You are on page 1of 17

Emerging Frontiers of Neuroengineering: A Network Science of Brain Connectivity

Danielle S. Bassett1,2 , Ankit N. Khambhati1 , and Scott T. Grafton3,4


1
Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104
2
Department of Electrical and Systems Engineering,
University of Pennsylvania, Philadelphia, PA, 19104
3
UCSB Brain Imaging Center and Department of Psychological and Brain Sciences,
University of California, Santa Barbara, CA 93106 USA and
4
Institute for Collaborative Biotechnologies, University of California, Santa Barbara, CA 93106 USA
(Dated: December 26, 2016)
Neuroengineering is faced with unique challenges in repairing or replacing complex neural systems
that are composed of many interacting parts. These interactions form intricate patterns over large
spatiotemporal scales, and produce emergent behaviors that are difficult to predict from individual
arXiv:1612.08059v1 [q-bio.NC] 23 Dec 2016

elements. Network science provides a particularly appropriate framework in which to study and
intervene in such systems, by treating neural elements (cells, volumes) as nodes in a graph and
neural interactions (synapses, white matter tracts) as edges in that graph. Here, we review the
emerging discipline of network neuroscience, which uses and develops tools from graph theory to
better understand and manipulate neural systems, from micro- to macroscales. We present examples
of how human brain imaging data is being modeled with network analysis and underscore potential
pitfalls. We then highlight current computational and theoretical frontiers, and emphasize their
utility in informing diagnosis and monitoring, brain-machine interfaces, and brain stimulation. A
flexible and rapidly evolving enterprise, network neuroscience provides a set of powerful approaches
and fundamental insights critical to the neuroengineers toolkit.

Could we graft new connections into the brain, to give (genes, neurons, columns, areas) that are related to one
someone back the abilities they had pre-injury [1]? Could another in many different ways (anatomical connections,
we decode the thoughts of someone who is caged inside functional relationships, material similarities) [15, 16]
their own body [2, 3]? Could we develop adaptive brain- (Fig. 1). These relationships form intricate patterns –
computer interfaces that evolve and adapt to remain ef- of synaptic connections, gene co-expression, connectome
fective for a child whose brain is continuously develop- fingerprints – that may differ across species [17, 18], or
ing [4, 5]? Answering these and many other seemingly across cohorts within a single species (e.g., in health ver-
over-ambitious questions is the fundamental aim of neu- sus disease) [19, 20]. From these patterns stem the very
roengineering [6], a relatively new domain of biomed- complicated phenomena of development, behavior, and
ical engineering that develops and uses computational cognition [21, 22].
and empirical techniques to understand and modulate Biological patterns like these are particularly difficult
the properties of neural systems. Particularly exciting to study for several reasons. First, the governing princi-
frontiers of neuroengineering include neuroimaging, neu- ples of pattern formation are often difficult to infer [23–
ral interfaces, neural prosthetics and robotics, and more 25], and thus mechanistic insights are difficult to come by.
general techniques for regeneration, enhancement, and Second, it is difficult to simplify patterns using coarse-
refinement of neural systems [7]. graining or other dimensionality reduction approaches
In the era of big data, neural systems are no exception while still maintaining the richness of the neurophysio-
to the rule of ever-increasing petabytes streaming in to logically relevant information [10]. While retaining nec-
servers around the world [8]. However, in many other essary information while simplifying the patterns is dif-
arenas, the amount of data being gathered has not posed ficult, so is studying each element in the pattern: with
an insurmountable obstacle. What is the fundamental thousands and sometimes millions of elements, the set of
difference that causes neuroscientific data to be so chal- interactions between them – particularly if they evolve
lenging? Is it a lack of a mechanistic understanding of in time – quickly becomes enormous and complicated.
how the brain works [9, 10]? Or an inability to physi- Indeed, as many fields have now come to realize, the in-
cally construct the hardware required to liaise with neu- tricacies of relational data call for a new conceptual and
ral systems for effective interventions [5]? We argue that mathematical framework [26, 27].
fundamental to both of these problems is the challenge
of dealing with complex relational data [11]. In develop-
ing a data science to meet these rising demands [12], we THE PECULIAR APPROPRIATENESS OF
must acknowledge that these data are far from indepen- NETWORK SCIENCE
dent: instead, data from neural systems are inherently
relational data [13]. Network science is an emerging interdisciplinary field
Relational data can be defined as any data that cod- that combines theories from statistical mechanics, com-
ifies relationships between elements [14]. The nervous putational techniques from computer science, statistics,
system is composed of units across many spatial scales applied mathematics, and visualization approaches to
2

