Skip to main content
Erschienen in: Neurocritical Care 1/2021

Open Access 01.07.2021 | The Curing Coma Campaign

Mechanisms Underlying Disorders of Consciousness: Bridging Gaps to Move Toward an Integrated Translational Science

verfasst von: Andrea I. Luppi, Joshua Cain, Lennart R. B. Spindler, Urszula J. Górska, Daniel Toker, Andrew E. Hudson, Emery N. Brown, Michael N. Diringer, Robert D. Stevens, Marcello Massimini, Martin M. Monti, Emmanuel A. Stamatakis, Melanie Boly, the Curing Coma Campaign and Its Contributing Collaborators

Erschienen in: Neurocritical Care | Sonderheft 1/2021

Abstract

Aim

In order to successfully detect, classify, prognosticate, and develop targeted therapies for patients with disorders of consciousness (DOC), it is crucial to improve our mechanistic understanding of how severe brain injuries result in these disorders.

Methods

To address this need, the Curing Coma Campaign convened a Mechanisms Sub-Group of the Coma Science Work Group (CSWG), aiming to identify the most pressing knowledge gaps and the most promising approaches to bridge them.

Results

We identified a key conceptual gap in the need to differentiate the neural mechanisms of consciousness per se, from those underpinning connectedness to the environment and behavioral responsiveness. Further, we characterised three fundamental gaps in DOC research: (1) a lack of mechanistic integration between structural brain damage and abnormal brain function in DOC; (2) a lack of translational bridges between micro- and macro-scale neural phenomena; and (3) an incomplete exploration of possible synergies between data-driven and theory-driven approaches.

Conclusion

In this white paper, we discuss research priorities that would enable us to begin to close these knowledge gaps. We propose that a fundamental step towards this goal will be to combine translational, multi-scale, and multimodal data, with new biomarkers, theory-driven approaches, and computational models, to produce an integrated account of neural mechanisms in DOC. Importantly, we envision that reciprocal interaction between domains will establish a “virtuous cycle,” leading towards a critical vantage point of integrated knowledge that will enable the advancement of the scientific understanding of DOC and consequently, an improvement of clinical practice.
Hinweise
This article is part of the collection “Neurocritical Care Through History”.
A correction to this article is available online at https://​doi.​org/​10.​1007/​s12028-022-01488-1.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction and Current State of Science

The last two decades have seen growing interest in the neuroscience of disorders of consciousness (DOC). Significant progress has led to the differentiation of clinical phenotypes, including the discovery of new syndromic entities such as cognitive-motor dissociation (CMD) syndrome, a condition characterized by behavioral unresponsiveness paired with evidence of covert consciousness (voluntary brain activity). At the same time, this progress has also highlighted the critical limitations in our practical ability to diagnose covert consciousness at the bedside, predict long-term trajectories and outcomes, and enhance neurological recovery with therapies that target specific biological mechanisms.
The discovery through functional magnetic resonance imaging (fMRI) studies [13] and scalp electroencephalography (EEG) [4] that ~15–20% of patients with DOC who lack overt behavioral responsiveness may nevertheless be covertly conscious highlights the critical need for diagnostic tools that rely on brain activity. Although some theory-driven approaches already seem promising [5, 6], further elucidation of the mechanisms underlying unconsciousness in DOC, as distinct from responsiveness, is a fundamental requirement to guide the development of new accurate bedside diagnostic tools. Additionally, almost 90% of patients with chronic DOC do not recover 1 year post injury [7]. Accordingly, there is a need to draw on large-scale and, ideally, longitudinal clinical studies to properly model prognostic trajectories. Although some new pharmacological (e.g., amantadine, zolpidem) and interventional therapies (e.g., deep brain stimulation [8], low-intensity-focused ultrasound [9]) have recently emerged for patients with DOC, both case reports and randomized controlled trials demonstrate only moderate success. Thus, a better mechanistic understanding of the different pathways leading to altered consciousness after brain injury could help develop more effective therapies as well as tailor more personalized treatment options to specific patients.
In the present white paper, we aim to identify the most pressing current gaps in our understanding of DOC and strategies for closing those gaps. Because DOCs are characterized by heterogeneity, both in phenotype (arousal and cognitive and motor functions) and brain function, understanding the specific neural mechanisms of DOC will require investigation using integrative perspectives. Specifically, we conceptualize three targets for such integration: linking brain structure and function, linking microscale and macroscale levels of analysis, and combining theory-driven and data-driven approaches to scientific discovery. We further envision that enhanced synergy between these three domains will provide a critical vantage point from which to empower diagnosis, prognosis, and treatment of patients, thereby establishing a virtuous cycle between scientific advances and clinical practice (highlighted in Fig. 1).

Methodology

A 13-member Mechanisms Work Group (co-authors of the present white paper) was convened as part of the Curing Coma Campaign, Coma Science Work Group to identify research gaps and approaches to address these gaps. The work group met weekly from June 19, 2020, to September 4, 2020, to develop consensus recommendations and on an as-needed basis while authoring the current article.
First, literature review and expert discussion were performed by clinical syndrome: coma, vegetative state/unresponsive wakefulness syndrome (VS/UWS), minimally conscious state (MCS), and CMD. After a meeting with the Coma Science Work Group endotype subgroup, a second round of literature review and expert discussion was organized to draft the current recommendations, grouped by research gap theme. The mechanisms are cast in terms of consciousness vs. environmental connectedness vs. responsiveness (see below). To make progress on understanding the relevant mechanisms, we have identified three key gaps that need to be filled (Fig. 1): (1) brain structure vs. brain function, (2) micro- vs macroscale neural mechanisms, and (3) theories vs. data-driven approaches.

Results of Gap Analysis

Mechanisms of Interest and Current Gaps: Consciousness, Environmental Connectedness, and Responsiveness

Disorders of consciousness are characterized by a wide array of symptoms and etiologies. Consequently, a number of classification schemes across different dimensions have been proposed that are based on aspects such as cognitive functions [10], awareness [11] and sensory, motor, and arousal behavioral functions [12]. With the acknowledgment that any one framework cannot fully capture all the constructs relevant to DOC, we propose to contextualize our recommendations within the framework provided by Sanders et al. [13], which distinguishes between consciousness, environmental connectedness, and responsiveness (C-EC-R), as outlined below.
Lack of responsiveness remains the clinical criterion for DOC. However, there is a clear conceptual distinction between responsiveness (specifically, nonreflexive behavioral responses) and consciousness (presence of subjective experience, regardless of what the experience is about [13]; note that here we will use this term to also encompass awareness). The possibility for a dissociation between these two dimensions is sharply illustrated by the (rare) case of patients experiencing intraoperative awareness during anesthesia, whose unresponsiveness due to paralysis is mistaken for evidence of unconsciousness [1416]. Likewise, independent studies during sleep and anesthesia (in which retrospective reports can be obtained on awakening) demonstrate that subjective experience frequently occurs even in the absence of any behavioral responsiveness [17, 18]. Within DOC, routine assessment based on overt responsiveness may be missing covert consciousness in about ~15–20% of patients [3, 4], a fact that has practical and ethical implications for the management of these patients.
In addition to consciousness and responsiveness, a third relevant dimension is environmental connectedness (as originally proposed in the context of anesthesia [13]). Environmental connectedness corresponds to consciousness of the external environment so that what happens in the environment influences the contents of one’s consciousness (unlike, e.g., during some dream states). On the basis of this framework of C-EC-R, patients with CMD can be characterized as being conscious and environmentally connected but not (overtly) responsive. An intriguingly similar example of dissociation between consciousness and responsiveness is found in patients studied under anesthesia by using the isolated forearm technique, whereby an inflated cuff on the arm prevents it from being paralyzed during general anesthesia [13]. Despite the absence of spontaneous behaviors, such patients can perform simple commands, such as squeezing the physician’s hand (although the potential confound of arousing nociception due to the inflated cuff should also be considered). Examples of conscious but environmentally disconnected states include dreaming and dissociative states induced by ketamine; examples of states that are conscious and environmentally connected but unresponsive are awareness during general anesthesia and sleep paralysis, whereby the paralysis that naturally occurs during rapid eye movement (REM) sleep persists for a short time after awakening [19]. Of course, it is also important to consider the notion of behavioral arousal; although likely neither sufficient for consciousness (given the presence of sleep–wake cycles in patients with VS/UWS) nor necessary for it (given the possibility of dreams during deep non-REM sleep), arousal may nevertheless be a background prerequisite for responsiveness and possibly also for full-fledged environmental connectedness (Fig. 2).

Current Gaps in Coma Science

Correctly characterizing each patient in terms of the three dimensions described in the C-EC-R framework may be valuable for improving diagnosis and determining the prognosis and appropriate standards of care; for instance, it is especially pressing to establish communication with unresponsive but connected patients and not only identify reliable biomarkers for conscious contents, such as emotional distress or pain (i.e., the conscious processing of nociceptive information), but also identify whether some environmental stimuli trigger them (environmental connectedness). Additionally, by capitalizing on the availability of subjective reports after awakening from sleep or anesthesia in the laboratory environment, it will be crucial to identify ways to detect environmental connectedness based on brain function alone.
By identifying the neural mechanisms that determine transitions between the dimensions of the C-EC-R framework, it may be feasible to devise targeted treatment strategies aimed at restoring each in their own right and to identify which personalized avenues may be the most advantageous for a given patient. Specifically, efforts should be made not only to investigate whether specific mechanisms identified from studies of anesthesia and sleep [13] could also apply to patients with DOC (e.g., to what extent could the isolated forearm scenario constitute a good model for CMD?) but also to investigate how these different states differ from one another (anesthesia is reversible on a short time scale, but DOC may not always be). Additionally, investigations should seek to identify possible dissociations between each of these aspects in subgroups of patients with DOC.
Crucially, the limitations of our present ability to discriminate between elements of the C-EC-R framework in clinical practice are symptomatic of deeper gaps that need to be addressed to obtain a mechanistic understanding of DOC. DOC can arise from a variety of causes, highlighting the need for a more comprehensive understanding of the intricate interactions between structure and function and of their temporary vs. permanent nature. How brain function in turn determines C-EC-R also needs to be clarified, with clear diagnostic utility and prognostic value for recovery. Likewise, a complete understanding of the mechanisms underlying the presence vs. absence of C-EC-R will need to span multiple levels of analysis [20] and biological detail: from the cellular and molecular microscale to macroscale systems and networks. Reconciling the distinct levels of analysis will require the concerted interaction of data-driven approaches combining multimodal data from large-scale studies with theoretical approaches able to integrate their findings into a coherent framework, as well as synthesis through computational modeling. Thus, against the backdrop of the clinical need to discriminate between consciousness, connectedness, and responsiveness in patients with DOC, we envision these complementary approaches as establishing a virtuous cycle to drive forward both scientific understanding and clinical practice.
Box 1 Brain connectivity and networks
The term “functional connectivity” (FC) (Fig. 3a) reflects the notion that similarity between patterns of activity of different brain regions (in terms of statistical dependency) may arise from interactions between those regions. At the spatial and temporal resolutions afforded by noninvasive neuroimaging techniques in humans, neural activity is most frequently estimated in terms of electrophysiology (using EEG or magnetoencephalography) or from the blood-oxygen-level-dependent signal measured by fMRI. Activity is simultaneously measured for each sensor (for EEG/magnetoencephalography) or each voxel (for fMRI) over a period of time.
On the basis of these measurements of brain activity, the most common ways to quantify FC are measures of linear association between pairs of regional time series (primarily, Pearson correlation, but also methods based on phase coherence or spectral properties of the signals), which are therefore agnostic to interactions between more than two elements and ignore the direction of information flow between the two regions. However, more sophisticated measures also exist, capable of addressing various shortcomings of traditional FC (although often at the expense of computational feasibility) [2126].
Distinct sets of brain regions, termed “resting-state networks,” spontaneously organize into consistently cofluctuating assemblies during both tasks and also at rest. Prominent among these resting-state networks are the frontoparietal control network and the default mode network: these networks typically exhibit inversely correlated time courses at rest [27, 28], but their interactions are consistently perturbed in unconscious individuals [29, 30].
Measures of effective connectivity have also been introduced to identify directed information flow (from region A to region B and not vice versa). Some effective connectivity approaches rely on probabilistic accounts to infer the direction of interactions from statistical relationships in the data (e.g., transfer entropy, Granger causality [3133]). Another approach to characterize the directionality and strength of interactions, albeit limited to a small number of brain areas, is dynamic causal modeling (DCM), which has also been applied to patients with DOC [34]. The DCM framework is used to infer the direction of connectivity between regions by comparing possible models of how regional signals were generated. First, alternative models are constructed on the basis of possible coupling between regions, viewed as nodes in a directed network. In a second step, the models are compared through Bayesian model selection to identify the model that best explains the empirically observed data [35]. Finally, effective connectivity can be assessed by a perturb-and-measure approach, in which causal interactions are measured by directly stimulating a subset of neurons and by measuring the responses of the rest of the system. In addition to functional and effective connectivity, structural connectivity can also be measured in vivo in humans from diffusion magnetic resonance imaging data (Fig. 3b), for instance, through diffusion tensor imaging (DTI), which can measure the relative diffusion of water molecules along white matter fibers connecting different regions (although without providing information about directionality) [36]. Thus, structural connectivity and FC can be related in the same individual [24, 25, 37, 38].
Whether functional or structural, the interactions between brain regions can be conceived as a network (Fig. 3c), and the mathematical study of networks, known as graph theory, can be used to obtain insights about such networks at multiple levels of resolution [39, 40]: from properties of individual nodes (e.g., degree, measuring how well connected they are [41, 42]) to network modules [26] to macroscale properties such as small-world organization [24, 25, 30, 4345].

