resting-state functional connectivity gradients in the mouse cortex and found robust spatial patterns across four data sets. The principal gradient of functional connectivity shows a striking overlap with an axis of neocortical evolution from two primordial origins. Additional gradients reflect sensory specialization and aspects of a sensory-to-transmodal hierarchy, and are associated with transcriptomic features. While some of these gradients strongly resemble observations in the human cortex, the overall pattern in the mouse cortex emphasizes the specialization of sensory areas over a global functional hierarchy.


Subject(s)
Biological Evolution , Neocortex/diagnostic imaging , Neural Pathways/diagnostic imaging , Animals , Brain Mapping , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/physiology , Connectome , Functional Neuroimaging , Magnetic Resonance Imaging , Mice , Mice, Inbred C57BL , Neocortex/physiology , Neural Pathways/physiology , Rest
5.
Neuroimage ; 225: 117456, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33069863

ABSTRACT

Hallucinogenic agents have been proposed as potent antidepressants; this includes the serotonin (5-HT) receptor 2A agonist psilocybin. In human subjects, psilocybin alters functional connectivity (FC) within the default-mode network (DMN), a constellation of inter-connected regions that displays altered FC in depressive disorders. In this study, we investigated the effects of psilocybin on FC across the entire brain with a view to investigate underlying mechanisms. Psilocybin effects were investigated in lightly-anaesthetized mice using resting-state fMRI. Dual-regression analysis identified reduced FC within the ventral striatum in psilocybin- relative to vehicle-treated mice. Refinement of the analysis using spatial references derived from both gene expression maps and viral tracer projection fields revealed two distinct effects of psilocybin: it increased FC between 5-HT-associated networks and cortical areas, including elements of the murine DMN, thalamus, and midbrain; it decreased FC within dopamine (DA)-associated striatal networks. These results suggest that interactions between 5-HT- and DA-regulated neural networks contribute to the neural and therefore psychological effects of psilocybin. Furthermore, they highlight how information on molecular expression patterns and structural connectivity can assist in the interpretation of pharmaco-fMRI findings.


Subject(s)
Brain/drug effects , Default Mode Network/drug effects , Psilocybin/pharmacology , Serotonin 5-HT2 Receptor Agonists/pharmacology , Animals , Brain/diagnostic imaging , Brain/metabolism , Corpus Striatum/diagnostic imaging , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Default Mode Network/diagnostic imaging , Default Mode Network/metabolism , Dopamine/metabolism , Functional Neuroimaging , Magnetic Resonance Imaging , Mesencephalon/diagnostic imaging , Mesencephalon/drug effects , Mesencephalon/metabolism , Mice , Neural Pathways/diagnostic imaging , Neural Pathways/drug effects , Neural Pathways/metabolism , Rest , Serotonin/metabolism , Thalamus/diagnostic imaging , Thalamus/drug effects , Thalamus/metabolism
6.
Int J Mol Sci ; 22(9)2021 May 10.
Article in English | MEDLINE | ID: mdl-34068707

ABSTRACT

BDNF plays a pivotal role in neuroplasticity events, vulnerability and resilience to stress-related disorders, being decreased in depressive patients and increased after antidepressant treatment. BDNF was found to be reduced in patients carrying the human polymorphism in the serotonin transporter promoter region (5-HTTLPR). The serotonin knockout rat (SERT-/-) is one of the animal models used to investigate the underlying molecular mechanisms of depression in humans. They present decreased BDNF levels, and anxiety- and depression-like behavior. To investigate whether upregulating BDNF would ameliorate the phenotype of SERT-/- rats, we overexpressed BDNF locally into the ventral hippocampus and submitted the animals to behavioral testing. The results showed that BDNF overexpression in the vHIP of SERT-/- rats promoted higher sucrose preference and sucrose intake; on the first day of the sucrose consumption test it decreased immobility time in the forced swim test and increased the time spent in the center of a novel environment. Furthermore, BDNF overexpression altered social behavior in SERT-/- rats, which presented increased passive contact with test partner and decreased solitary behavior. Finally, it promoted decrease in plasma corticosterone levels 60 min after restraint stress. In conclusion, modulation of BDNF IV levels in the vHIP of SERT-/- rats led to a positive behavioral outcome placing BDNF upregulation in the vHIP as a potential target to new therapeutic approaches to improve depressive symptoms.