How does one go about translating neural systems into


the language of network science [19, 37–43]? The first
critical step is to determine which constituent elements
are the fundamental unit of interest that is measurable
in the particular experiment under consideration [44].
These elements – which might be single neurons, neu-
ronal ensembles, genes, or large-scale brain areas – will
be treated as nodes or vertices in the network [45]. Then,
one must define the connections, interactions, or relation-
ships of interest between network nodes. These links –
which might be white matter tracts between large-scale
brain areas, chemical or electrical synapses between neu-
rons, or co-expression patterns among genes – will be
treated as network edges. Once nodes and edges have
been defined, the network itself – the pattern of edges
linking nodes – can be studied from the point of view of
FIG. 1. Relational data in biological systems. Repeating a graph in mathematics [46, 47].
genotypic and phenotypic patterns emerge frequently in the
study of biological systems. These biological patterns are ex-
pressed across multiple scales of granularity. Illustrated here THE MATHEMATICS OF NETWORK SCIENCE
are three different scales of biological elements (behavioral, IN NEURAL SYSTEMS
structural, genetic) in different animal species, with lines rep-
resenting conceptual relationships between elements. At the
macro-scale, we observe behavioral similarities across differ- In the field of mathematics, a graph G = (V, E) is com-
ent species, such as the ability to fly in birds and fruit flies. posed of a node set V and an edge set E [46, 47]. We store
However, a closer lens on the neurological substrate of this this information in an adjacency matrix A, whose ele-
behavior may tell a different story: that meso-scale struc- ments indicate the strength of edges between nodes. The
tural brain architecture differs significantly between birds and representation of data in a graph enables the investigator
fruit flies, and is more similar between insects (e.g. fruit flies to characterize the patterns of connectivity locally sur-
and ants) and between mammals (e.g. mice and cats). De-
rounding a single node or globally taking into account all
spite differences in structural brain architecture, we might
find that animals of different species share commonalities in edges. In addition to local and global structure, tools are
genetic code that manifest similarly in physical attributes. available to probe so-called mesoscale structure in the
While differences in each element yield unique qualities to graph, which can be defined as structure that is present
each individual animal species, examining relational data can at intermediate length scales in the system (Fig. 2).
provide a more comprehensive view on the functional role of To give the reader some simple intuitions, we briefly
each element ecologically. describe examples of local, meso-scale, and global statis-
tics that can be computed from graphs of neural systems.
First, a common local statistic that has proven particu-
larly effective in characterizing neural systems is the clus-
tering coefficient of a node, which can be defined as the
probe, perturb, and predict the behavior of complex sys- fraction of a node’s neighbors that are also connected to
tems in technology, biology, and sociology [28]. While one another [48]. In essence, this statistic is sensitive to
historically developed to understand social interactions the density of triangles in the graph, and is thought to
or friendship webs similar to those supported (or elicited) play a non-trivial role in local information processing in
by Facebook or Twitter, network science is a peculiarly neural systems [49] (although for caveats see [50, 51]). A
appropriate framework in which to tackle the challenges common global statistic that has proven useful in charac-
of neural data sciences to better engineer artificial and terizing neural systems is the characteristic path length,
natural neural systems. which is defined as the average shortest path between
In particular, rather than reducing complex relational all possible node pairs [28]. This statistic is sensitive to
data to a list of independent parts, network science pro- long-distance connections that provide short cuts from
vides tools to explicitly characterize the pattern of in- one side of the network to another, and is thought to play
teractions between neural elements [29]. In addition to a role in the swift transmission of information across the
these descriptive tools, it provides benchmark graphical system [43]. Interestingly, early work demonstrated that
models for statistical comparison and inference [30–32], humans displaying brain wiring patterns with shorter
mathematical models to quantify and predict the flow of characteristic path length also had higher IQ than those
information [33] or communication [34] through neural with longer characteristic path length [52], suggesting the
circuits, and predictive tools to forecast how networks sensitivity of network statistics to architectures that sup-
might change in response to injury [35] or therapeutic port healthy human cognitive function. However, it is
interventions [36]. worth noting that short characteristic path lengths do
3