Brain Structure vs. Brain Function

Advancing the science of DOC requires a precise mapping of the heterogenous structural and functional brain alterations observed in DOC to clinically relevant dimensions (e.g., C-EC-R) across a variety of temporal and spatial scales. Although it is sometimes possible to precisely map neurological dysfunction to a specific location of damaged tissue, it is important to emphasize that brain regions are intricately interconnected, such that local structural or functional changes may well have far-reaching or even global repercussions on other components of the network (diaschisis [46]). Classical lesion-based methods have found success focusing on specific regions of interest (ROI); however, future work should attempt an integration of these approaches with the notion that there exists a many-to-many mapping between brain structure and functional brain states [47]. A full mapping between structural and functional correlates of DOC will require the leveraging of multimodal neuroimaging and neurophysiological techniques, combined with novel analytical methods for integrating them, including the emerging approach of whole-brain computational modeling (Box 2).
Historically, circumscribed lesions and changes in activity within damaged brains have been used to identify ROI and model simplified circuits that may be relevant to DOC symptoms. For instance, the influential mesocircuit model proposes that because of pathological changes following severe brain injuries, a reduction of thalamocortical and thalamostriatal outflow withdraws drive to the frontal cortex and striatum, thus implicating basal ganglia–thalamocortical circuits in the symptoms of DOC [10]. Compared to network or computational perspectives, these focal lesion-based models have the benefit of providing clear targets for treatment (e.g., via deep brain stimulation, low-intensity-focused ultrasound, pharmacological). For instance, emphasis on the role of the thalamus in DOC has compelled the development of techniques for its stimulation, which have been associated with improved behavioral responsiveness in a subset of patients with DOC [9, 48]. Yet the precise functional roles of key ROI, such as the thalamus, remain far from fully characterized; indeed, it remains unclear why nearly full ablation of the thalamus does not appear to result in a loss of consciousness in rodent models [49] despite its consistent association with DOC. Thus, focal characterization of structure–function relationships retains unique value but must continue to be refined.
For instance, although the original mesocircuit model emphasizes pallido-thalamo-cortical communication, this model has been updated by the recent discovery of direct structural connections between the globus pallidus and the cortex, first in rodents [50] and then in humans [51] by using DTI. In parallel, DCM (see Box 1) was applied to the mesocircuit and specifically implicated pallidocortical communication in the transition between states of consciousness under anesthesia [52]. This example illustrates the kind of multidisciplinary workflow that should inform the placement of ROI within increasingly complex frameworks (network, computational) for understanding the structural and functional relationships underlying DOC.
Compared to ROI approaches, the structural correlates of DOC may be further detailed by employing mass-univariate, voxel-wise analysis, which can produce mappings of behavioral symptoms to structural alterations at the millimeter scale (e.g., voxel-level symptom mapping [53, 54]; also see voxel-level shape analysis [55, 56]). Similar approaches have been applied to the structural connectome derived from DTI (known as connectome-based lesion symptoms mapping [57, 58]). Although these methods allow for a more spatially precise connection between large-scale functions (e.g., behavioral arousal) and structural damage, they do not capture the perhaps more elusive reorganizations in functional networks that often follow structural insult and that ultimately produce changes in behavior (e.g., diaschisis) as well as inadequate or maladaptive compensatory mechanisms, which may also produce DOC symptoms. Indeed, regions that are only secondarily affected may be particularly promising treatment targets because of their relative retained structural integrity, which may hold greater potential to reach preinjury levels of functioning. To capture the relationship between gray matter atrophy, white matter disconnection, and functional interactions, there is a need to better integrate structural correlates with the full range of functional modalities available to us instead of behavioral symptoms alone.
A jumping-off point for multimodal integration in DOC may be to overlay the structural correlates of DOC with the healthy human connectome (both structural and functional) to derive likely locations for a diaschisis effect, a method that avoids the often challenging process of multimodal data collection within patients themselves [59, 60]. Such an approach could be used to build whole-brain computational models, including the known structural correlates of DOC and known large-scale functional correlates (Box 2).
Box 2 Whole-brain computational models
Whole-brain computational models represent a powerful set of tools to study macroscale mechanistic questions in neuroscience [6264]. Such models typically combine two fundamental ingredients (Fig. 4a): (1) information about brain network structure (e.g., obtained from diffusion-weighted imaging in humans or invasive tract tracing in animals) and (2) a model of regional neural activity, ranging from Kuramoto or Hopf oscillators to the dynamic mean-field model obtained by mean-field reduction of integrate-and-fire spiking neurons with excitatory and inhibitory populations [65]. The complex interactions of these two key components can give rise to rich and biologically realistic functional dynamics analogous to those observed from fMRI and EEG [65, 66]. Although the required level of neurobiological detail will vary according to the specific question under investigation, the more biologically inspired models (e.g., dynamic mean-field) can also be enriched with further information, such as regional myelination or the regional distribution of specific receptors obtained from positron-emission tomography (PET) [64, 6770]
Importantly, in silico computational models offer several advantages: their parameters are fully available to inspection and manipulation by the researcher, and they can be perturbed in ways that would not be possible in either humans or animals [6770]. Computational modeling allows formulation and testing of specific mechanisms, a key feature not provided by other techniques (e.g., neuroimaging). Moreover, the same model can be subjected to different kinds of perturbations to investigate which pharmacological or structural interventions have equivalent results on the model’s function (Fig. 4b), a powerful avenue to interrogate potential similarities between anesthesia and DOC. Finally, the advent of computational models offers the unique promise to develop personalized models from each patient’s multimodal neuroimaging data, and subsequently perform systematic perturbation of the model to evaluate the potential effects of different treatment approaches, with the ultimate goal of informing which therapeutic modalities may be applicable for a patient
However, multimodal data collection in patients with DOC—and new methods for merging modalities with minimal information loss [60]—will be necessary to account for the inevitable translational gaps between healthy patients, computational models, and real patients with DOC. Some recent studies have pioneered these approaches; for instance, EEG markers have been linked to subcortical damage in acute [71] and chronic [72] DOC, and recent evidence indicates that preserved fractal (self-similar) character of structural brain networks is associated with covert consciousness on the basis of fMRI response to mental imagery tasks [73]. Inspiration for future joint investigations of structure and function may be drawn from other models of disrupted consciousness, such as anesthesia, in which links between structural and functional networks have recently been identified [24, 38].
To achieve clinical relevance, it will be crucial not only to collect increasingly abundant and multimodal data but also to distill from them the biomarkers that are most predictive of consciousness, connectedness, and responsiveness, either in terms of brain states or information structures, which may better explain how functions such as C-EC-R emerge instead of only from where [62]. For instance, the framework of connectome harmonic decomposition [47, 74] allows for functional brain activity to be decomposed in terms of different contributions of the human structural connectome [47]. Such connectome harmonics are wave-like patterns of spatial oscillations that represent how information spreads over the structural connectivity of the human brain. Through this approach, a common neural signature was recently identified between reduced consciousness in DOC and under anesthesia [74] and thus may represent a general signature of consciousness that can inform theoretical interpretations.
Of course, a plethora of other measures have emerged in recent years to describe network properties, including graph-theoretic and information-theory-based measures. The perturbational complexity index (PCI), which measures the complexity of EEG signals following causal perturbation by pulses of transcranial magnetic stimulation (TMS), appears particularly adept at detecting covert consciousness during sleep and anesthesia, as well as in patients with DOC, without the need for behavioral response [6]. Other measures of this kind, although perhaps less accurate for diagnosis, have also shown significant prognostic value [41, 75]. Future approaches should seek relationships and convergence between metrics derived from information theory, graph theory, and dynamical systems theory and strive to connect them with structural measures. Contemporary examples include the recent link found between PCI and subcortical atrophy [76] and the recent association between focal cortical lesions and the generation of pathological slow waves, disconnection, and lost complexity [77, 78].
Alongside sleep or anesthesia, seizures present yet another way to interrogate the generalizability of brain states to the C-EC-R framework in unique contexts (e.g., presence of fast-spiking high metabolism in seizures compared with anesthesia and DOC). This is especially relevant given that specific deficits in C-EC-R can all present during seizures in patients with chronic epilepsy [7981]. The loss of informational complexity typically observed during seizures [82] and the restored behavioral arousal following subcortical (pons, thalamus) stimulation during focal seizures in rats [83] suggests that generalizability is likely to be found; however, this area is ripe for more investigation.
Importantly, the first steps toward the integration of structure and function in DOC are largely taking place by experimental, analytic, and computational integration of the various neuroimaging methods that probe macroscale phenomena. However, a full mapping of structure–function relationships must inevitably dive deeper into the microscale neural substrate on which macroscale networks arise. Indeed, modeling the biological correlates of DOC across all spatial scales is likely to improve the location of individual patients within the heterogeneous space of DOC manifestations (e.g., as defined by C-EC-R). Thus, in the next section, we detail the gaps that exist in our understanding of microscale neural underpinnings of DOC and coma and how macro and micro levels of understanding may be bridged.

Linking Micro- and Macroscales

A comprehensive understanding of DOC requires that insights from macroscopic and mesoscopic levels of inquiry sampled by neuroimaging are integrated with insights from more microscopic scales: the relevant contributions at the systems, cellular, genetic, and molecular levels. Because these can largely only be studied directly in animal models, bridging this gap necessitates a closer association of preclinical and clinical research and the generation of hypotheses that can be bidirectionally tested in both animal and human research approaches.
Animal models that allow relevant direct measurements of such microscopic facets remain limited for coma [84] and fully absent for VS/UWS and MCS. The development of functionally relevant animal models will need to distinguish between the various neurological events (e.g., traumatic brain injury, anoxia, hypoxia among others) associated with DOC and the subsequent neurological syndromes of DOC (e.g., MCS, UWS/VS) [85, 86] to characterize both etiology-specific and generalizable DOC mechanisms. This could be further enhanced through comparison with results from sleep and anesthesia research, in which microscopic mechanisms have been more comprehensively probed in preclinical work [87], producing mechanistic frameworks that span from whole-brain phenomena, such as individual susceptibility, to anesthetic-state transitions (neural inertia), all the way to genetic susceptibility factors for anesthesia [38, 88, 89]. Indeed, work in rodents and cortical slices alike has recently demonstrated that neuronal “off” periods determine a dramatic collapse of large-scale interactions and complexity during non-REM sleep and anesthesia [9092], which can also be assessed by using EEG coupled with TMS in humans with brain damage [77, 78].
In terms of experimental approaches, novel noninvasive in vivo optogenetic techniques allow for the modulation of very specific and deeply seated targets in animal models [93, 94]. Leveraging these technological developments will enable systematic hypothesis-driven investigations of mechanisms that have been suggested both in previous anesthetic, sleep, and lesion studies in both animals and humans. Importantly, combining optogenetic stimulation with simultaneous high-resolution neuroimaging [95] could provide biomarkers to serve as direct translational interfaces between animal and human research.
Firstly, a systems-level microscopic perspective requires a comprehensive investigation of subcortical structures in neuroimaging given the implications of these structures in animal and translational research across trauma, anesthesia, and sleep. Specifically, the historical dichotomy between the cortex as the substrate of contents of consciousness and the thalamus and brainstem as substrates of arousal has oversimplified the various and diverse roles of subcortical systems [96]. As a first step, efforts should be directed toward a detailed mapping of how key subcortical structures (e.g., thalamus, brainstem nuclei, and basal ganglia) interact with specific cortical layers by using high-field structural and functional neuroimaging and complementary animal models [97, 98]. A relevant example of how current oversimplified views of the subcortical–cortical interplay can be refined into region- and lamina-specific accounts is provided by Redinbaugh et al. [97], who used thalamic stimulation in the anesthetized macaque to reveal that consciousness-relevant thalamic influence differs between deep and superficial cortical layers.
Similarly, critical insights at the microscopic systems level are to be gained from the brainstem neuromodulatory nuclei, which have been extensively studied in animals by using anesthetic and lesion approaches [99101]. Their associated transmitter systems and brain-wide neuromodulatory projections have been variously implicated as causing coma [60, 102, 103]. In healthy patients, they have been found to possibly drive both tonic and phasic large-scale in vivo brain activity [104106]. Established and novel single-photon emission computed tomography approaches [107] and innovative magnetic resonance sequences (e.g., magnetization transfer images [108]), combined with increased magnetic resonance field strengths, will allow these nuclei and their brain-wide projections to be more directly probed. These approaches should delineate whether dysfunction of these nuclei alters their modulation of the whole-brain connectome, which in turn may cause the striking macroscopic network disruptions commonly observed in DOC. This should use complementary approaches in both animal and patient research as well as computational modeling. Specifically, recent identification of the dopaminergic system’s relevance for wakefulness from anesthesia models has begun to provide a mechanistic foundation of why dopamine may have emerged as a key pharmacological treatment target in DOC [99, 109112]. To maximize therapeutic potential, comprehensive assessments of associated brainstem nuclei should also aim to delineate whether their dysfunctions in patients with DOC are pre- or postsynaptic using approaches, such as those demonstrated by Fridman et al. [113], combining PET with bolus administrations of pre- or postsynaptically acting pharmacological agents to characterize a presynaptic dopaminergic release deficit in patients with DOC.
Further progress along the microscopic perspective requires that differential involvements of particular neuronal types and subtypes in consciousness alterations be delineated. These studies will largely have to be delivered by using single-unit recordings and conditional expression approaches in novel animal models to delineate the relevance of neuronal types, e.g., pyramidal neurons [114]. For instance, anesthetic-induced decoupling between apical and basal compartments of layer 5 pyramidal neurons in rodents impairs large-scale functional integration in the brain [115]. These animal experiments can, however, also be complemented by work in human neuroimaging by using whole-brain transcriptomic maps from microarray data, as demonstrated by Craig et al. [116], who used this approach to delineate that GABAergic cortical interneuron subtypes are differently affected in anesthesia.
Indeed, it is key to not only characterize the involvement of different neuronal subtypes but equally characterize the role of glia and glial subtypes in DOC [117], especially given that widespread white matter damage is commonly associated with these conditions [118, 119]. Both animal models of glial modulation and alteration in response to trauma/anoxia [120] and new approaches in future neuroimaging studies [121] will have to constructively integrate white matter structure and function [121, 122]. An area of particular promise is the study of how microglia mediate systemic and regional inflammation following both traumatic brain injury and anoxia [123], leading to DOC. PET ligands for activated microglia and concomitant longitudinal collection of inflammatory and cell death markers in blood and/or cerebrospinal fluid should also distinguish inflammation in acute and chronic phases of DOC [124]. Animal models should aim to distinguish neuroprotective and pathogenic inflammatory effects in vivo [125] and distinguish whether pharmacological intervention can induce neuroprotective states, leading to more favorable outcomes. Similar multimodal techniques can also be used to begin to probe the roles of other glial cell types, such as astrocytes, whose role in overall homeostasis of the central nervous system has been demonstrated in traumatic brain injury and thus may play a similar role on the DOC spectrum [126, 127].
Finally, the subcellular and thus most microscopic level holds great promise for future explorations. In particular, it is necessary to identify genetic and molecular mediators of clinical progression and outcome in DOC. Beyond relying on analogies from anesthesia, it is required to delineate specific receptors and particular pathways that may be associated with the development or persistence of DOC. Although no specific genetic factors have yet been associated with DOC, genetic risk factors for adverse outcomes in traumatic brain injury/anoxia serve as useful and possibly intertwined starting points [128]. However, because human genome-wide association studies require prohibitively large population sizes, insights across all other levels of microscopic analysis (see Fig. 5) should be combined into biologically and physiologically relevant collections of gene candidates to be assessed with approaches such as transcriptome-wide association studies [129]. These could identify viable genetic candidates that in turn could be incorporated into both animal (assessing, e.g., loss-of-function and gain-of-function alterations) and human experiments (e.g., using microarray data [116]), thereby feeding back relevant, testable knowledge across all levels of analysis at which we need to consider DOC mechanisms as well as the theoretical frameworks that build on them.