Subject(s)
Anxiety Disorders/drug therapy , Brain-Derived Neurotrophic Factor/genetics , Serotonin Plasma Membrane Transport Proteins/genetics , Stress, Psychological/drug therapy , Animals , Anti-Anxiety Agents/pharmacology , Antidepressive Agents/pharmacology , Anxiety Disorders/genetics , Anxiety Disorders/pathology , Corticosterone/genetics , Disease Models, Animal , Gene Expression Regulation , Gene Knockout Techniques , Hippocampus/metabolism , Hippocampus/pathology , Humans , Male , Neuronal Plasticity/genetics , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/genetics , Rats , Serotonin/genetics , Stress, Psychological/genetics , Stress, Psychological/pathology
7.
Neuroimage ; 205: 116278, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31614221

ABSTRACT

Preclinical applications of resting-state functional magnetic resonance imaging (rsfMRI) offer the possibility to non-invasively probe whole-brain network dynamics and to investigate the determinants of altered network signatures observed in human studies. Mouse rsfMRI has been increasingly adopted by numerous laboratories worldwide. Here we describe a multi-centre comparison of 17 mouse rsfMRI datasets via a common image processing and analysis pipeline. Despite prominent cross-laboratory differences in equipment and imaging procedures, we report the reproducible identification of several large-scale resting-state networks (RSN), including a mouse default-mode network, in the majority of datasets. A combination of factors was associated with enhanced reproducibility in functional connectivity parameter estimation, including animal handling procedures and equipment performance. RSN spatial specificity was enhanced in datasets acquired at higher field strength, with cryoprobes, in ventilated animals, and under medetomidine-isoflurane combination sedation. Our work describes a set of representative RSNs in the mouse brain and highlights key experimental parameters that can critically guide the design and analysis of future rodent rsfMRI investigations.


Subject(s)
Brain/physiology , Connectome/methods , Image Processing, Computer-Assisted/methods , Magnetic Resonance Imaging/methods , Nerve Net/physiology , Animals , Brain/diagnostic imaging , Connectome/standards , Female , Image Processing, Computer-Assisted/standards , Magnetic Resonance Imaging/standards , Male , Mice , Mice, Inbred C57BL , Nerve Net/diagnostic imaging , Reproducibility of Results
8.
J Neurosci ; 37(34): 8092-8101, 2017 08 23.
Article in English | MEDLINE | ID: mdl-28716961