not appear to be the full story [42, 53], and measures of nodes also known as modules; the nodes in a module are
segregated information processing also play an important more densely connected to other nodes in the same mod-
role in brain function [54]. ule than to nodes in other modules [55, 56]. This modular
architecture is thought to support specialization of func-
tion, each module performing a different role in support
of neurophysiological processes from synchronization to
cognition [54]. In contrast, a network with core-periphery
structure contains a set of core nodes that are densely in-
terconnected with all other nodes in the network, and a
set of periphery nodes that are sparsely interconnected
with all other nodes in the network [57, 58]. This or-
ganization is thought to support the integration of in-
formation across neuronal assemblies, neural circuits, or
large-scale functional modules [59], in each of which the
top-down web os often referred to as a rich club [60].
The multiscale nature of these network tools are par-
ticularly useful for neural systems, which are thought
to perform inherently different computations at differ-
ent levels of the network hierarchy [61]. For example,
information is thought to be processed in local cortical
areas before being passed across modules along rich-club
edges (in a so-called small-world [42, 53]), allowing inte-
grative computations and coherent behavioral responses
[62, 63]. Understanding this multiscale architecture and
FIG. 2. Multi-scale topology in brain networks. Brain its functional role in neural system dynamics is critical
networks express fundamental organizing principles across for developing effective interventions that capitalize on
multiple spatial scales. Brain networks are modeled as a existing structure and dynamic properties rather than
collection of nodes – representing regions of interest with fighting against them.
presumably coherent functional responsibilities – and edges
– structural connections or functional interactions between
brain regions. (A) Node centrality describes the importance
of individual nodes in terms of their connectivity relative to HOW DO WE BUILD BRAIN NETWORKS?
other nodes in the network. Nodes with more connections or
stronger edges tend to be hubs (red), while nodes with less
Using the mathematical tools of network science to
connections tend to be isolated (blue). (B) Clustering coef-
ficient, a measure of connectivity between the neighbors of
understand neural data requires one to explicitly build
a node, is another local measure of network topology. Un- network models. How does one go about doing so? This
like network topologies with strong hubness qualities, as in topic fully warrants a review of its own: describing meth-
A, networks with strong clustering coefficient demonstrate a ods to build brain networks from spiking data [64], cal-
high density of triangles that is believed to facilitate local cium transients and microelectrode arrays [65], mesoscale
information processing. (C) Modularity is a meso-scale topo- tract tracing [53], genetic expression [66], and large-scale
logical property that captures communities of nodes that are neuroimaging [43] and across species from cat [67] and
tightly connected to one another and weakly connected to macaque [68], to C. elegans [62], mouse [69], rat [70],
nodes in other communities. Modular organization underlies drosophila [18, 71], and human [72] (to offer a sparse
a rich functional specialization within individual communi- list!). Because we cannot do justice to the full richness of
ties. Here, nodes of different communities are colored red,
this question here, we focus our presentation on human
blue, or pink. (D) Networks with core-periphery structure
exhibit a set of tightly-connected nodes (core; red) sparsely brain imaging data, which has historically provided the
connected to a set of isolated nodes (periphery; blue). This largest source of data for testing the utility of network
organization is in stark contrast to the modular organization science to characterize complex neural systems. Thou-
in C. The core-periphery architecture is characteristic in net- sands of healthy subjects and patient populations have
works that integrate information from isolated regions in a been scanned, primarily by magnetic resonance imaging
central area. to identify both structural and functional properties of
the nervous system. Given the dominant role of imag-
ing, here we familiarize the reader with the strategies
used to transform brain scans into data structures that
In addition to local and global structure, meso-scale are amenable to network analysis. However, we empha-
organization provides a window into the properties of size that the network tools we describe are fully translat-
groups of nodes. Two common mesoscale structures able to other neural systems, and are commonly applied
are modularity and core-periphery structure. A network in EEG [73–75], MEG [76], ECOG [77, 78], and fNIRS
with modular structure is one that contains groups of [79, 80] as well (for a more thorough review of these ap-
4

plication areas, see [13]). across a range of atlases or spatial resolutions.