Theory vs. Data-Driven Approaches

Recent research continues to enable a better understanding of the neuronal causes and clinical manifestations of DOC. Among the approaches that have recently been developed, we may distinguish purely empirical ones and those driven by assumptions of certain theories of consciousness. However, in their current state, it seems that both theories and data-driven approaches are unable to make predictions precise enough to enable a clear distinction between the neural mechanisms of the three dimensions of the C-EC-R framework. Yet this mechanistic understanding would carry immense clinical and ethical significance. For example, one of the key issues for clinicians is the ability to better predict the presence vs. absence of pain in unresponsive patients [130] and what may trigger it. Obtaining such diagnostic biomarkers would require a mechanistic understanding of the foundations of presence vs. absence of both consciousness and environmental connectedness [13] without reliance on behavior alone. Thus, a convergence between data-driven metrics (behavioral and neural) and theoretical perspectives on consciousness and environmental connectedness is needed to improve our ability to estimate the degree of residual consciousness and environmental connectedness (e.g., the presence of suffering) in unresponsive patients.
Recently, we have witnessed a formidable growth of new metrics based on neuroimaging [131] for the diagnosis of consciousness and the prognosis of recovery after severe brain injury. For example, various studies have shown a reduction in entropy (a measure of signal diversity or unpredictability) when consciousness fades, e.g., in sleep, anesthesia, and DOC [30, 61, 132, 133]. Although measures of this kind may demonstrate impressive predictive power, it remains unclear if certain metrics are relevant for consciousness per se or rather only represent epiphenomenal correlates (e.g., decline in entropy might be observed when transitioning from fast to slow oscillatory activity during anesthesia), and the above-mentioned EEG complexity has also been dissociated from responsiveness in anesthetized rodents [134]. Thus, although appealing, relying on promising but theoretically ambiguous data-driven metrics in a clinical setting runs the risk of introducing a discrepancy between what is intended to be detected (e.g., consciousness) and what is being measured; a similar problem is the so-called black box in medical artificial intelligence [135], whereby classification (diagnosis) and prediction (prognosis) can be successful but for opaque reasons [135].
Various prominent theories of consciousness tend to emphasize different neurophysiological underpinnings. For example, integrated information theory (IIT) suggests that consciousness requires a specific kind of causal interaction between elements of the system capable of supporting the integration of information [136]. On the basis of its early theoretical concepts [137139] that explicitly linked consciousness to complexity, defined as coexistence of functional differentiation and functional integration in the brain, measurement of EEG responses to magnetic perturbation (TMS) led to successful detection of residual awareness after brain injury (PCI [5]). Alternatively, the global neuronal workspace theory proposes that information processed in parallel by specialized modules needs to compete for access to a global workspace of frontoparietal circuits, whereupon it is integrated and subsequently broadcasted to the entire brain, becoming available for conscious processing [140142]. Some measures derived from graph theory (e.g., “small-worldness,” which reflects the balance of local segregation and global integration in a network [40]) have been argued to capture a decrease in such broadcasting in conditions of fading responsiveness [30, 43, 45, 143, 144]. However, there is still a need to more specifically associate these theoretical frameworks (among the many others) with concrete biomarkers so that empirical data may more clearly inform on the validity of particular theoretical interpretations over others. To do so, there is a pressing need to explicitly clarify, for example, the constructs of integration and information, as featured in different theoretical accounts, as well as their underlying assumptions.
In response to the current gaps and inconsistencies identified above, both data- and theory-driven approaches should advance toward unified terminology and contents that can be readily compared. First, we should aim for precise theoretical predictions for environmental disconnection and unconsciousness that are distinct from behavioral responsiveness, both at the information level and in terms of possible biological mechanisms. Indeed, there can be distinct biological mechanisms leading to the same functional or informational end point (e.g., multiple possible causes leading to brain states of low complexity). Both data- and theory-driven biomarkers for DOC should first be verified in conditions in which subjective reports are available (sleep, anesthesia, seizures) and then be precisely described in relation to their target: the distinction in C-EC-R (see Fig. 2).
To ameliorate the inconsistencies outlined here, theories should help to identify markers that may be most discriminative, and in the case of existing data-driven markers, their proponents should be able to explain their relevance to particular theoretical perspectives. Ideally, data-driven approaches should focus on a comprehensive set of markers indexing different biological levels, e.g., from metabolism and resting-state EEG/fMRI measures to effective connectivity or task-based paradigms. Similarly, comprehensive theoretical frameworks should span different descriptive levels (e.g., circuit level, information level, and topological level) and explicitly connect their predictions to different types of empirical markers.
We acknowledge that comparisons between existing theories are challenging because they sometimes operate on different definitions of consciousness. Furthermore, biological mechanistic frameworks, such as the mesocircuit hypothesis [10], may be compatible with a mathematical framework, such as IIT, when viewed as providing the required background conditions (e.g., neuromodulators maintaining adequate excitability) that enable the physical substrate of consciousness itself to function [76, 145]. Although some recent efforts have attempted to reconcile concepts derived from different theories of consciousness [146, 152], a direct involvement of the proponents of each theory in an adversarial context is most helpful to identify commonalities and differences in theoretical predictions and come up with specific experiments through which their predictions can be explicitly compared. For example, some incompatible predictions of global neuronal workspace and IIT are now being explicitly tested [147]. Ultimately, such adversarial collaboration may provide different degrees of data-driven support for different theoretical frameworks to make inferences about the presence of consciousness and/or environmental connectedness in patients with DOC.

Conclusion and Future Directions

In this position paper, we suggest that bringing together and integrating different levels of analyses and modalities, different scales, and different approaches, while at the same time attending to discrete relevant dimensions (C-EC-R), will prove a challenging but vital approach to push forward our mechanistic understanding of DOC (Table 1).
Table 1
Future research needs for investigating mechanisms of consciousness toward improved diagnosis, prognosis, and treatment of DOC
Research need: establish a framework for differentiating clinical subtypes of DOC with concepts of C-EC-R
Research need: identify links between structural brain damage and abnormal brain function in DOC
Form a complete mapping of DOC structural damage to functions of interest across multiple temporal and spatial scales
Identify structural correlates of relevant biomarkers of brain function in DOC patients derived from different approaches, e.g., information theory, graph theory, and dynamical systems theory
Research need: provide an integrated understanding of DOC across biological micro- and macroscales
Develop clinically relevant animal models of DOC for translational research approaches
Identify the role of subcortical structures and their interplay with the cortex in heterogeneous DOC
Associate microscale (neuronal and nonneuronal), and subcellular (molecular and genetic) mediators, with in vivo manifestations in patients with DOC
Research need: induce integration between theory-driven and data-driven approaches
Develop precise theoretical predictions and further biomarkers to address each dimension of the C-EC-R framework
Build a comprehensive set of data-driven and theory-driven biomarkers addressing different levels of analysis
Compare and develop existing theories in adversarial collaboration between theory leaders
Research need: integrate levels of description, imaging modalities, and theoretical approaches
Analyze multilevel and multimodal data from large-scale data sets to construct more realistic computational models of DOC
Develop personalized medicine models to guide treatment based on individual patients’ multimodal data
C-EC-R consciousness, environmental connectedness, and responsiveness, DOC disorders of consciousness
To achieve these ambitious goals, the field will need to leverage multimodal data in the same patients over time but also in diverse patients and across different states of altered consciousness (e.g., sleep, anesthesia, seizures), as well as in animal models, to enable the generalization of results. Several categories of techniques stand out for their ability to provide both improvements within each area and further integration between them, namely, (1) increasingly multilevel animal models, (2) analytic techniques for aggregating the various neural correlates of DOC (e.g., machine learning), and (3) increasingly multilevel whole-brain computational models.
Although direct animal models of DOC remain absent and our ability to disentangle consciousness from responsiveness in animals remains limited because of subjective reports being unattainable, animal models continue to be an important avenue for expanding our understanding of the biological mechanisms of DOC. Specifically, the expansion of multimodal methods for experimental manipulation in animals provides opportunities to selectively probe mechanisms and neuronal populations in the brain. Such work in animals could provide translational bridges from foundational microscale alterations and effectors to the neuroimaging findings commonly observed in patients with DOC [148] and thus catalyze the identification of translatable treatment targets and strategies.
To make sense of the wealth of data that such increasingly multimodal and multivariate approaches will produce, interrogation through new analytic tools will also be called for. Machine learning based on neuroimaging data has already been used to predict prognosis in patients with DOC [41, 42] as well as to differentiate states of consciousness (wakefulness from DOC or anesthesia) [75]. Nevertheless, predictive models derived from machine learning can help to both reduce redundancy between metrics and identify synergy between many identifiable neural correlates of DOC to distill clinically relevant predictions (e.g., probability of recovery, likelihood of covert consciousness).
In this vein, the increasing richness of empirical data can be leveraged to develop increasingly realistic computational models of DOC. In silico whole-brain models offer an especially promising avenue, with their potential to combine macroscale information about brain structure and function with considerations of microscale neurobiology [6264] (Box 2) as well as related empirical data and theoretical perspectives at multiple levels (e.g., how systems-level accounts of DOC interact with information perspectives). Indeed, successful models of anesthetic states have already been put forth [149151]. Therefore, computational models could provide means to address each of the three gaps we have identified, with the potential to aid personalized medicine.
Throughout this article, we have emphasized that closing the gaps we have identified must be understood as a synergistic endeavor. Addressing gaps at one level (e.g., the relationship between micro- and macroscale) will also inform our understanding of the interactions between brain structure and function. At the same time, closer translational interfaces between these areas of study will thereby inevitably emerge. Likewise, in addition to addressing the specific questions and challenges outlined here, large-scale multimodal data sets across humans and animals will provide the basis on which data-driven approaches and subsequent modeling can be developed further and refined. Finally, bidirectional interaction between scientific investigation and clinical practice will continue to play a fundamental role, with models and biomarkers informing prognosis and treatment while being informed by clinical insight, engendering a virtuous cycle. Nevertheless, we do not consider bridging these gaps as the end point of coma science but only as a new vantage point—a vantage point of integrated knowledge across levels, imaging modalities, and theoretical approaches—from which to pursue our goal: curing coma.

Acknowledgements

The authors wish to acknowledge the Curing Coma Campaign collaborators participating in the overall program and Angela Comanducci for helpful discussion. Figure 4 was created with biorender.com.

Conflicts of interest

The authors declare no conflicts of interest.

Source of Support

This work was supported by the Neurocritical Care Society; the Tiny Blue Dot Foundation; the Templeton World Charity Foundation, Inc.; grant NInDS K23NS112473; National Institutes of Health, National Institute of General Medical Sciences grant R01GM135420; National Institute of General Medical Sciences grant 1K08GM121961; the Johns Hopkins University Discovery Award and Stimulating and Advancing ACCM Research Award; the European Union’s Horizon 2020 Framework Program for Research and Innovation under the Specific Grant Agreement No. 945539 (Human Brain Project SGA3); the Canadian Institute for Advanced Research (RCZB/072 RG93193); the Stephen Erskine Fellowship (Queens’ College, Cambridge, UK); the Cambridge European Trust; and the Gates Cambridge Trust.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Jetzt e.Med zum Sonderpreis bestellen!