ABSTRACT

Translational neuroimaging requires approaches and techniques that can bridge between multiple different species and disease states. One candidate method that offers insights into the brain's functional connectivity (FC) is resting-state fMRI (rs-fMRI). In both humans and nonhuman primates, patterns of FC (often referred to as the functional connectome) have been related to the underlying structural connectivity (SC; also called the structural connectome). Given the recent rise in preclinical neuroimaging of mouse models, it is an important question whether the mouse functional connectome conforms to the underlying SC. Here, we compared FC derived from rs-fMRI in female mice with the underlying monosynaptic structural connectome as provided by the Allen Brain Connectivity Atlas. We show that FC between interhemispheric homotopic cortical and hippocampal areas, as well as in cortico-striatal pathways, emerges primarily via monosynaptic structural connections. In particular, we demonstrate that the striatum (STR) can be segregated according to differential rs-fMRI connectivity patterns that mirror monosynaptic connectivity with isocortex. In contrast, for certain subcortical networks, FC emerges along polysynaptic pathways as shown for left and right STR, which do not share direct anatomical connections, but high FC is putatively driven by a top-down cortical control. Finally, we show that FC involving cortico-thalamic pathways is limited, possibly confounded by the effect of anesthesia, small regional size, and tracer injection volume. These findings provide a critical foundation for using rs-fMRI connectivity as a translational tool to study complex brain circuitry interactions and their pathology due to neurological or psychiatric diseases across species.SIGNIFICANCE STATEMENT A comprehensive understanding of how the anatomical architecture of the brain, often referred to as the "connectome," corresponds to its function is arguably one of the biggest challenges for understanding the brain and its pathologies. Here, we use the mouse as a model for comparing functional connectivity (FC) derived from resting-state fMRI with gold standard structural connectivity measures based on tracer injections. In particular, we demonstrate high correspondence between FC measurements of cortico-cortical and cortico-striatal regions and their anatomical underpinnings. This work provides a critical foundation for studying the pathology of these circuits across mouse models and human patients.


Subject(s)
Brain/physiology , Connectome/methods , Magnetic Resonance Imaging/methods , Nerve Net/physiology , Animals , Female , Mice , Mice, Inbred C57BL
9.
Neuroimage ; 149: 190-199, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28159688

ABSTRACT

Studies in mice using resting-state functional magnetic resonance imaging (rs-fMRI) have provided opportunities to investigate the effects of pharmacological manipulations on brain function and map the phenotypes of mouse models of human brain disorders. Mouse rs-fMRI is typically performed under anaesthesia, which induces both regional suppression of brain activity and disruption of large-scale neural networks. Previous comparative studies using rodents investigating various drug effects on long-distance functional connectivity (FC) have reported agent-specific FC patterns, however, effects of regional suppression are sparsely explored. Here we examined changes in regional connectivity under six different anaesthesia conditions using mouse rs-fMRI with the goal of refining the framework of understanding the brain activation under anaesthesia at a local level. Regional homogeneity (ReHo) was used to map local synchronization in the brain, followed by analysis of several brain areas based on ReHo maps. The results revealed high local coherence in most brain areas. The primary somatosensory cortex and caudate-putamen showed agent-specific properties. Lower local coherence in the cingulate cortex was observed under medetomidine, particularly when compared to the combination of medetomidine and isoflurane. The thalamus was associated with retained local coherence across anaesthetic levels and multiple nuclei. These results show that anaesthesia induced by the investigated anaesthetics through different molecular targets promote agent-specific regional connectivity. In addition, ReHo is a data-driven method with minimum user interaction, easy to use and fast to compute. Given that examination of the brain at a local level is widely applied in human rs-fMRI studies, our results show its sensitivity to extract information on varied neuronal activity under six different regimens relevant to mouse functional imaging. These results, therefore, will inform future rs-fMRI studies on mice and the type of anaesthetic agent used, and will help to bridge observations between this burgeoning research field and ongoing human research across analytical scales.


Subject(s)
Anesthetics/pharmacology , Brain Mapping/methods , Brain/drug effects , Neural Pathways/drug effects , Animals , Female , Image Processing, Computer-Assisted , Isoflurane/pharmacology , Magnetic Resonance Imaging , Medetomidine/pharmacology , Mice , Mice, Inbred C57BL , Propofol/pharmacology , Rest , Urethane/pharmacology
10.
Neuroimage ; 152: 497-508, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28315459