The most common measurements of human brain con-
nectivity, whether they are functional or structural net-
works, rely on scans obtained by magnetic resonance WHAT DO BRAIN NETWORKS OFFER
imaging (MRI). There are three basic types of brain scans NEUROENGINEERING?
that are typically used in network construction. The first
is an “anatomic” scan. This is a T1 weighted, high res- After building networks from imaging data, one can
olution (<1mm isotropic) sequence that can distinguish then use these networks to address pressing questions in
gray matter from underlying white matter. Many soft- neuroengineering. While we cannot exhaustively cover
ware tools are available for segmenting these two types all possible uses of these tools currently in the litera-
of tissues and for partitioning the gray matter into a set ture, here we highlight their utility in neural mapping
of local regions that form the network nodes, as shown in and connectivity estimation, diagnosis and monitoring,
Fig. 3. There are many atlases available for partitioning and rehabilitation and treatment.
the gray matter, varying from ∼50-1000 separate regions.
The second type of scan is a “functional” MRI. This is a
series of T2* weighted scans acquired at sampling rates Diagnosis and monitoring
as fast as 2.21 Hz (although more typically at 0.5 Hz) and
at a lower spatial resolution (∼3mm non-isotropic). This
tissue contrast is sensitive to changes in blood oxygen Accurately diagnosing disorders of the human con-
level dependent (BOLD) signals, which vary as a func- nectome and monitoring their progression are particu-
tion of cortical activity (whether neuronal or synaptic). larly critical applications of network-based tools to neu-
These scans can be acquired with the subject at rest, ral systems. Diseases thought to be accompanied by con-
the so-called “resting state” MRI or while performing a nectome abnormalities or alterations include schizophre-
particular task, the so-called “task based” fMRI [81, 82]. nia [76, 84, 85], autism [86, 87], epilepsy [78, 88], and
The time series of brain activity, averaged across all vox- Alzheimer’s disease [89–91], among others [19, 92]. The
els in each local region can then be extracted. To create pattern of alterations in a given condition can be de-
an adjacency matrix reflecting functional connectivity, scribed in the form of a graph, as can the patterns that
pairs of time series are related by correlation, partial cor- are similar or different between a pair of conditions.
relation, wavelet filtered correlation or coherence within In some cases, these network changes occur early in a
a particular frequency band. disease, offering potential as diagnostic biomarkers [93].
Bolstering this possibility, several studies have demon-
The third method of imaging is based on diffusion
strated that by incorporating network statistics as fea-
imaging. This involves the acquisition of a set of scans,
tures in machine learning algorithms, it is possible to
each of which is sensitive to the magnitude of water dif-
classify groups of individuals with and without a partic-
fusion in a particular direction in 3-dimensions. The set
ular condition, from aging [94] to major depression [95].
of oriented diffusion scans are then combined to estimate
While diagnosis and classification are binary decisions,
voxel-wise distributions of water diffusion [83]. The brain
one can also continuously monitor brain networks within
is then seeded uniformly at subvoxel resolution and a
a single individual [96] either during drastic changes such
probable path of diffusion through the full volume is cal-
as those accompanying disease progression, or during mi-
culated, resulting in a virtual tract of diffusion, referred
nor changes in mental state such as those induced by
to as a “streamline”. These streamlines are virtual es-
driving fatigue [97].
timates of possible water diffusion that can correspond
to true white matter fascicles or tracts. The white mat-
ter fascicles and tracts are thought to be the primary
means for information sharing between distinct gray mat- Rehabilitation and treatment
ter regions, analogous to wiring that connects distinct
computer modules [63, 72]. To create an adjacency ma- The sensitivity of network measures to brain state of-
trix reflecting this structural connectivity, the strength of fers the generalizable potential for graph statistics to be
connectivity between pairs of regions can be estimated by used as indicators of the efficacy of rehabilitation and
the number of streamlines, the density of streamlines or treatment. Initial studies support this potential efficacy
the number of streamlines normalized by the length they by demonstrating appreciable changes in network orga-
traverse. The set of all real connections in the brain is nization induced by memory rehabilitation treatment (a
referred to as the human connectome. While it is not broad intervention useful across multiple clinical condi-
yet possible to characterize this full set of connections, tions [98]), seizure therapy (an intervention for severe
it can be approximated by the streamlines reconstructed depression [73]), and motor imagery (a frequent inter-
with diffusion imagng. This lower dimensional connec- vention for stroke [99]). Common techniques to affect
tome can then be characterized with the tools of network these interventions include neurofeedback where humans
science. Given that it is an approximation, it is valuable learn to control the activity or connectivity in certain
to test the robustness of any particular network property parts of their brain to enhance mental function [100, 101].
5