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

Jetzt bestellen und 100 € sparen!

e.Med Anästhesiologie

Kombi-Abonnement

Mit e.Med Anästhesiologie erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes AINS, den Premium-Inhalten der AINS-Fachzeitschriften, inklusive einer gedruckten AINS-Zeitschrift Ihrer Wahl.

e.Med Neurologie & Psychiatrie

Kombi-Abonnement

Mit e.Med Neurologie & Psychiatrie erhalten Sie Zugang zu CME-Fortbildungen der Fachgebiete, den Premium-Inhalten der dazugehörigen Fachzeitschriften, inklusive einer gedruckten Zeitschrift Ihrer Wahl.

Weitere Produktempfehlungen anzeigen
Literatur
1.
Zurück zum Zitat Owen AM, Coleman MR, Boly M, Davis MH, Laureys S, Pickard JD. Detecting awareness in the vegetative state. Science. 2006;313:1402.PubMed Owen AM, Coleman MR, Boly M, Davis MH, Laureys S, Pickard JD. Detecting awareness in the vegetative state. Science. 2006;313:1402.PubMed
2.
Zurück zum Zitat Monti MM, Vanhaudenhuyse A, Coleman MR, Boly M, Pickard JD, Tshibanda L, et al. Willful modulation of brain activity in disorders of consciousness. N Engl J Med. 2010;362:579–89.PubMed Monti MM, Vanhaudenhuyse A, Coleman MR, Boly M, Pickard JD, Tshibanda L, et al. Willful modulation of brain activity in disorders of consciousness. N Engl J Med. 2010;362:579–89.PubMed
3.
Zurück zum Zitat Naci L, Sinai L, Owen AM. Detecting and interpreting conscious experiences in behaviorally non-responsive patients. Neuroimage. 2017;145:304–13.PubMed Naci L, Sinai L, Owen AM. Detecting and interpreting conscious experiences in behaviorally non-responsive patients. Neuroimage. 2017;145:304–13.PubMed
4.
Zurück zum Zitat Claassen J, Doyle K, Matory A, Couch C, Burger KM, Velazquez A, et al. Detection of brain activation in unresponsive patients with acute brain injury. N Engl J Med. 2019;380:2497–505.PubMed Claassen J, Doyle K, Matory A, Couch C, Burger KM, Velazquez A, et al. Detection of brain activation in unresponsive patients with acute brain injury. N Engl J Med. 2019;380:2497–505.PubMed
5.
Zurück zum Zitat Casali AG, Gosseries O, Rosanova M, Boly M, Sarasso S, Casali KR, et al. A theoretically based index of consciousness independent of sensory processing and behavior. Sci Transl Med. 2013;5:198ra105.PubMed Casali AG, Gosseries O, Rosanova M, Boly M, Sarasso S, Casali KR, et al. A theoretically based index of consciousness independent of sensory processing and behavior. Sci Transl Med. 2013;5:198ra105.PubMed
6.
Zurück zum Zitat Casarotto S, Comanducci A, Rosanova M, Sarasso S, Fecchio M, Napolitani M, et al. Stratification of unresponsive patients by an independently validated index of brain complexity. Ann Neurol. 2016;80:718–29.PubMedPubMedCentral Casarotto S, Comanducci A, Rosanova M, Sarasso S, Fecchio M, Napolitani M, et al. Stratification of unresponsive patients by an independently validated index of brain complexity. Ann Neurol. 2016;80:718–29.PubMedPubMedCentral
7.
Zurück zum Zitat Estraneo A, Moretta P, Loreto V, Santoro L, Trojano L. Clinical and neuropsychological long-term outcomes after late recovery of responsiveness: a case series. Arch Phys Med Rehabil. 2014;95:711–6.PubMed Estraneo A, Moretta P, Loreto V, Santoro L, Trojano L. Clinical and neuropsychological long-term outcomes after late recovery of responsiveness: a case series. Arch Phys Med Rehabil. 2014;95:711–6.PubMed
8.
Zurück zum Zitat Schnakers C, Monti MM. Disorders of consciousness after severe brain injury: therapeutic options. Curr Opin Neurol. 2017;30:573–9.PubMed Schnakers C, Monti MM. Disorders of consciousness after severe brain injury: therapeutic options. Curr Opin Neurol. 2017;30:573–9.PubMed
9.
Zurück zum Zitat Cain JA, Spivak NM, Coetzee JP, Crone JS, Johnson MA, Lutkenhoff ES, et al. Ultrasonic thalamic stimulation in chronic disorders of consciousness. Brain Stimul. 2021;14:301–3.PubMed Cain JA, Spivak NM, Coetzee JP, Crone JS, Johnson MA, Lutkenhoff ES, et al. Ultrasonic thalamic stimulation in chronic disorders of consciousness. Brain Stimul. 2021;14:301–3.PubMed
10.
Zurück zum Zitat Schiff ND. Recovery of consciousness after brain injury: a mesocircuit hypothesis. Trends Neurosci. 2010;33:1–9.PubMed Schiff ND. Recovery of consciousness after brain injury: a mesocircuit hypothesis. Trends Neurosci. 2010;33:1–9.PubMed
11.
Zurück zum Zitat Laureys S. The neural correlate of (un)awareness: lessons from the vegetative state. Trends Cogn Sci. 2005;9:556–9.PubMed Laureys S. The neural correlate of (un)awareness: lessons from the vegetative state. Trends Cogn Sci. 2005;9:556–9.PubMed
12.
Zurück zum Zitat Giacino JT. The vegetative and minimally conscious states: consensus-based criteria for establishing diagnosis and prognosis. NeuroRehabilitation. 2004;19:293–8.PubMed Giacino JT. The vegetative and minimally conscious states: consensus-based criteria for establishing diagnosis and prognosis. NeuroRehabilitation. 2004;19:293–8.PubMed
13.
Zurück zum Zitat Sanders RD, Tononi G, Laureys S, Sleigh JW. Unresponsiveness ≠ unconsciousness. Anesthesiology. 2012;116:946–59.PubMed Sanders RD, Tononi G, Laureys S, Sleigh JW. Unresponsiveness ≠ unconsciousness. Anesthesiology. 2012;116:946–59.PubMed
14.
Zurück zum Zitat Moerman N, Bonke B, Oosting J. Awareness and recall during general anesthesia. Facts and feelings Anesthesiology. 1993;79:454–64.PubMed Moerman N, Bonke B, Oosting J. Awareness and recall during general anesthesia. Facts and feelings Anesthesiology. 1993;79:454–64.PubMed
15.
Zurück zum Zitat Ghoneim MM, Block RI, Haffarnan M, Mathews MJ. Awareness during anesthesia: risk factors, causes and sequelae: a review of reported cases in the literature. Anesth Analg. 2009;108:527–35.PubMed Ghoneim MM, Block RI, Haffarnan M, Mathews MJ. Awareness during anesthesia: risk factors, causes and sequelae: a review of reported cases in the literature. Anesth Analg. 2009;108:527–35.PubMed
16.
Zurück zum Zitat Sebel PS, Bowdle TA, Ghoneim MM, Rampil IJ, Padilla RE, Gan TJ, et al. The incidence of awareness during anesthesia: a multicenter United States study. Anesth Analg. 2004;99:833–9.PubMed Sebel PS, Bowdle TA, Ghoneim MM, Rampil IJ, Padilla RE, Gan TJ, et al. The incidence of awareness during anesthesia: a multicenter United States study. Anesth Analg. 2004;99:833–9.PubMed
17.
Zurück zum Zitat Nir Y, Tononi G. Dreaming and the brain: from phenomenology to neurophysiology. Trends Cogn Sci. 2010;14:88–100.PubMedPubMedCentral Nir Y, Tononi G. Dreaming and the brain: from phenomenology to neurophysiology. Trends Cogn Sci. 2010;14:88–100.PubMedPubMedCentral
18.
Zurück zum Zitat Leslie K, Sleigh J, Paech MJ, Voss L, Lim CW, Sleigh C. Dreaming and electroencephalographic changes during anesthesia maintained with propofol or desflurane. Anesthesiology. 2009;111:547–55.PubMed Leslie K, Sleigh J, Paech MJ, Voss L, Lim CW, Sleigh C. Dreaming and electroencephalographic changes during anesthesia maintained with propofol or desflurane. Anesthesiology. 2009;111:547–55.PubMed
19.
Zurück zum Zitat Cheyne JA, Newby-Clark IR, Rueffer SD. Relations among hypnagogic and hypnopompic experiences associated with sleep paralysis. J Sleep Res. 1999;8:313–7.PubMed Cheyne JA, Newby-Clark IR, Rueffer SD. Relations among hypnagogic and hypnopompic experiences associated with sleep paralysis. J Sleep Res. 1999;8:313–7.PubMed
20.
Zurück zum Zitat Marr D. Vision: a computational investigation into the human representation and processing of visual information. Cambridge (MA): The MIT Press; 2010. Marr D. Vision: a computational investigation into the human representation and processing of visual information. Cambridge (MA): The MIT Press; 2010.
21.
Zurück zum Zitat Crone JS, Schurz M, Höller Y, Bergmann J, Monti M, Schmid E, et al. Impaired consciousness is linked to changes in effective connectivity of the posterior cingulate cortex within the default mode network. Neuroimage. 2015;110:101–9.PubMed Crone JS, Schurz M, Höller Y, Bergmann J, Monti M, Schmid E, et al. Impaired consciousness is linked to changes in effective connectivity of the posterior cingulate cortex within the default mode network. Neuroimage. 2015;110:101–9.PubMed
22.
Zurück zum Zitat Deco G, Vidaurre D, Kringelbach ML. Revisiting the global workspace orchestrating the hierarchical organization of the human brain. Nat Hum Behav. 2021;5:497–511.PubMedPubMedCentral Deco G, Vidaurre D, Kringelbach ML. Revisiting the global workspace orchestrating the hierarchical organization of the human brain. Nat Hum Behav. 2021;5:497–511.PubMedPubMedCentral
23.
Zurück zum Zitat Mediano PAM, Rosas F, Carhart-Harris RL, Seth AK, Barrett AB. Beyond integrated information: A taxonomy of information dynamics phenomena. arXiv. 2019; arXiv:1909.02297. Mediano PAM, Rosas F, Carhart-Harris RL, Seth AK, Barrett AB. Beyond integrated information: A taxonomy of information dynamics phenomena. arXiv. 2019; arXiv:​1909.​02297.
24.
Zurück zum Zitat Barttfeld P, Uhrig L, Sitt JD, Sigman M, Jarraya B, Dehaene S. Signature of consciousness in the dynamics of resting-state brain activity. Proc Natl Acad Sci USA. 2015;112:887–92.PubMedPubMedCentral Barttfeld P, Uhrig L, Sitt JD, Sigman M, Jarraya B, Dehaene S. Signature of consciousness in the dynamics of resting-state brain activity. Proc Natl Acad Sci USA. 2015;112:887–92.PubMedPubMedCentral
25.
Zurück zum Zitat Zamani Esfahlani F, Jo Y, Faskowitz J, Byrge L, Kennedy DP, Sporns O, et al. High-amplitude cofluctuations in cortical activity drive functional connectivity. Proc Natl Acad Sci USA. 2020;117:28393–401.PubMedPubMedCentral Zamani Esfahlani F, Jo Y, Faskowitz J, Byrge L, Kennedy DP, Sporns O, et al. High-amplitude cofluctuations in cortical activity drive functional connectivity. Proc Natl Acad Sci USA. 2020;117:28393–401.PubMedPubMedCentral
26.
Zurück zum Zitat Shine JM, Bissett PG, Bell PT, Koyejo O, Balsters JH, Gorgolewski KJ, et al. The dynamics of functional brain networks: integrated network states during cognitive task performance. Neuron. 2016;92:544–54.PubMedPubMedCentral Shine JM, Bissett PG, Bell PT, Koyejo O, Balsters JH, Gorgolewski KJ, et al. The dynamics of functional brain networks: integrated network states during cognitive task performance. Neuron. 2016;92:544–54.PubMedPubMedCentral
27.
Zurück zum Zitat Raichle ME, MacLeod AM, Snyder AZ, Powers WJ, Gusnard DA, Shulman GL. A default mode of brain function. Proc Natl Acad Sci USA. 2001;98:676–82.PubMedPubMedCentral Raichle ME, MacLeod AM, Snyder AZ, Powers WJ, Gusnard DA, Shulman GL. A default mode of brain function. Proc Natl Acad Sci USA. 2001;98:676–82.PubMedPubMedCentral
28.
Zurück zum Zitat Fox MD, Snyder AZ, Vincent JL, Corbetta M, Van Essen DC, Raichle ME. The human brain is intrinsically organized into dynamic, anticorrelated functional networks. Proc Natl Acad Sci USA. 2005;102:9673–8.PubMedPubMedCentral Fox MD, Snyder AZ, Vincent JL, Corbetta M, Van Essen DC, Raichle ME. The human brain is intrinsically organized into dynamic, anticorrelated functional networks. Proc Natl Acad Sci USA. 2005;102:9673–8.PubMedPubMedCentral
29.
Zurück zum Zitat Di Perri C, Bahri MA, Amico E, Thibaut A, Heine L, Antonopoulos G, et al. Neural correlates of consciousness in patients who have emerged from a minimally conscious state: a cross-sectional multimodal imaging study. Lancet Neurol. 2016;15:830–42.PubMed Di Perri C, Bahri MA, Amico E, Thibaut A, Heine L, Antonopoulos G, et al. Neural correlates of consciousness in patients who have emerged from a minimally conscious state: a cross-sectional multimodal imaging study. Lancet Neurol. 2016;15:830–42.PubMed
30.
Zurück zum Zitat Luppi AI, Craig MM, Pappas I, Finoia P, Williams GB, Allanson J, et al. Consciousness-specific dynamic interactions of brain integration and functional diversity. Nat Commun. 2019;10:4616.PubMedPubMedCentral Luppi AI, Craig MM, Pappas I, Finoia P, Williams GB, Allanson J, et al. Consciousness-specific dynamic interactions of brain integration and functional diversity. Nat Commun. 2019;10:4616.PubMedPubMedCentral
31.
Zurück zum Zitat Barrett AB, Murphy M, Bruno M-A, Noirhomme Q, Boly M, Laureys S, et al. Granger causality analysis of steady-state electroencephalographic signals during propofol-induced anaesthesia. PLoS ONE. 2012;7:e29072.PubMedPubMedCentral Barrett AB, Murphy M, Bruno M-A, Noirhomme Q, Boly M, Laureys S, et al. Granger causality analysis of steady-state electroencephalographic signals during propofol-induced anaesthesia. PLoS ONE. 2012;7:e29072.PubMedPubMedCentral
32.
Zurück zum Zitat Schreiber T. Measuring information transfer. Phys Rev Lett. 2000;85:461–4.PubMed Schreiber T. Measuring information transfer. Phys Rev Lett. 2000;85:461–4.PubMed
33.
Zurück zum Zitat Massey J. Causality, feedback and directed information. Proceedings of the 1990 International Symposium on Information Theory and its Applications. 1990. Massey J. Causality, feedback and directed information. Proceedings of the 1990 International Symposium on Information Theory and its Applications. 1990.
34.
Zurück zum Zitat Boly M, Garrido MI, Gosseries O, Bruno M-A, Boveroux P, Schnakers C, et al. Preserved feedforward but impaired top-down processes in the vegetative state. Science. 2011;332:858–62.PubMed Boly M, Garrido MI, Gosseries O, Bruno M-A, Boveroux P, Schnakers C, et al. Preserved feedforward but impaired top-down processes in the vegetative state. Science. 2011;332:858–62.PubMed
35.
Zurück zum Zitat Stephan KE, Harrison LM, Kiebel SJ, David O, Penny WD, Friston KJ. Dynamic causal models of neural system dynamics: current state and future extensions. J Biosci. 2007;32:129–44.PubMedPubMedCentral Stephan KE, Harrison LM, Kiebel SJ, David O, Penny WD, Friston KJ. Dynamic causal models of neural system dynamics: current state and future extensions. J Biosci. 2007;32:129–44.PubMedPubMedCentral
36.
Zurück zum Zitat Hagmann P, Cammoun L, Gigandet X, Meuli R, Honey CJ, Wedeen VJ, et al. Mapping the structural core of human cerebral cortex. PLoS Biol. 2008;6:e159.PubMedPubMedCentral Hagmann P, Cammoun L, Gigandet X, Meuli R, Honey CJ, Wedeen VJ, et al. Mapping the structural core of human cerebral cortex. PLoS Biol. 2008;6:e159.PubMedPubMedCentral