ABSTRACT

Functional connectivity (FC) derived from resting-state functional magnetic resonance imaging (rs-fMRI) allows for the integrative study of neuronal processes at a macroscopic level. The majority of studies to date have assumed stationary interactions between brain regions, without considering the dynamic aspects of network organization. Only recently has the latter received increased attention, predominantly in human studies. Applying dynamic FC (dFC) analysis to mice is attractive given the relative simplicity of the mouse brain and the possibility to explore mechanisms underlying network dynamics using pharmacological, environmental or genetic interventions. Therefore, we have evaluated the feasibility and research potential of mouse dFC using the interventions of social stress or anesthesia duration as two case-study examples. By combining a sliding-window correlation approach with dictionary learning, several dynamic functional states (dFS) with a complex organization were identified, exhibiting highly dynamic inter- and intra-modular interactions. Each dFS displayed a high degree of reproducibility upon changes in analytical parameters and across datasets. They fluctuated at different degrees as a function of anesthetic depth, and were sensitive indicators of pathology as shown for the chronic psychosocial stress mouse model of depression. Dynamic functional states are proposed to make a major contribution to information integration and processing in the healthy and diseased brain.


Subject(s)
Brain Mapping/methods , Brain/physiology , Social Behavior , Stress, Psychological/physiopathology , Anesthetics, Inhalation/administration & dosage , Animals , Brain/drug effects , Female , Image Processing, Computer-Assisted , Isoflurane/administration & dosage , Magnetic Resonance Imaging , Male , Mice, Inbred C57BL , Neural Pathways/drug effects , Neural Pathways/physiology
11.
Neuroimage ; 134: 1-11, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27033685

ABSTRACT

Alzheimer's disease is a fatal neurodegenerative disorder affecting the aging population. Neuroimaging methods, in particular magnetic resonance imaging (MRI), have helped reveal alterations in the brain structure, metabolism, and function of patients and in groups at risk of developing AD, yet the nature of these alterations is poorly understood. Neuroimaging in mice is attractive for investigating mechanisms underlying functional and structural changes associated with AD pathology. Several preclinical murine models of AD have been generated based on transgenic insertion of human mutated APP genes. Depending on the specific mutations, mouse strains express different aspects of amyloid pathology, e.g. intracellular amyloid-ß (Aß) aggregates, parenchymal plaques, or cerebral amyloid angiopathy. We have applied multi-parametric MRI in three transgenic mouse lines to compare changes in brain function with resting-state fMRI and structure with diffusion tensor imaging and high resolution anatomical imaging. E22ΔAß developing intracellular Aß aggregates did not present functional or structural alterations compared to their wild-type littermates. PSAPP mice displaying parenchymal amyloid plaques displayed mild functional changes within the supplementary and barrel field cortices, and increased isocortical volume relative to controls. Extensive reduction in functional connectivity in the sensory-motor cortices and within the default mode network, as well as local volume increase in the midbrain relative to wild-type have been observed in ArcAß mice bearing intracellular Aß aggregates as well as parenchymal and vascular amyloid deposits. Patterns of functional and structural changes appear to be strain-specific and not directly related to amyloid deposition.


Subject(s)
Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Brain/pathology , Brain/physiopathology , Disease Models, Animal , Magnetic Resonance Imaging/methods , Multimodal Imaging/methods , Alzheimer Disease/diagnostic imaging , Amyloid beta-Protein Precursor/genetics , Amyloidosis/pathology , Amyloidosis/physiopathology , Animals , Brain/diagnostic imaging , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Reproducibility of Results , Sensitivity and Specificity
12.
Neuroimage ; 142: 544-552, 2016 Nov 15.
Article in English | MEDLINE | ID: mdl-27520750