FIG. 3. Constructing Connectomes from MRI Data. To generate human connectomes with magnetic resonance imaging,
an anatomic scan delineating gray matter is partitioned into a set of nodes. This is combined with either diffusion scans of white
matter structural connections or time series of brain activity measured by functional MRI, resulting in a weighted connectivity
matrix.

Notably, graph statistics of functional network architec- standing necessary to intervene in a way that benefits
ture have proven sensitive to cognitive workload dur- the system. A particularly exciting current frontier in
ing these interventions, offering task-independent mark- neuroengineering lies in mapping neural systems using a
ers for monitoring and matching participants ability and variety of imaging techniques [106], and in estimating the
task difficulty during neurofeedback training [102]. Neu- connectivity between neural elements using sophisticated
rofeedback approaches often utilize exquisitely calibrated statistical algorithms [107]. In these contexts, network
brain-computer interfaces [103], systems that can also science offers explicit tools to characterize the maps, and
dual as neural prosthetics. When applying these tech- to use empirical estimates of connectivity to inform the
niques to clinical populations, a pressing question arises design of new networks. Indeed, the concept of network
due to limited resources: Who will benefit most? Can we design is a relatively new one in biological systems. When
choose the intervention that best fits a given individual? applied to the neural domain, network design includes the
Interestingly, emerging data suggest that organizational building of computational models of neural dynamics, as
characteristics of a person’s functional network architec- well as physical models [108] via micropatterning, mi-
ture as measured by EEG can be used to predict who crofabricated multielectrode arrays, and low-density neu-
will be receptive to motor imagery treatment [75, 104]. ronal culture techniques [109]. Together, these algorith-
These initial studies underscore the potential of network mic and empirical approaches provide exciting avenues
representations of neural data to provide sensitive and to map the neural connectome across scales and species
specific markers of the receptiveness of neural circuits to [13], and to better understand the dynamics that produce
induced network structure change [105]. cognition and behavior [21].

Neural mapping and neural connectivity estimation CONSTRUCTING AND USING BRAIN
NETWORKS FOR NEUROENGINEERING
While neuroengineering is often thought of as a field
of clinical translation, basic science plays a fundamental This brief survey of the literature demonstrates that
role in giving the investigator the knowledge and under- brain networks offer exciting capabilities in addressing
6