37.
Zurück zum Zitat Amico E, Marinazzo D, Di Perri C, Heine L, Annen J, Martial C, et al. Mapping the functional connectome traits of levels of consciousness. Neuroimage. 2017;148:201–11.PubMed Amico E, Marinazzo D, Di Perri C, Heine L, Annen J, Martial C, et al. Mapping the functional connectome traits of levels of consciousness. Neuroimage. 2017;148:201–11.PubMed
38.
Zurück zum Zitat Uhrig L, Sitt JD, Jacob A, Tasserie J, Barttfeld P, Dupont M, et al. Resting-state dynamics as a cortical signature of anesthesia in monkeys. Anesthesiology. 2018;129:942–58.PubMed Uhrig L, Sitt JD, Jacob A, Tasserie J, Barttfeld P, Dupont M, et al. Resting-state dynamics as a cortical signature of anesthesia in monkeys. Anesthesiology. 2018;129:942–58.PubMed
39.
Zurück zum Zitat Sporns O. From simple graphs to the connectome: networks in neuroimaging. Neuroimage. 2012;62:881–6.PubMed Sporns O. From simple graphs to the connectome: networks in neuroimaging. Neuroimage. 2012;62:881–6.PubMed
40.
Zurück zum Zitat Bullmore E, Sporns O. The economy of brain network organization. Nat Rev Neurosci. 2012;13:336–49.PubMed Bullmore E, Sporns O. The economy of brain network organization. Nat Rev Neurosci. 2012;13:336–49.PubMed
41.
Zurück zum Zitat Chennu S, Annen J, Wannez S, Thibaut A, Chatelle C, Cassol H, et al. Brain networks predict metabolism, diagnosis and prognosis at the bedside in disorders of consciousness. Brain. 2017;140:2120–32.PubMed Chennu S, Annen J, Wannez S, Thibaut A, Chatelle C, Cassol H, et al. Brain networks predict metabolism, diagnosis and prognosis at the bedside in disorders of consciousness. Brain. 2017;140:2120–32.PubMed
42.
Zurück zum Zitat Pappas I, Adapa RM, Menon DK, Stamatakis EA. Brain network disintegration during sedation is mediated by the complexity of sparsely connected regions. Neuroimage. 2019;186:221–33.PubMed Pappas I, Adapa RM, Menon DK, Stamatakis EA. Brain network disintegration during sedation is mediated by the complexity of sparsely connected regions. Neuroimage. 2019;186:221–33.PubMed
43.
Zurück zum Zitat Achard S, Delon-Martin C, Vértes PE, Renard F, Schenck M, Schneider F, et al. Hubs of brain functional networks are radically reorganized in comatose patients. Proc Natl Acad Sci USA. 2012;109:20608–13.PubMedPubMedCentral Achard S, Delon-Martin C, Vértes PE, Renard F, Schenck M, Schneider F, et al. Hubs of brain functional networks are radically reorganized in comatose patients. Proc Natl Acad Sci USA. 2012;109:20608–13.PubMedPubMedCentral
44.
Zurück zum Zitat Crone JS, Soddu A, Höller Y, Vanhaudenhuyse A, Schurz M, Bergmann J, et al. Altered network properties of the fronto-parietal network and the thalamus in impaired consciousness. Neuroimage Clin. 2013;4:240–8.PubMedPubMedCentral Crone JS, Soddu A, Höller Y, Vanhaudenhuyse A, Schurz M, Bergmann J, et al. Altered network properties of the fronto-parietal network and the thalamus in impaired consciousness. Neuroimage Clin. 2013;4:240–8.PubMedPubMedCentral
45.
Zurück zum Zitat Monti MM, Lutkenhoff ES, Rubinov M, Boveroux P, Vanhaudenhuyse A, Gosseries O, et al. Dynamic change of global and local information processing in propofol-induced loss and recovery of consciousness. PLoS Comput Biol. 2013;9:e1003271. Monti MM, Lutkenhoff ES, Rubinov M, Boveroux P, Vanhaudenhuyse A, Gosseries O, et al. Dynamic change of global and local information processing in propofol-induced loss and recovery of consciousness. PLoS Comput Biol. 2013;9:e1003271.
46.
Zurück zum Zitat Carrera E, Tononi G. Diaschisis: past, present, future. Brain. 2014;137:2408–22.PubMed Carrera E, Tononi G. Diaschisis: past, present, future. Brain. 2014;137:2408–22.PubMed
47.
Zurück zum Zitat Atasoy S, Donnelly I, Pearson J. Human brain networks function in connectome-specific harmonic waves. Nat Commun. 2016;7:10340.PubMedPubMedCentral Atasoy S, Donnelly I, Pearson J. Human brain networks function in connectome-specific harmonic waves. Nat Commun. 2016;7:10340.PubMedPubMedCentral
48.
Zurück zum Zitat Schiff ND, Giacino JT, Kalmar K, Victor JD, Baker K, Gerber M, et al. Behavioural improvements with thalamic stimulation after severe traumatic brain injury. Nature. 2007;448:600–3.PubMed Schiff ND, Giacino JT, Kalmar K, Victor JD, Baker K, Gerber M, et al. Behavioural improvements with thalamic stimulation after severe traumatic brain injury. Nature. 2007;448:600–3.PubMed
49.
Zurück zum Zitat Fuller PM, Sherman D, Pedersen NP, Saper CB, Lu J. Reassessment of the structural basis of the ascending arousal system. J Comp Neurol. 2011;519:933–56.PubMedPubMedCentral Fuller PM, Sherman D, Pedersen NP, Saper CB, Lu J. Reassessment of the structural basis of the ascending arousal system. J Comp Neurol. 2011;519:933–56.PubMedPubMedCentral
50.
Zurück zum Zitat Chen MC, Ferrari L, Sacchet MD, Foland-Ross LC, Qiu M-H, Gotlib IH, et al. Identification of a direct GABAergic pallidocortical pathway in rodents. Eur J Neurosci. 2015;41:748–59.PubMedPubMedCentral Chen MC, Ferrari L, Sacchet MD, Foland-Ross LC, Qiu M-H, Gotlib IH, et al. Identification of a direct GABAergic pallidocortical pathway in rodents. Eur J Neurosci. 2015;41:748–59.PubMedPubMedCentral
52.
Zurück zum Zitat Crone JS, Lutkenhoff ES, Bio BJ, Laureys S, Monti MM. Testing proposed neuronal models of effective connectivity within the cortico-basal ganglia-thalamo-cortical loop during loss of consciousness. Cereb Cortex. 2017;27:2727–38.PubMed Crone JS, Lutkenhoff ES, Bio BJ, Laureys S, Monti MM. Testing proposed neuronal models of effective connectivity within the cortico-basal ganglia-thalamo-cortical loop during loss of consciousness. Cereb Cortex. 2017;27:2727–38.PubMed
53.
Zurück zum Zitat Bates E, Wilson SM, Saygin AP, Dick F, Sereno MI, Knight RT, et al. Voxel-based lesion-symptom mapping. Nat Neurosci. 2003;6:448–50.PubMed Bates E, Wilson SM, Saygin AP, Dick F, Sereno MI, Knight RT, et al. Voxel-based lesion-symptom mapping. Nat Neurosci. 2003;6:448–50.PubMed
54.
Zurück zum Zitat Tyler LK, Marslen-Wilson W, Stamatakis EA. Dissociating neuro-cognitive component processes: voxel-based correlational methodology. Neuropsychologia. 2005;43:771–8.PubMed Tyler LK, Marslen-Wilson W, Stamatakis EA. Dissociating neuro-cognitive component processes: voxel-based correlational methodology. Neuropsychologia. 2005;43:771–8.PubMed
55.
Zurück zum Zitat Lutkenhoff ES, McArthur DL, Hua X, Thompson PM, Vespa PM, Monti MM. Thalamic atrophy in antero-medial and dorsal nuclei correlates with six-month outcome after severe brain injury. Neuroimage Clin. 2013;3:396–404.PubMedPubMedCentral Lutkenhoff ES, McArthur DL, Hua X, Thompson PM, Vespa PM, Monti MM. Thalamic atrophy in antero-medial and dorsal nuclei correlates with six-month outcome after severe brain injury. Neuroimage Clin. 2013;3:396–404.PubMedPubMedCentral
56.
Zurück zum Zitat Lutkenhoff ES, Chiang J, Tshibanda L, Kamau E, Kirsch M, Pickard JD, et al. Thalamic and extrathalamic mechanisms of consciousness after severe brain injury. Ann Neurol. 2015;78:68–76.PubMed Lutkenhoff ES, Chiang J, Tshibanda L, Kamau E, Kirsch M, Pickard JD, et al. Thalamic and extrathalamic mechanisms of consciousness after severe brain injury. Ann Neurol. 2015;78:68–76.PubMed
57.
Zurück zum Zitat Gleichgerrcht E, Fridriksson J, Rorden C, Bonilha L. Connectome-based lesion-symptom mapping (CLSM): a novel approach to map neurological function. Neuroimage Clin. 2017;16:461–7.PubMedPubMedCentral Gleichgerrcht E, Fridriksson J, Rorden C, Bonilha L. Connectome-based lesion-symptom mapping (CLSM): a novel approach to map neurological function. Neuroimage Clin. 2017;16:461–7.PubMedPubMedCentral
58.
Zurück zum Zitat Zheng ZS, Reggente N, Lutkenhoff E, Owen AM, Monti MM. Disentangling disorders of consciousness: insights from diffusion tensor imaging and machine learning. Hum Brain Mapp. 2017;38:431–43.PubMed Zheng ZS, Reggente N, Lutkenhoff E, Owen AM, Monti MM. Disentangling disorders of consciousness: insights from diffusion tensor imaging and machine learning. Hum Brain Mapp. 2017;38:431–43.PubMed
59.
Zurück zum Zitat Boes AD, Prasad S, Liu H, Liu Q, Pascual-Leone A, Caviness VS, et al. Network localization of neurological symptoms from focal brain lesions. Brain. 2015;138:3061–75.PubMedPubMedCentral Boes AD, Prasad S, Liu H, Liu Q, Pascual-Leone A, Caviness VS, et al. Network localization of neurological symptoms from focal brain lesions. Brain. 2015;138:3061–75.PubMedPubMedCentral
60.
Zurück zum Zitat Fischer DB, Boes AD, Demertzi A, Evrard HC, Laureys S, Edlow BL, et al. A human brain network derived from coma-causing brainstem lesions. Neurology. 2016;87:2427–34.PubMedPubMedCentral Fischer DB, Boes AD, Demertzi A, Evrard HC, Laureys S, Edlow BL, et al. A human brain network derived from coma-causing brainstem lesions. Neurology. 2016;87:2427–34.PubMedPubMedCentral
61.
Zurück zum Zitat Demertzi A, Tagliazucchi E, Dehaene S, Deco G, Barttfeld P, Raimondo F, et al. Human consciousness is supported by dynamic complex patterns of brain signal coordination. Sci Adv. 2019;5:eaat7603. Demertzi A, Tagliazucchi E, Dehaene S, Deco G, Barttfeld P, Raimondo F, et al. Human consciousness is supported by dynamic complex patterns of brain signal coordination. Sci Adv. 2019;5:eaat7603.
62.
Zurück zum Zitat Cofré R, Herzog R, Mediano PAM, Piccinini J, Rosas FE, Sanz Perl Y, et al. Whole-brain models to explore altered states of consciousness from the bottom up. Brain Sci. 2020;10:626.PubMedCentral Cofré R, Herzog R, Mediano PAM, Piccinini J, Rosas FE, Sanz Perl Y, et al. Whole-brain models to explore altered states of consciousness from the bottom up. Brain Sci. 2020;10:626.PubMedCentral
63.
Zurück zum Zitat Kringelbach ML, Deco G. Brain states and transitions: insights from computational neuroscience. Cell Rep. 2020;32:108128. Kringelbach ML, Deco G. Brain states and transitions: insights from computational neuroscience. Cell Rep. 2020;32:108128.
65.
Zurück zum Zitat Cabral J, Kringelbach ML, Deco G. Functional connectivity dynamically evolves on multiple time-scales over a static structural connectome: models and mechanisms. Neuroimage. 2017;160:84–96.PubMed Cabral J, Kringelbach ML, Deco G. Functional connectivity dynamically evolves on multiple time-scales over a static structural connectome: models and mechanisms. Neuroimage. 2017;160:84–96.PubMed
66.
Zurück zum Zitat Bensaid S, Modolo J, Merlet I, Wendling F, Benquet P. COALIA: a computational model of human EEG for consciousness research. Front Syst Neurosci. 2019;13:59.PubMedPubMedCentral Bensaid S, Modolo J, Merlet I, Wendling F, Benquet P. COALIA: a computational model of human EEG for consciousness research. Front Syst Neurosci. 2019;13:59.PubMedPubMedCentral
67.
Zurück zum Zitat Demirtaş M, Burt JB, Helmer M, Ji JL, Adkinson BD, Glasser MF, et al. Hierarchical heterogeneity across human cortex shapes large-scale neural dynamics. Neuron. 2019;101:1181-94.e13.PubMedPubMedCentral Demirtaş M, Burt JB, Helmer M, Ji JL, Adkinson BD, Glasser MF, et al. Hierarchical heterogeneity across human cortex shapes large-scale neural dynamics. Neuron. 2019;101:1181-94.e13.PubMedPubMedCentral
68.
Zurück zum Zitat Wang P, Kong R, Kong X, Liégeois R, Orban C, Deco G, et al. Inversion of a large-scale circuit model reveals a cortical hierarchy in the dynamic resting human brain. Sci Adv. 2019;5:eaat7854. Wang P, Kong R, Kong X, Liégeois R, Orban C, Deco G, et al. Inversion of a large-scale circuit model reveals a cortical hierarchy in the dynamic resting human brain. Sci Adv. 2019;5:eaat7854.
69.
Zurück zum Zitat Deco G, Cruzat J, Cabral J, Knudsen GM, Carhart-Harris RL, Whybrow PC, et al. Whole-brain multimodal neuroimaging model using serotonin receptor maps explains non-linear functional effects of LSD. Curr Biol. 2018;28:3065-74.e6.PubMed Deco G, Cruzat J, Cabral J, Knudsen GM, Carhart-Harris RL, Whybrow PC, et al. Whole-brain multimodal neuroimaging model using serotonin receptor maps explains non-linear functional effects of LSD. Curr Biol. 2018;28:3065-74.e6.PubMed
70.
Zurück zum Zitat Kringelbach ML, Cruzat J, Cabral J, Knudsen GM, Carhart-Harris R, Whybrow PC, et al. Dynamic coupling of whole-brain neuronal and neurotransmitter systems. Proc Natl Acad Sci USA. 2020;117:9566–76.PubMedPubMedCentral Kringelbach ML, Cruzat J, Cabral J, Knudsen GM, Carhart-Harris R, Whybrow PC, et al. Dynamic coupling of whole-brain neuronal and neurotransmitter systems. Proc Natl Acad Sci USA. 2020;117:9566–76.PubMedPubMedCentral
71.
Zurück zum Zitat Schnakers C, Lutkenhoff ES, Bio BJ, McArthur DL, Vespa PM, Monti MM. Acute EEG spectra characteristics predict thalamic atrophy after severe TBI. J Neurol Neurosurg Psychiatry. 2019;90:617–9.PubMed Schnakers C, Lutkenhoff ES, Bio BJ, McArthur DL, Vespa PM, Monti MM. Acute EEG spectra characteristics predict thalamic atrophy after severe TBI. J Neurol Neurosurg Psychiatry. 2019;90:617–9.PubMed
73.
Zurück zum Zitat Luppi AI, Craig MM, Coppola P, Peattie ARD, Finoia P, Williams GB, et al. Preserved fractal character of structural brain networks is associated with covert consciousness after severe brain injury. Neuroimage Clin. 2021;30:102682. Luppi AI, Craig MM, Coppola P, Peattie ARD, Finoia P, Williams GB, et al. Preserved fractal character of structural brain networks is associated with covert consciousness after severe brain injury. Neuroimage Clin. 2021;30:102682.
75.
Zurück zum Zitat Demertzi A, Antonopoulos G, Heine L, Voss HU, Crone JS, de Los AC, et al. Intrinsic functional connectivity differentiates minimally conscious from unresponsive patients. Brain. 2015;138:2619–31.PubMed Demertzi A, Antonopoulos G, Heine L, Voss HU, Crone JS, de Los AC, et al. Intrinsic functional connectivity differentiates minimally conscious from unresponsive patients. Brain. 2015;138:2619–31.PubMed
76.