ABSTRACT

Human depression, for which chronic psychosocial stress is a major risk factor, is characterized by consistent alterations in neurocircuitry. For example, there is increased functional connectivity (FC) within and between regions comprising the default mode network (DMN) including prefrontal cortex and cingulate cortex. Alterations in network FC are associated with specific aspects of psychopathology. In mice, chronic psychosocial stress (CPS) leads to depression-relevant behavior, including increased fear learning, learned helplessness, fatigue and decreased motivation for reward. Using multimodal in vivo magnetic resonance imaging (MRI) and spectroscopy (MRS), we investigated CPS effects on function and structure in the mouse brain under light anesthesia. Mice underwent a baseline MRI/MRS session, followed by 15-day CPS (n=26) or control handling (n=27), and a post-treatment MRI/MRS session. In BOLD fMRI, relative to controls, CPS mice exhibited robust, reproducible increases in FC within 8 of 9 identified cortical networks, including the prefrontal and cingulate cortices that contribute to the "mouse DMN". CPS mice exhibited increases in between-network FC, including amygdala - prefrontal cortex and amygdala - cingulate cortex. MRS identified metabolic alterations in CPS mice as increased inositol levels in amygdala and increased glycerophosphorylcholine levels in prefrontal cortex. Diffusion-weighted MRI detected increased fractional anisotropic values in the cingulum. This study demonstrates that chronic psychosocial stress induces FC states in the mouse brain analogous to those observed in depression, as well as cerebral metabolism and white matter pathway alterations that contribute to understanding of pathological processes. It also demonstrates the importance of brain imaging to the establishment of valid animal models in translational psychiatry.


Subject(s)
Amygdala , Behavior, Animal/physiology , Connectome/methods , Depression , Gyrus Cinguli , Magnetic Resonance Imaging/methods , Prefrontal Cortex , Stress, Psychological , Amygdala/diagnostic imaging , Amygdala/metabolism , Amygdala/physiopathology , Animals , Depression/diagnostic imaging , Depression/etiology , Depression/metabolism , Depression/physiopathology , Diffusion Tensor Imaging/methods , Disease Models, Animal , Glycerylphosphorylcholine/metabolism , Gyrus Cinguli/diagnostic imaging , Gyrus Cinguli/metabolism , Gyrus Cinguli/physiopathology , Inositol/metabolism , Magnetic Resonance Spectroscopy/methods , Male , Mice , Mice, Inbred C57BL , Prefrontal Cortex/diagnostic imaging , Prefrontal Cortex/metabolism , Prefrontal Cortex/physiopathology , Stress, Psychological/complications , Stress, Psychological/diagnostic imaging , Stress, Psychological/metabolism , Stress, Psychological/physiopathology
13.
J Neurosci ; 34(41): 13780-9, 2014 Oct 08.
Article in English | MEDLINE | ID: mdl-25297104

ABSTRACT

Impairment of brain functional connectivity (FC) is thought to be an early event occurring in diseases with cerebral amyloidosis, such as Alzheimer's disease. Regions sustaining altered functional networks have been shown to colocalize with regions marked with amyloid plaques burden suggesting a strong link between FC and amyloidosis. Whether the decline in FC precedes amyloid plaque deposition or is a consequence thereof is currently unknown. The sequence of events during early stages of the disease is difficult to capture in humans due to the difficulties in providing an early diagnosis and also in view of the heterogeneity among patients. Transgenic mouse lines overexpressing amyloid precursor proteins develop cerebral amyloidosis and constitute an attractive model system for studying the relationship between plaque and functional changes. In this study, ArcAß transgenic and wild-type mice were imaged using resting-state fMRI methods across their life-span in a cross-sectional design to analyze changes in FC in relation to the pathology. Transgenic mice show compromised development of FC during the first months of postnatal life compared with wild-type animals, resulting in functional impairments that affect in particular the sensory-motor cortex already in preplaque stage. These functional alterations were accompanied by structural changes as reflected by reduced fractional anisotropy values, as derived from diffusion tensor imaging. Our results suggest cerebral amyloidosis in mice is preceded by impairment of neuronal networks and white matter structures. FC analysis in mice is an attractive tool for studying the implications of impaired neuronal networks in models of cerebral amyloid pathology.