pressing questions in neuroengineering. In this section, ent cognitive processes. However, there are numerous
describe important considerations in constructing and other sources of noise that can contribute to spurious in-
using brain networks in the context of human imaging. creases of functional connectivity [114–116]. One of the
While we focus on human neuroimaging, these (or simi- most important influences on functional connectivity is
lar) considerations are likely to be important in the col- variations in amplitude or rate in the cardiac and respi-
lection and analysis of other types of data (multi-unit ration cycles. Respiration rate (∼0.2 Hz) and depth of
recording, optical imaging) as they become available for breathing can clearly influence local BOLD signal [117].
network analysis. Cardiac rate (∼1 Hz) also influences BOLD signal [118].
These effects are regionally complex, with respiration ef-
fects more apparent near the ventricles, and cardiac ef-
Image Acquisition fects more apparent near the largest arteries. Higher or-
der effects of the cardiac and respiratory cycle may also
The rapid growth of imaging-based network science has be present in the tissue beyond a simple linear projection
been accompanied by a parallel recognition that func- of the pulse and bellows signals. For example, the chest
tional and structural MRI data can be corrupted by a wall expansion will influence global magnetic field inho-
broad array of technical, physiologic and anatomic fac- mogeneity, while CSF pulsatility (via the cardiac pulse)
tors that, if not handled properly, lead to major errors in may be periodic with the chest expansion. Thus, the in-
network modeling. The good news is that MRI is a ma- fluence of both on functional connectivity analyses will
ture technology and it is rare for data to be corrupted by be dependent on an individual subject’s physiologic state
artifacts secondary to unreliable hardware or poor pulse and unique anatomy.
sequence designs. The bad news is that brain imaging is There are many retrospective strategies for removing
commonly corrupted by more subtle physical-anatomic the effects of cardiac and respiratory cycle variation on
properties that can be difficult to surmount with conven- the BOLD time series from each voxel. If heart rate
tional hardware and pulse sequences [110]. Both diffusion and respiration have been independently measured, then
weighted imaging (used for structural connectomics) and software tools such as “RETROICOR” can be used [119].
T2* weighted imaging (to detect changes of BOLD sig- It uses a Fourier expansion of the non-brain physiologic
nals in functional connectomics) are highly sensitive to signals with 8-20 regressors. While this method works
susceptibility artifacts. The most troublesome cause is an well for both linear and higher order artifacts, there is
air-tissue interface that leads to very localized non-linear a trade-off in that increasing the number of regressors
image distortion and signal irregularity. For example, the during RETROICOR correction will remove a greater
medial and inferior temporal and orbital frontal cortex of amount of relevant brain signal [120].
the brain are adjacent to air filled petrous and ethmoid For many experimental situations, independent mea-
sinuses. The resulting artifacts lead to missing stream- sures of heart rate and respiration are not available
lines projecting into these areas or unreliable estimates and methods besides RETROICOR are needed. Here
of functional activity within the distorted gray matter we mention techniques based on independent component
regions. These distortions are difficult to correct post analysis (ICA) of the rsMRI data. ICA decomposes the
hoc. The degree of signal dropout and missing data varies functional time series for all voxels into patterns of ac-
enormously between individual subjects. Thus, network tivity consisting of a set of spatial maps, each of which
analyses that are aggregated over a population need to has a corresponding time series that when added linearly,
carefully evaluate the influence of missing data on the sum to the original voxel-wise time series. A set of ICA
underlying connectivity matrices. A second challenge in components will represent both brain activity and noise
brain MRI, particularly for diffusion imaging is the effect components. Ideally, these sources of brain and non-
of eddy currents. Eddy currents are loops of electrical brain activity are independent. If so, then these latter
current induced within the brain tissue by the changing noise components can be removed and a new noise free
magnetic field required to generate images. This causes times series can be reconstructed. The challenge then,
spatial distortion within each image slice and can be par- is to find an unbiased, efficient method for identifying
ticularly impactful in diffusion imaging. A third chal- those components reflecting noise. Manual classification
lenge, also leading to geometric distortion, arises from of ICA components is very difficult, and requires expert
magnetic field inhomogeneity and phase encoding errors. knowledge. One semi-automated ICA-based X-noiseifier
There are numerous software tools available for correct- called “FIX” [121] uses a machine learning approach to
ing both types of distortion post hoc. aid with this process. For each ICA component a large
number of distinct spatial and temporal features are gen-
erated, each describing the proportion of temporal fluc-
Pitfalls unique to functional imaging networks tuations observed at high frequencies. These features are
fed into a multi-level classifier. After training by hand-
Ideally, all of the functional connections, whether for classification across a sufficient number of datasets, the
a resting state [111–113] or task-based network would be classifier can then be used with new datasets.
determined by patterned brain activity reflecting inher- An alternative approach is to estimate pulse and res-
7