Zurück zum Zitat Lutkenhoff ES, Johnson MA, Casarotto S, Massimini M, Monti MM. Subcortical atrophy correlates with the perturbational complexity index in patients with disorders of consciousness. Brain Stimul. 2020;13:1426–35.PubMed Lutkenhoff ES, Johnson MA, Casarotto S, Massimini M, Monti MM. Subcortical atrophy correlates with the perturbational complexity index in patients with disorders of consciousness. Brain Stimul. 2020;13:1426–35.PubMed
77.
Zurück zum Zitat Sarasso S, D’Ambrosio S, Fecchio M, Casarotto S, Viganò A, Landi C, et al. Local sleep-like cortical reactivity in the awake brain after focal injury. Brain. 2020;143:3672–84.PubMedPubMedCentral Sarasso S, D’Ambrosio S, Fecchio M, Casarotto S, Viganò A, Landi C, et al. Local sleep-like cortical reactivity in the awake brain after focal injury. Brain. 2020;143:3672–84.PubMedPubMedCentral
78.
Zurück zum Zitat Rosanova M, Fecchio M, Casarotto S, Sarasso S, Casali AG, Pigorini A, et al. Sleep-like cortical OFF-periods disrupt causality and complexity in the brain of unresponsive wakefulness syndrome patients. Nat Commun. 2018;9:4427.PubMedPubMedCentral Rosanova M, Fecchio M, Casarotto S, Sarasso S, Casali AG, Pigorini A, et al. Sleep-like cortical OFF-periods disrupt causality and complexity in the brain of unresponsive wakefulness syndrome patients. Nat Commun. 2018;9:4427.PubMedPubMedCentral
79.
Zurück zum Zitat Johanson M, Revonsuo A, Chaplin J, Wedlund J-E. Level and contents of consciousness in connection with partial epileptic seizures. Epilepsy Behav. 2003;4:279–85.PubMed Johanson M, Revonsuo A, Chaplin J, Wedlund J-E. Level and contents of consciousness in connection with partial epileptic seizures. Epilepsy Behav. 2003;4:279–85.PubMed
80.
Zurück zum Zitat Johanson M, Valli K, Revonsuo A, Chaplin JE, Wedlund J-E. Alterations in the contents of consciousness in partial epileptic seizures. Epilepsy Behav. 2008;13:366–71.PubMed Johanson M, Valli K, Revonsuo A, Chaplin JE, Wedlund J-E. Alterations in the contents of consciousness in partial epileptic seizures. Epilepsy Behav. 2008;13:366–71.PubMed
81.
Zurück zum Zitat Laccheo I, Sonmezturk H, Bhatt AB, Tomycz L, Shi Y, Ringel M, et al. Non-convulsive status epilepticus and non-convulsive seizures in neurological ICU patients. Neurocrit Care. 2015;22:202–11.PubMed Laccheo I, Sonmezturk H, Bhatt AB, Tomycz L, Shi Y, Ringel M, et al. Non-convulsive status epilepticus and non-convulsive seizures in neurological ICU patients. Neurocrit Care. 2015;22:202–11.PubMed
82.
Zurück zum Zitat Mateos DM, Guevara Erra R, Wennberg R, Perez Velazquez JL. Measures of entropy and complexity in altered states of consciousness. Cogn Neurodyn. 2018;12:73–84.PubMed Mateos DM, Guevara Erra R, Wennberg R, Perez Velazquez JL. Measures of entropy and complexity in altered states of consciousness. Cogn Neurodyn. 2018;12:73–84.PubMed
83.
Zurück zum Zitat Kundishora AJ, Gummadavelli A, Ma C, Liu M, McCafferty C, Schiff ND, et al. Restoring conscious arousal during focal limbic seizures with deep brain stimulation. Cereb Cortex. 2017;27:1964–75.PubMed Kundishora AJ, Gummadavelli A, Ma C, Liu M, McCafferty C, Schiff ND, et al. Restoring conscious arousal during focal limbic seizures with deep brain stimulation. Cereb Cortex. 2017;27:1964–75.PubMed
84.
Zurück zum Zitat Pais-Roldán P, Edlow BL, Jiang Y, Stelzer J, Zou M, Yu X. Multimodal assessment of recovery from coma in a rat model of diffuse brainstem tegmentum injury. Neuroimage. 2019;189:615–30.PubMed Pais-Roldán P, Edlow BL, Jiang Y, Stelzer J, Zou M, Yu X. Multimodal assessment of recovery from coma in a rat model of diffuse brainstem tegmentum injury. Neuroimage. 2019;189:615–30.PubMed
85.
Zurück zum Zitat O’Donnell JC, Browne KD, Kilbaugh TJ, Chen HI, Whyte J, Cullen DK. Challenges and demand for modeling disorders of consciousness following traumatic brain injury. Neurosci Biobehav Rev. 2019;98:336–46.PubMed O’Donnell JC, Browne KD, Kilbaugh TJ, Chen HI, Whyte J, Cullen DK. Challenges and demand for modeling disorders of consciousness following traumatic brain injury. Neurosci Biobehav Rev. 2019;98:336–46.PubMed
86.
Zurück zum Zitat Wojnarowicz MW, Fisher AM, Minaeva O, Goldstein LE. Considerations for experimental animal models of concussion, traumatic brain injury, and chronic traumatic encephalopathy-these matters matter. Front Neurol. 2017;8:240.PubMedPubMedCentral Wojnarowicz MW, Fisher AM, Minaeva O, Goldstein LE. Considerations for experimental animal models of concussion, traumatic brain injury, and chronic traumatic encephalopathy-these matters matter. Front Neurol. 2017;8:240.PubMedPubMedCentral
87.
Zurück zum Zitat Brown EN, Purdon PL, Van Dort CJ. General anesthesia and altered states of arousal: a systems neuroscience analysis. Annu Rev Neurosci. 2011;34:601–28.PubMedPubMedCentral Brown EN, Purdon PL, Van Dort CJ. General anesthesia and altered states of arousal: a systems neuroscience analysis. Annu Rev Neurosci. 2011;34:601–28.PubMedPubMedCentral
88.
Zurück zum Zitat Proekt A, Hudson AE. A stochastic basis for neural inertia in emergence from general anaesthesia. Br J Anaesth. 2018;121:86–94.PubMedPubMedCentral Proekt A, Hudson AE. A stochastic basis for neural inertia in emergence from general anaesthesia. Br J Anaesth. 2018;121:86–94.PubMedPubMedCentral
89.
Zurück zum Zitat Friedman EB, Sun Y, Moore JT, Hung H-T, Meng QC, Perera P, et al. A conserved behavioral state barrier impedes transitions between anesthetic-induced unconsciousness and wakefulness: evidence for neural inertia. PLoS ONE. 2010;5:e11903. Friedman EB, Sun Y, Moore JT, Hung H-T, Meng QC, Perera P, et al. A conserved behavioral state barrier impedes transitions between anesthetic-induced unconsciousness and wakefulness: evidence for neural inertia. PLoS ONE. 2010;5:e11903.
90.
Zurück zum Zitat Pigorini A, Sarasso S, Proserpio P, Szymanski C, Arnulfo G, Casarotto S, et al. Bistability breaks-off deterministic responses to intracortical stimulation during non-REM sleep. Neuroimage. 2015;112:105–13.PubMed Pigorini A, Sarasso S, Proserpio P, Szymanski C, Arnulfo G, Casarotto S, et al. Bistability breaks-off deterministic responses to intracortical stimulation during non-REM sleep. Neuroimage. 2015;112:105–13.PubMed
91.
Zurück zum Zitat D’Andola M, Rebollo B, Casali AG, Weinert JF, Pigorini A, Villa R, et al. Bistability, causality, and complexity in cortical networks: an in vitro perturbational study. Cereb Cortex. 2018;28:2233–42.PubMed D’Andola M, Rebollo B, Casali AG, Weinert JF, Pigorini A, Villa R, et al. Bistability, causality, and complexity in cortical networks: an in vitro perturbational study. Cereb Cortex. 2018;28:2233–42.PubMed
92.
Zurück zum Zitat Dasilva M, Camassa A, Navarro-Guzman A, Pazienti A, Perez-Mendez L, Zamora-López G, et al. Modulation of cortical slow oscillations and complexity across anesthesia levels. Neuroimage. 2021;224:117415. Dasilva M, Camassa A, Navarro-Guzman A, Pazienti A, Perez-Mendez L, Zamora-López G, et al. Modulation of cortical slow oscillations and complexity across anesthesia levels. Neuroimage. 2021;224:117415.
94.
Zurück zum Zitat Taylor NE, Van Dort CJ, Kenny JD, Pei J, Guidera JA, Vlasov KY, et al. Optogenetic activation of dopamine neurons in the ventral tegmental area induces reanimation from general anesthesia. Proc Natl Acad Sci USA. 2016;113:12826–31.PubMedPubMedCentral Taylor NE, Van Dort CJ, Kenny JD, Pei J, Guidera JA, Vlasov KY, et al. Optogenetic activation of dopamine neurons in the ventral tegmental area induces reanimation from general anesthesia. Proc Natl Acad Sci USA. 2016;113:12826–31.PubMedPubMedCentral
95.
Zurück zum Zitat Reynaud O, da Silva AR, Gruetter R, Jelescu IO. Multi-slice passband bSSFP for human and rodent fMRI at ultra-high field. J Magn Reson. 2019;305:31–40.PubMed Reynaud O, da Silva AR, Gruetter R, Jelescu IO. Multi-slice passband bSSFP for human and rodent fMRI at ultra-high field. J Magn Reson. 2019;305:31–40.PubMed
96.
Zurück zum Zitat Satpute AB, Kragel PA, Barrett LF, Wager TD, Bianciardi M. Deconstructing arousal into wakeful, autonomic and affective varieties. Neurosci Lett. 2019;693:19–28.PubMed Satpute AB, Kragel PA, Barrett LF, Wager TD, Bianciardi M. Deconstructing arousal into wakeful, autonomic and affective varieties. Neurosci Lett. 2019;693:19–28.PubMed
97.
Zurück zum Zitat Redinbaugh MJ, Phillips JM, Kambi NA, Mohanta S, Andryk S, Dooley GL, et al. Thalamus modulates consciousness via layer-specific control of cortex. Neuron. 2020;106:66-75.e12.PubMedPubMedCentral Redinbaugh MJ, Phillips JM, Kambi NA, Mohanta S, Andryk S, Dooley GL, et al. Thalamus modulates consciousness via layer-specific control of cortex. Neuron. 2020;106:66-75.e12.PubMedPubMedCentral
98.
Zurück zum Zitat Bastos AM, Donoghue JA, Brincat SL, Mahnke M, Yanar J, Correa J, et al. Neural effects of propofol-induced unconsciousness and its reversal using thalamic stimulation. Elife. 2021;10:e60824. Bastos AM, Donoghue JA, Brincat SL, Mahnke M, Yanar J, Correa J, et al. Neural effects of propofol-induced unconsciousness and its reversal using thalamic stimulation. Elife. 2021;10:e60824.
99.
Zurück zum Zitat Palmiter RD. Dopamine signaling as a neural correlate of consciousness. Neuroscience. 2011;198:213–20.PubMed Palmiter RD. Dopamine signaling as a neural correlate of consciousness. Neuroscience. 2011;198:213–20.PubMed
100.
Zurück zum Zitat Brown EN, Pavone KJ, Naranjo M. Multimodal general anesthesia: theory and practice. Anesth Analg. 2018;127:1246–58.PubMedPubMedCentral Brown EN, Pavone KJ, Naranjo M. Multimodal general anesthesia: theory and practice. Anesth Analg. 2018;127:1246–58.PubMedPubMedCentral
101.
102.
Zurück zum Zitat Edlow BL, Takahashi E, Wu O, Benner T, Dai G, Bu L, et al. Neuroanatomic connectivity of the human ascending arousal system critical to consciousness and its disorders. J Neuropathol Exp Neurol. 2012;71:531–46.PubMed Edlow BL, Takahashi E, Wu O, Benner T, Dai G, Bu L, et al. Neuroanatomic connectivity of the human ascending arousal system critical to consciousness and its disorders. J Neuropathol Exp Neurol. 2012;71:531–46.PubMed
103.
Zurück zum Zitat Edlow BL, Haynes RL, Takahashi E, Klein JP, Cummings P, Benner T, et al. Disconnection of the ascending arousal system in traumatic coma. J Neuropathol Exp Neurol. 2013;72:505–23.PubMed Edlow BL, Haynes RL, Takahashi E, Klein JP, Cummings P, Benner T, et al. Disconnection of the ascending arousal system in traumatic coma. J Neuropathol Exp Neurol. 2013;72:505–23.PubMed
104.
Zurück zum Zitat van den Brink RL, Pfeffer T, Warren CM, Murphy PR, Tona K-D, van der Wee NJA, et al. Catecholaminergic neuromodulation shapes intrinsic MRI functional connectivity in the human brain. J Neurosci. 2016;36:7865–76.PubMedPubMedCentral van den Brink RL, Pfeffer T, Warren CM, Murphy PR, Tona K-D, van der Wee NJA, et al. Catecholaminergic neuromodulation shapes intrinsic MRI functional connectivity in the human brain. J Neurosci. 2016;36:7865–76.PubMedPubMedCentral
105.
Zurück zum Zitat Shine JM, Breakspear M, Bell PT, Ehgoetz Martens KA, Shine R, Koyejo O, et al. Human cognition involves the dynamic integration of neural activity and neuromodulatory systems. Nat Neurosci. 2019;22:289–96.PubMed Shine JM, Breakspear M, Bell PT, Ehgoetz Martens KA, Shine R, Koyejo O, et al. Human cognition involves the dynamic integration of neural activity and neuromodulatory systems. Nat Neurosci. 2019;22:289–96.PubMed
106.
Zurück zum Zitat Avery MC, Krichmar JL. Neuromodulatory systems and their interactions: a review of models, theories, and experiments. Front Neural Circuits. 2017;11:108.PubMedPubMedCentral Avery MC, Krichmar JL. Neuromodulatory systems and their interactions: a review of models, theories, and experiments. Front Neural Circuits. 2017;11:108.PubMedPubMedCentral
107.
108.
Zurück zum Zitat Ye R, Rua C, O’Callaghan C, Jones PS, Hezemans FH, Kaalund SS, et al. An in vivo probabilistic atlas of the human locus coeruleus at ultra-high field. Neuroimage. 2021;225:117487. Ye R, Rua C, O’Callaghan C, Jones PS, Hezemans FH, Kaalund SS, et al. An in vivo probabilistic atlas of the human locus coeruleus at ultra-high field. Neuroimage. 2021;225:117487.
109.
Zurück zum Zitat Solt K, Van Dort CJ, Chemali JJ, Taylor NE, Kenny JD, Brown EN. Electrical stimulation of the ventral tegmental area induces reanimation from general anesthesia. Anesthesiology. 2014;121:311–9.PubMed Solt K, Van Dort CJ, Chemali JJ, Taylor NE, Kenny JD, Brown EN. Electrical stimulation of the ventral tegmental area induces reanimation from general anesthesia. Anesthesiology. 2014;121:311–9.PubMed
110.
Zurück zum Zitat Taylor NE, Chemali JJ, Brown EN, Solt K. Activation of D1 dopamine receptors induces emergence from isoflurane general anesthesia. Anesthesiology. 2013;118:30–9.PubMed Taylor NE, Chemali JJ, Brown EN, Solt K. Activation of D1 dopamine receptors induces emergence from isoflurane general anesthesia. Anesthesiology. 2013;118:30–9.PubMed
111.
Zurück zum Zitat Edlow BL, Claassen J, Schiff ND, Greer DM. Recovery from disorders of consciousness: mechanisms, prognosis and emerging therapies. Nat Rev Neurol. 2021;17:135–56.PubMed Edlow BL, Claassen J, Schiff ND, Greer DM. Recovery from disorders of consciousness: mechanisms, prognosis and emerging therapies. Nat Rev Neurol. 2021;17:135–56.PubMed
112.
Zurück zum Zitat Solt K, Cotten JF, Cimenser A, Wong KFK, Chemali JJ, Brown EN. Methylphenidate actively induces emergence from general anesthesia. Anesthesiology. 2011;115:791–803.PubMed Solt K, Cotten JF, Cimenser A, Wong KFK, Chemali JJ, Brown EN. Methylphenidate actively induces emergence from general anesthesia. Anesthesiology. 2011;115:791–803.PubMed
113.
Zurück zum Zitat Fridman EA, Osborne JR, Mozley PD, Victor JD, Schiff ND. Presynaptic dopamine deficit in minimally conscious state patients following traumatic brain injury. Brain. 2019;142:1887–93.PubMedPubMedCentral Fridman EA, Osborne JR, Mozley PD, Victor JD, Schiff ND. Presynaptic dopamine deficit in minimally conscious state patients following traumatic brain injury. Brain. 2019;142:1887–93.PubMedPubMedCentral
114.