Subject(s)
Amyloidosis/pathology , Neural Pathways/physiology , White Matter/anatomy & histology , Aging/physiology , Alzheimer Disease/pathology , Amyloidosis/genetics , Animals , Anisotropy , Female , Genotype , Humans , Male , Mice , Mice, Transgenic , Nerve Net/pathology , White Matter/growth & development
14.
Neuroimage ; 123: 11-21, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26296501

ABSTRACT

The use of resting state fMRI (rs-fMRI) in translational research is a powerful tool to assess brain connectivity and investigate neuropathology in mouse models. However, despite encouraging initial results, the characterization of consistent and robust resting state networks in mice remains a methodological challenge. One key reason is that the quality of the measured MR signal is degraded by the presence of structural noise from non-neural sources. Notably, in the current pipeline of the Human Connectome Project, a novel approach has been introduced to clean rs-fMRI data, which involves automatic artifact component classification and data cleaning (FIX). FIX does not require any external recordings of physiology or the segmentation of CSF and white matter. In this study, we evaluated the performance of FIX for analyzing mouse rs-fMRI data. Our results showed that FIX can be easily applied to mouse datasets and detects true signals with 100% accuracy and true noise components with very high accuracy (>98%), thus reducing both within- and between-subject variability of rs-fMRI connectivity measurements. Using this improved pre-processing pipeline, maps of 23 resting state circuits in mice were identified including two networks that displayed default mode network-like topography. Hierarchical clustering grouped these neural networks into meaningful larger functional circuits. These mouse resting state networks, which are publicly available, might serve as a reference for future work using mouse models of neurological disorders.


Subject(s)
Artifacts , Brain Mapping/methods , Brain/physiology , Magnetic Resonance Imaging/methods , Signal Processing, Computer-Assisted , Algorithms , Animals , Cluster Analysis , Image Processing, Computer-Assisted , Male , Mice , Mice, Inbred C57BL , Neural Pathways/physiology
15.
Neuroimage ; 120: 55-63, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26166624

ABSTRACT

Functional proton magnetic resonance spectroscopy (1H-MRS) enables the non-invasive assessment of neural activity by measuring signals arising from endogenous metabolites in a time resolved manner. Proof-of-principle of this approach has been demonstrated in humans and rats; yet functional 1H-MRS has not been applied in mice so far, although it would be of considerable interest given the many genetically engineered models of neurological disorders established in this species only. Mouse 1H-MRS is challenging as the high demands on spatial resolution typically result in long data acquisition times not commensurable with functional studies. Here, we propose an approach based on spectroscopic imaging in combination with the acquisition of the free induction decay to maximize signal intensity. Highly resolved metabolite maps have been recorded from mouse brain with 12 min temporal resolution. This enabled monitoring of metabolic changes following the administration of bicuculline, a GABA-A receptor antagonist. Changes in levels of metabolites involved in energy metabolism (lactate and phosphocreatine) and neurotransmitters (glutamate) were investigated in a region-dependent manner and shown to scale with the bicuculline dose. GABAergic inhibition induced spectral changes characteristic for increased neurotransmitter turnover and oxidative stress. In contrast to metabolic readouts, BOLD and CBV fMRI responses did not scale with the bicuculline dose indicative of the failure of neurovascular coupling. Nevertheless fMRI measurements supported the notion of increased oxidative stress revealed by functional MRS. Hence, the combined analysis of metabolic and hemodynamic changes in response to stimulation provides complementary insight into processes associated with neural activity.


Subject(s)
Brain/metabolism , Epilepsy/metabolism , GABA-A Receptor Antagonists/pharmacology , Magnetic Resonance Imaging/methods , Magnetic Resonance Spectroscopy/methods , Neurovascular Coupling/physiology , Oxidative Stress/physiology , Proton Magnetic Resonance Spectroscopy/methods , Animals , Bicuculline/pharmacology , Brain/physiopathology , Epilepsy/chemically induced , Female , Mice , Mice, Inbred C57BL
16.
Neuroimage ; 94: 372-384, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24495809