piratory variability for a subject directly from an inde- mon motion-induced signal change. This type of signal
pendent set of fMRI data, utilizing temporal indepen- change has been described as the “predominant effect of
dent component analysis [122]. The method assumes motion” in a sample ranging from 8 to 23 years old [127].
that non-brain physiologic noise is spatially stationary. In recognition of potential artifacts from rapid motion
For example, noise associated with the carotid arteries (or RF spikes), software has been developed to address
will be in the same location across different rsMRI scans them. Rather than using the realignment information,
from the same subject. Once the underlying and in- these methods search for global spikes in signal intensity.
dependently derived spatial weighting matrix is identi- There is one final challenge with head motion artifacts.
fied by ICA in one dataset, it can be applied to a sep- Within each volumetric acquisition, a stack of slices are
arate rsMRI time series from the same subject to pro- acquired sequentially, typically by first sampling the odd
duce the temporal pattern of noise. The resulting car- slices and then the even slices. It is not uncommon for
diac and respiratory estimators can then be used with a brief head movement to demagnetize a subset of slices,
RETROICOR or similar correction methods. While this causing artifacts in every other slice. This is not de-
method works well, it requires an independent sample of tected in the transformation matrix and may not alter
functional data. global signal intensity. New tools are emerging to detect
It has been assumed that global BOLD activity, mea- unexpected signal spikes within single slices [131]. For
sured over the whole brain, will remain constant across a useful study benchmarking confound regression strate-
a time series. Any fluctuation would be due to instru- gies for the control of motion artifact in studies of func-
mentation issues or non-brain physiologic effects. How- tional connectivity, see [128].
ever, recent studies have examined the effect of removing
the global mean signal from the time course on subse-
quent connectivity analyses. Interestingly, multiple stud- Pitfalls unique to structural imaging networks
ies show that a significant portion of the global mean
signal is in fact related to the average signal within par- There are many sampling schemes for acquiring a set of
ticular resting state brain networks [123, 124] and that oriented diffusion scans. These include diffusion tensor
removing the global signal can result in spurious neg- imaging (DTI), which sample an object at a uniformly
ative correlations [125] and reduces reproducibility of spaced set of angles and at a constant magnetic gradi-
many network metrics [126]. Despite these disadvan- ent strength. When the gradient strength or number of
tages, global signal regression can be helpful in develop- directions are increased the angular resolution improves
mental and clinical cohorts to correct for motion-related (Q-ball and high angular resolution diffusion imaging
artifact [127, 128]. “HARDI”) but with the tradeoff of reduced signal to
Indeed, it is almost impossible for a person to remain noise in the scans. Multiple shells of gradient strengths
motionless in an MRI scanner. Breathing, swallowing can be applied (multishell diffusion imaging) or a uni-
and volitional movements can create motion that propa- form distribution across gradient strength and direction
gates to the head. A brain placed in the MRI field will can be applied (diffusion spectrum imaging) [132]. Crit-
become magnetized over ∼6 seconds. If the brain moves, ically, each of these methods requires a different mathe-
then it will no longer be magnetized in the same direc- matical technique for converting diffusion images to prob-
tion and there can be a massive increase in the signal abilistic estimates of local water diffusion, resulting in
until the brain has remagnetized to the new magnetic varying success at modeling the connectivity in differ-
orientation. To account for the effects of this motion- ent brain areas where there can be water diffusion in
induced noise, a variety of retrospective methods have multiple directions (the crossing fiber problem). Meth-
been proposed. Most assume that the change in signal ods using lower angular resolution such as DTI consis-
intensity will be global, occurring within a single time tently underestimate the number of possible streamlines
sample of the rsMRI time series. One of the most com- by an order of magnitude compared to multishell and DSI
mon methods is to use linear transformations to fit each methods. Missing connections can also arise because an
time sample to one time point. The resulting transfor- insufficient number of seeds are introduced to generate
mation weights (translations and rotations) can be used the underlying streamline set. Whatever the cause, al-
to adjust global signal intensity or be included in a re- lowing for missing data can significantly alter graph met-
gression model as a covariate of non-interest [129, 130]. rics [81]. On the other hand, commonly used algorithms
However, the “filtering” of time series data with motion for generating streamlines can create noisy, anatomically
parameters is problematic because they do not model implausible connections that must be removed by length
continuous motion directly. Rather, they capture net and/or angle thresholds. Most current algorithms for
displacement at the temporal resolution of the sampling generating streamline connections suffer from a length
frequency (∼0.5 Hz). If the head is displaced rapidly, bias [133, 134]. The shorter a connection, the easier it
and returns to the same position within a single sam- is to be reconstructed. Thus, a structural network will
pling period, then there is no net displacement, but a be more likely to represent short connections than long.
large signal spike in the data. This will profoundly alter Similarly, the odds are more likely that a streamline will
the strength of connectivity between areas with a com- be reconstructed if it is in a thick white matter fascicle
8