Zurück zum Zitat Aru J, Suzuki M, Larkum ME. Cellular mechanisms of conscious processing. Trends Cogn Sci. 2020;24:814–25.PubMed Aru J, Suzuki M, Larkum ME. Cellular mechanisms of conscious processing. Trends Cogn Sci. 2020;24:814–25.PubMed
115.
Zurück zum Zitat Suzuki M, Larkum ME. General anesthesia decouples cortical pyramidal neurons. Cell. 2020;180:666-76.e13.PubMed Suzuki M, Larkum ME. General anesthesia decouples cortical pyramidal neurons. Cell. 2020;180:666-76.e13.PubMed
116.
Zurück zum Zitat Craig MM, Misic B, Pappas I, Adapa RM, Menon DK, Stamatakis EA. Propofol sedation-induced alterations in brain connectivity reflect parvalbumin interneurone distribution in human cerebral cortex. Br J Anaesth. 2021;126:835–44.PubMed Craig MM, Misic B, Pappas I, Adapa RM, Menon DK, Stamatakis EA. Propofol sedation-induced alterations in brain connectivity reflect parvalbumin interneurone distribution in human cerebral cortex. Br J Anaesth. 2021;126:835–44.PubMed
118.
Zurück zum Zitat Wu X, Zhang J, Cui Z, Tang W, Shao C, Hu J, et al. White matter deficits underlying the impaired consciousness level in patients with disorders of consciousness. Neurosci Bull. 2018;34:668–78.PubMedPubMedCentral Wu X, Zhang J, Cui Z, Tang W, Shao C, Hu J, et al. White matter deficits underlying the impaired consciousness level in patients with disorders of consciousness. Neurosci Bull. 2018;34:668–78.PubMedPubMedCentral
119.
Zurück zum Zitat Stafford CA, Owen AM, Fernández-Espejo D. The neural basis of external responsiveness in prolonged disorders of consciousness. Neuroimage Clin. 2019;22:101791. Stafford CA, Owen AM, Fernández-Espejo D. The neural basis of external responsiveness in prolonged disorders of consciousness. Neuroimage Clin. 2019;22:101791.
120.
Zurück zum Zitat Karve IP, Taylor JM, Crack PJ. The contribution of astrocytes and microglia to traumatic brain injury. Br J Pharmacol. 2016;173:692–702.PubMed Karve IP, Taylor JM, Crack PJ. The contribution of astrocytes and microglia to traumatic brain injury. Br J Pharmacol. 2016;173:692–702.PubMed
121.
Zurück zum Zitat Grajauskas LA, Frizzell T, Song X, D’Arcy RCN. White matter fMRI activation cannot be treated as a nuisance regressor: overcoming a historical blind spot. Front Neurosci. 2019;13:1024.PubMedPubMedCentral Grajauskas LA, Frizzell T, Song X, D’Arcy RCN. White matter fMRI activation cannot be treated as a nuisance regressor: overcoming a historical blind spot. Front Neurosci. 2019;13:1024.PubMedPubMedCentral
122.
Zurück zum Zitat Takata N, Sugiura Y, Yoshida K, Koizumi M, Hiroshi N, Honda K, et al. Optogenetic astrocyte activation evokes BOLD fMRI response with oxygen consumption without neuronal activity modulation. Glia. 2018;66:2013–23.PubMed Takata N, Sugiura Y, Yoshida K, Koizumi M, Hiroshi N, Honda K, et al. Optogenetic astrocyte activation evokes BOLD fMRI response with oxygen consumption without neuronal activity modulation. Glia. 2018;66:2013–23.PubMed
123.
Zurück zum Zitat Simon DW, McGeachy MJ, Bayır H, Clark RSB, Loane DJ, Kochanek PM. The far-reaching scope of neuroinflammation after traumatic brain injury. Nat Rev Neurol. 2017;13:171–91.PubMedPubMedCentral Simon DW, McGeachy MJ, Bayır H, Clark RSB, Loane DJ, Kochanek PM. The far-reaching scope of neuroinflammation after traumatic brain injury. Nat Rev Neurol. 2017;13:171–91.PubMedPubMedCentral
124.
Zurück zum Zitat Scott G, Hellyer PJ, Ramlackhansingh AF, Brooks DJ, Matthews PM, Sharp DJ. Thalamic inflammation after brain trauma is associated with thalamo-cortical white matter damage. J Neuroinflammation. 2015;12:224.PubMedPubMedCentral Scott G, Hellyer PJ, Ramlackhansingh AF, Brooks DJ, Matthews PM, Sharp DJ. Thalamic inflammation after brain trauma is associated with thalamo-cortical white matter damage. J Neuroinflammation. 2015;12:224.PubMedPubMedCentral
125.
Zurück zum Zitat Corps KN, Roth TL, McGavern DB. Inflammation and neuroprotection in traumatic brain injury. JAMA Neurol. 2015;72:355–62.PubMedPubMedCentral Corps KN, Roth TL, McGavern DB. Inflammation and neuroprotection in traumatic brain injury. JAMA Neurol. 2015;72:355–62.PubMedPubMedCentral
127.
Zurück zum Zitat Theparambil SM, Hosford PS, Ruminot I, Kopach O, Reynolds JR, Sandoval PY, et al. Astrocytes regulate brain extracellular pH via a neuronal activity-dependent bicarbonate shuttle. Nat Commun. 2020;11:5073.PubMedPubMedCentral Theparambil SM, Hosford PS, Ruminot I, Kopach O, Reynolds JR, Sandoval PY, et al. Astrocytes regulate brain extracellular pH via a neuronal activity-dependent bicarbonate shuttle. Nat Commun. 2020;11:5073.PubMedPubMedCentral
128.
Zurück zum Zitat Zeiler FA, McFadyen C, Newcombe VFJ, Synnot A, Donoghue EL, Ripatti S, et al. Genetic influences on patient-oriented outcomes in traumatic brain injury: a living systematic review of non-apolipoprotein e single-nucleotide polymorphisms. J Neurotrauma. 2021;38:1107–23.PubMedPubMedCentral Zeiler FA, McFadyen C, Newcombe VFJ, Synnot A, Donoghue EL, Ripatti S, et al. Genetic influences on patient-oriented outcomes in traumatic brain injury: a living systematic review of non-apolipoprotein e single-nucleotide polymorphisms. J Neurotrauma. 2021;38:1107–23.PubMedPubMedCentral
129.
Zurück zum Zitat Wainberg M, Sinnott-Armstrong N, Mancuso N, Barbeira AN, Knowles DA, Golan D, et al. Opportunities and challenges for transcriptome-wide association studies. Nat Genet. 2019;51:592–9.PubMedPubMedCentral Wainberg M, Sinnott-Armstrong N, Mancuso N, Barbeira AN, Knowles DA, Golan D, et al. Opportunities and challenges for transcriptome-wide association studies. Nat Genet. 2019;51:592–9.PubMedPubMedCentral
130.
Zurück zum Zitat Chatelle C, Thibaut A, Whyte J, De Val MD, Laureys S, Schnakers C. Pain issues in disorders of consciousness. Brain Inj. 2014;28:1202–8.PubMed Chatelle C, Thibaut A, Whyte J, De Val MD, Laureys S, Schnakers C. Pain issues in disorders of consciousness. Brain Inj. 2014;28:1202–8.PubMed
131.
Zurück zum Zitat Varley TF, Luppi AI, Pappas I, Naci L, Adapa R, Owen AM, et al. Consciousness and brain functional complexity in propofol anaesthesia. Sci Rep. 2020;10:1018.PubMedPubMedCentral Varley TF, Luppi AI, Pappas I, Naci L, Adapa R, Owen AM, et al. Consciousness and brain functional complexity in propofol anaesthesia. Sci Rep. 2020;10:1018.PubMedPubMedCentral
132.
Zurück zum Zitat Liu X, Lauer KK, Ward BD, Roberts CJ, Liu S, Gollapudy S, et al. Regional entropy of functional imaging signals varies differently in sensory and cognitive systems during propofol-modulated loss and return of behavioral responsiveness. Brain Imaging Behav. 2019;13:514–25.PubMedPubMedCentral Liu X, Lauer KK, Ward BD, Roberts CJ, Liu S, Gollapudy S, et al. Regional entropy of functional imaging signals varies differently in sensory and cognitive systems during propofol-modulated loss and return of behavioral responsiveness. Brain Imaging Behav. 2019;13:514–25.PubMedPubMedCentral
133.
Zurück zum Zitat Schartner M, Seth A, Noirhomme Q, Boly M, Bruno M-A, Laureys S, et al. Complexity of multi-dimensional spontaneous EEG decreases during propofol induced general anaesthesia. PLoS ONE. 2015;10:e0133532. Schartner M, Seth A, Noirhomme Q, Boly M, Bruno M-A, Laureys S, et al. Complexity of multi-dimensional spontaneous EEG decreases during propofol induced general anaesthesia. PLoS ONE. 2015;10:e0133532.
134.
Zurück zum Zitat Pal D, Li D, Dean JG, Brito MA, Liu T, Fryzel AM, et al. Level of consciousness is dissociable from electroencephalographic measures of cortical connectivity, slow oscillations, and complexity. J Neurosci. 2020;40:605–18.PubMedPubMedCentral Pal D, Li D, Dean JG, Brito MA, Liu T, Fryzel AM, et al. Level of consciousness is dissociable from electroencephalographic measures of cortical connectivity, slow oscillations, and complexity. J Neurosci. 2020;40:605–18.PubMedPubMedCentral
135.
Zurück zum Zitat Castelvecchi D. Can we open the black box of AI? Nature. 2016;538:20–3.PubMed Castelvecchi D. Can we open the black box of AI? Nature. 2016;538:20–3.PubMed
136.
Zurück zum Zitat Oizumi M, Albantakis L, Tononi G. From the phenomenology to the mechanisms of consciousness: Integrated Information Theory 3.0. PLoS Comput Biol. 2014;10:e1003588. Oizumi M, Albantakis L, Tononi G. From the phenomenology to the mechanisms of consciousness: Integrated Information Theory 3.0. PLoS Comput Biol. 2014;10:e1003588.
137.
Zurück zum Zitat Tononi G, Edelman GM. Consciousness and complexity. Science. 1998;282:1846–51.PubMed Tononi G, Edelman GM. Consciousness and complexity. Science. 1998;282:1846–51.PubMed
139.
Zurück zum Zitat Tononi G, Boly M, Massimini M, Koch C. Integrated information theory: from consciousness to its physical substrate. Nat Rev Neurosci. 2016;17:450–61.PubMed Tononi G, Boly M, Massimini M, Koch C. Integrated information theory: from consciousness to its physical substrate. Nat Rev Neurosci. 2016;17:450–61.PubMed
140.
Zurück zum Zitat Baars BJ. Global workspace theory of consciousness: toward a cognitive neuroscience of human experience. Prog Brain Res. 2005;150:45–53.PubMed Baars BJ. Global workspace theory of consciousness: toward a cognitive neuroscience of human experience. Prog Brain Res. 2005;150:45–53.PubMed
141.
Zurück zum Zitat Dehaene S, Changeux J-P. Experimental and theoretical approaches to conscious processing. Neuron. 2011;70:200–27.PubMed Dehaene S, Changeux J-P. Experimental and theoretical approaches to conscious processing. Neuron. 2011;70:200–27.PubMed
142.
Zurück zum Zitat Mashour GA, Roelfsema P, Changeux J-P, Dehaene S. Conscious processing and the global neuronal workspace hypothesis. Neuron. 2020;105:776–98.PubMedPubMedCentral Mashour GA, Roelfsema P, Changeux J-P, Dehaene S. Conscious processing and the global neuronal workspace hypothesis. Neuron. 2020;105:776–98.PubMedPubMedCentral
143.
Zurück zum Zitat Barttfeld P, Bekinschtein TA, Salles A, Stamatakis EA, Adapa R, Menon DK, et al. Factoring the brain signatures of anesthesia concentration and level of arousal across individuals. Neuroimage Clin. 2015;9:385–91.PubMedPubMedCentral Barttfeld P, Bekinschtein TA, Salles A, Stamatakis EA, Adapa R, Menon DK, et al. Factoring the brain signatures of anesthesia concentration and level of arousal across individuals. Neuroimage Clin. 2015;9:385–91.PubMedPubMedCentral
144.
Zurück zum Zitat Vatansever D, Schröter M, Adapa RM, Bullmore ET, Menon DK, Stamatakis EA. Reorganisation of brain hubs across altered states of consciousness. Sci Rep. 2020;10:3402.PubMedPubMedCentral Vatansever D, Schröter M, Adapa RM, Bullmore ET, Menon DK, Stamatakis EA. Reorganisation of brain hubs across altered states of consciousness. Sci Rep. 2020;10:3402.PubMedPubMedCentral
145.
Zurück zum Zitat Boly M, Massimini M, Tsuchiya N, Postle BR, Koch C, Tononi G. Are the neural correlates of consciousness in the front or in the back of the cerebral cortex? Clinical and neuroimaging evidence. J Neurosci. 2017;37:9603–13.PubMedPubMedCentral Boly M, Massimini M, Tsuchiya N, Postle BR, Koch C, Tononi G. Are the neural correlates of consciousness in the front or in the back of the cerebral cortex? Clinical and neuroimaging evidence. J Neurosci. 2017;37:9603–13.PubMedPubMedCentral
146.
Zurück zum Zitat Safron A. An integrated world modeling theory (IWMT) of consciousness: combining integrated information and global neuronal workspace theories with the free energy principle and active inference framework; toward solving the hard problem and characterizing agentic causation. Front Artif Intell. 2020;3:30.PubMedPubMedCentral Safron A. An integrated world modeling theory (IWMT) of consciousness: combining integrated information and global neuronal workspace theories with the free energy principle and active inference framework; toward solving the hard problem and characterizing agentic causation. Front Artif Intell. 2020;3:30.PubMedPubMedCentral
148.
Zurück zum Zitat Berlingeri M, Magnani FG, Salvato G, Rosanova M, Bottini G. Neuroimaging studies on disorders of consciousness: a meta-analytic evaluation. J Clin Med. 2019;8:516.PubMedCentral Berlingeri M, Magnani FG, Salvato G, Rosanova M, Bottini G. Neuroimaging studies on disorders of consciousness: a meta-analytic evaluation. J Clin Med. 2019;8:516.PubMedCentral
149.
Zurück zum Zitat Ching S, Purdon PL, Vijayan S, Kopell NJ, Brown EN. A neurophysiological-metabolic model for burst suppression. Proc Natl Acad Sci USA. 2012;109:3095–100.PubMedPubMedCentral Ching S, Purdon PL, Vijayan S, Kopell NJ, Brown EN. A neurophysiological-metabolic model for burst suppression. Proc Natl Acad Sci USA. 2012;109:3095–100.PubMedPubMedCentral
150.
Zurück zum Zitat Ching S, Cimenser A, Purdon PL, Brown EN, Kopell NJ. Thalamocortical model for a propofol-induced alpha-rhythm associated with loss of consciousness. Proc Natl Acad Sci USA. 2010;107:22665–70.PubMedPubMedCentral Ching S, Cimenser A, Purdon PL, Brown EN, Kopell NJ. Thalamocortical model for a propofol-induced alpha-rhythm associated with loss of consciousness. Proc Natl Acad Sci USA. 2010;107:22665–70.PubMedPubMedCentral
151.
Zurück zum Zitat Vijayan S, Ching S, Purdon PL, Brown EN, Kopell NJ. Thalamocortical mechanisms for the anteriorization of α rhythms during propofol-induced unconsciousness. J Neurosci. 2013;33:11070–5.PubMedPubMedCentral Vijayan S, Ching S, Purdon PL, Brown EN, Kopell NJ. Thalamocortical mechanisms for the anteriorization of α rhythms during propofol-induced unconsciousness. J Neurosci. 2013;33:11070–5.PubMedPubMedCentral
Metadaten
Titel
Mechanisms Underlying Disorders of Consciousness: Bridging Gaps to Move Toward an Integrated Translational Science
verfasst von
Andrea I. Luppi
Joshua Cain
Lennart R. B. Spindler
Urszula J. Górska
Daniel Toker
Andrew E. Hudson
Emery N. Brown
Michael N. Diringer
Robert D. Stevens
Marcello Massimini
Martin M. Monti
Emmanuel A. Stamatakis
Melanie Boly
the Curing Coma Campaign and Its Contributing Collaborators
Publikationsdatum
01.07.2021
Verlag
Springer US
Erschienen in
Neurocritical Care / Ausgabe Sonderheft 1/2021
Print ISSN: 1541-6933
Elektronische ISSN: 1556-0961
DOI
https://doi.org/10.1007/s12028-021-01281-6