ABSTRACT

Functional magnetic resonance (fMRI) in mice has become an attractive tool for mechanistic studies, for characterizing models of human disease, and for evaluation of novel therapies. Yet, controlling the physiological state of mice is challenging, but nevertheless important as changes in cardiovascular parameters might affect the hemodynamic readout which constitutes the basics of the fMRI signal. In contrast to rats, fMRI studies in mice report less robust brain activation of rather widespread character to innocuous sensory stimulation. Anesthesia is known to influence the characteristics of the fMRI signal. To evaluate modulatory effects imposed by the anesthesia on stimulus-evoked fMRI responses, we compared blood oxygenation level dependent (BOLD) and cerebral blood volume (CBV) signal changes to electrical hindpaw stimulation using the four commonly used anesthetics isoflurane, medetomidine, propofol and urethane. fMRI measurements were complemented by assessing systemic physiological parameters throughout the experiment. Unilateral stimulation of the hindpaw elicited widespread fMRI responses in the mouse brain displaying a bilateral pattern irrespective of the anesthetic used. Analysis of magnitude and temporal profile of BOLD and CBV signals indicated anesthesia-specific modulation of cerebral hemodynamic responses and differences observed for the four anesthetics could be largely explained by their known effects on animal physiology. Strikingly, independent of the anesthetic used our results reveal that fMRI responses are influenced by stimulus-induced cardiovascular changes, which indicate an arousal response, even to innocuous stimulation. This may mask specific fMRI signal associated to the stimulus. Hence, studying the processing of peripheral input in mice using fMRI techniques constitutes a major challenge and adapted paradigms and/or alternative fMRI readouts should also be considered when studying sensory processing in mice.


Subject(s)
Anesthetics, General/administration & dosage , Brain Mapping/methods , Evoked Potentials, Somatosensory/drug effects , Evoked Potentials, Somatosensory/physiology , Magnetic Resonance Imaging/methods , Somatosensory Cortex/drug effects , Somatosensory Cortex/physiology , Animals , Dose-Response Relationship, Drug , Female , Isoflurane/administration & dosage , Medetomidine/administration & dosage , Mice , Mice, Inbred C57BL , Physical Stimulation/methods , Propofol/administration & dosage , Reproducibility of Results , Sensitivity and Specificity , Touch , Urethane/administration & dosage
17.
Neuroimage ; 102 Pt 2: 838-47, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25175535

ABSTRACT

Resting state-fMRI (rs-fMRI) in mice allows studying mechanisms underlying functional connectivity (FC) as well as alterations of FC occurring in murine models of neurological diseases. Mouse fMRI experiments are typically carried out under anesthesia to minimize animal movement and potential distress during examination. Yet, anesthesia inevitably affects FC patterns. Such effects have to be understood for proper interpretation of data. We have compared the influence of four commonly used anesthetics on rs-fMRI. Rs-fMRI data acquired under isoflurane, propofol, and urethane presented similar patterns when accounting for anesthesia depth. FC maps displayed bilateral correlation with respect to cortical seeds, but no significant inter-hemispheric striatal connectivity. In contrast, for medetomidine, we detected bilateral striatal but compromised inter-hemispheric cortical connectivity. The spatiotemporal patterns of the rs-fMRI signal have been rationalized considering anesthesia depth and pharmacodynamic properties of the anesthetics. Our results bridge the results from different studies from the burgeoning field of mouse rs-fMRI and offer a framework for understanding the influences of anesthetics on FC patterns. Utilizing this information, we suggest the combined use of medetomidine and isoflurane representing the two proposed classes of anesthetics; the combination of low doses of the two anesthetics retained strong correlations both within cortical and subcortical structures, without the potential seizure-inducing effects of medetomidine, rendering this regimen an attractive anesthesia for rs-fMRI in mice.