Weitere Artikel der Sonderheft 1/2021

Neurocritical Care 1/2021 Zur Ausgabe

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Nicht Creutzfeldt Jakob, sondern Abführtee-Vergiftung

29.05.2024 Hyponatriämie Nachrichten

Eine ältere Frau trinkt regelmäßig Sennesblättertee gegen ihre Verstopfung. Der scheint plötzlich gut zu wirken. Auf Durchfall und Erbrechen folgt allerdings eine Hyponatriämie. Nach deren Korrektur kommt es plötzlich zu progredienten Kognitions- und Verhaltensstörungen.

Schutz der Synapsen bei Alzheimer

29.05.2024 Morbus Alzheimer Nachrichten

Mit einem Neurotrophin-Rezeptor-Modulator lässt sich möglicherweise eine bestehende Alzheimerdemenz etwas abschwächen: Erste Phase-2-Daten deuten auf einen verbesserten Synapsenschutz.

Sozialer Aufstieg verringert Demenzgefahr

24.05.2024 Demenz Nachrichten

Ein hohes soziales Niveau ist mit die beste Versicherung gegen eine Demenz. Noch geringer ist das Demenzrisiko für Menschen, die sozial aufsteigen: Sie gewinnen fast zwei demenzfreie Lebensjahre. Umgekehrt steigt die Demenzgefahr beim sozialen Abstieg.

Hirnblutung unter DOAK und VKA ähnlich bedrohlich

17.05.2024 Direkte orale Antikoagulanzien Nachrichten

Kommt es zu einer nichttraumatischen Hirnblutung, spielt es keine große Rolle, ob die Betroffenen zuvor direkt wirksame orale Antikoagulanzien oder Marcumar bekommen haben: Die Prognose ist ähnlich schlecht.

Update Neurologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.