Subject(s)
Anesthetics/pharmacology , Brain/drug effects , Brain/physiology , Magnetic Resonance Imaging , Nerve Net/drug effects , Nerve Net/physiology , Anesthesia/standards , Animals , Clinical Protocols , Female , Isoflurane/pharmacology , Medetomidine/pharmacology , Mice , Mice, Inbred C57BL , Propofol/pharmacology , Rest/physiology , Urethane/pharmacology
18.
Mol Imaging ; 132014.
Article in English | MEDLINE | ID: mdl-25248521

ABSTRACT

Intratumoral hypoxia changes the metabolism of gliomas, leading to a more aggressive phenotype with increased resistance to radio- and chemotherapy. Hypoxia triggers a signaling cascade with hypoxia-inducible factor (HIF) as a key regulator. We monitored activation of the HIF pathway longitudinally in murine glioma tumors. GL261 cells, stably transfected with a luciferase reporter driven under the control of a promoter comprising the HIF target gene motive hypoxia response element, were implanted either subcutaneously or orthotopically. In vivo experiments were carried out using bioluminescence imaging. Tumors were subsequently analyzed using immunofluorescence staining for hypoxia, endothelial cells, tumor perfusion, and glucose transporter expression. Transient upregulation of the HIF signaling was observed in both subcutaneous and orthotopic gliomas. Immunofluorescence staining confirmed hypoxic regions in subcutaneous and, to a lesser extent, intracranial tumors. Subcutaneous tumors showed substantial necrosis, which might contribute to the decreased bioluminescence output observed toward the end of the experiment. Orthotopic tumors were less hypoxic than subcutaneous ones and did not develop extensive necrotic areas. Although this may be the result of the overall smaller size of orthotopic tumors, it might also reflect differences in the local environment, such as the better intrinsic vascularization of brain tissue compared to the subcutaneous tissue compartment.


Subject(s)
Glioma/metabolism , Glioma/pathology , Luminescent Measurements/methods , Magnetic Resonance Imaging/methods , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Hypoxia , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Genes, Reporter , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Luciferases/metabolism , Luminescent Agents/metabolism , Mice , Mice, Nude , Neoplasm Transplantation , Signal Transduction
19.
eNeuro ; 11(3)2024 Mar.
Article in English | MEDLINE | ID: mdl-38499355

ABSTRACT

Fueled by the recent and controversial brain-wide association studies in humans, the animal neuroimaging community has also begun questioning whether using larger sample sizes is necessary for ethical and effective scientific progress. In this opinion piece, we illustrate two opposing views on sample size extremes in MRI-based animal neuroimaging.


Subject(s)
Magnetic Resonance Imaging , Neuroimaging , Animals , Humans , Neuroimaging/methods , Magnetic Resonance Imaging/methods , Brain/diagnostic imaging
20.
Nat Commun ; 15(1): 6708, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39112455

ABSTRACT

Functional magnetic resonance imaging in rodents holds great potential for advancing our understanding of brain networks. Unlike the human community, there remains no standardized resource in rodents for image processing, analysis and quality control, posing significant reproducibility limitations. Our software platform, Rodent Automated Bold Improvement of EPI Sequences, is a pipeline designed to address these limitations for preprocessing, quality control, and confound correction, along with best practices for reproducibility and transparency. We demonstrate the robustness of the preprocessing workflow by validating performance across multiple acquisition sites and both mouse and rat data. Building upon a thorough investigation into data quality metrics across acquisition sites, we introduce guidelines for the quality control of network analysis and offer recommendations for addressing issues. Taken together, this software platform will allow the emerging community to adopt reproducible practices and foster progress in translational neuroscience.


Subject(s)
Brain , Image Processing, Computer-Assisted , Magnetic Resonance Imaging , Software , Animals , Magnetic Resonance Imaging/methods , Rats , Mice , Image Processing, Computer-Assisted/methods , Brain/diagnostic imaging , Brain/physiology , Reproducibility of Results , Data Accuracy , Brain Mapping/methods , Male , Quality Control
SELECTION OF CITATIONS
SEARCH DETAIL