with many fibers oriented in a common direction than if The tools of temporal networks are particularly use-
it is in a thin fascicle. To address the length bias, some ful in modeling plasticity and learning with the aim of
authors normalize the streamline count between two gray predicting recovery [142]. In initial efforts, temporal net-
matter regions by the physical size of those same regions. works have been used to reveal common patterns of net-
Clearly, standardization in these acquisition, reconstruc- work reconfiguration that occur as healthy adult individ-
tion and counting procedures is essential for reproducibil- uals learn a new motor-visual skill over the course of days
ity and generalizability. to weeks [59, 143–146]. Interestingly, individuals that dis-
played greater network flexibility, particularly in areas of
the brain critical for cognitive control [146] learned more
quickly than individuals with less flexible brains [143].
FRONTIERS IN COMPUTATIONAL SCIENCE
AND SYSTEMS ENGINEERING
While these studies initially applied temporal network
techniques to motor skill learning with the aim of inform-
ing rehabilitation after stroke [147], there are many open
With these empirical considerations in mind, it is nev- questions about how sensitive these techniques might be
ertheless clear that brain imaging has provided a fertile to neural or cognitive plasticity underlying other types of
test bed for developing and testing novel tools from net- learning [148, 149], or to other dynamic processes that are
work science. Yet, it is likewise clear that this is only of important to neuroengineering in other clinical con-
a first wave of innovation. Indeed, network neuroscience texts.
offers to the field of neuroengineering two distinct sets Beyond temporal networks, one can extend the multi-
of frontiers: one in the development of computational layer network construct to represent relationships across
and systems engineering approaches, and the other in different imaging modalities [150]: for example, calcium
translating current and future advances directly to clin- transients and local field potentials, or structural MRI
ical populations. In this section, we briefly review new and EEG, or diffusion imaging and functional MRI [151].
directions in algorithmic development, computational ar- Alternatively, one could think of letting each layer rep-
chitectures, signal processing techniques, and statistics resent a different frequency band [152, 153] or a different
that support the extraction, representation, and char- patient in a clinical cohort. Indeed, the potential ap-
acterization of meaningful relational patterns in neural plications of these multilayer representations across neu-
data. We also discuss the nascent application of control roimaging contexts is surprisingly broad, and future ef-
theory to these networks, and highlight their potential forts will likely include a careful assessment of their util-
utility in guiding clinical interventions. ity in uncovering conserved and variable properties of
networked neural systems.

Dynamic and multilayer networks


Statistical tools, frameworks, and null models
A commonly faced challenge in applying network anal-
yses to neural data is that the processes we often wish to An important burgeoning area of work lies in build-
understand are inherently dynamic processes [135–137]. ing, testing, and validating appropriate statistical meth-
Rehabilitation, response to treatment, monitoring dis- ods and models for network inference. Because networks
ease progression, and tracking BCI learning are all evolv- are not simple mathematical objects, the tools required
ing processes that occur over a range of time scales. Yet, to capture and compare them extend beyond what tradi-
networks in their simplest forms are static: a fixed set tional statistics offers [154]. Many efforts have focused on
of network nodes are connected by a single estimate of developing sophisticated permutation-based methods for
connectivity. In extending these static descriptions to in- network comparison [155, 156], and some have extended
corporate time, the applied mathematics community has these methods to assess differences in network functions
defined so-called multilayer networks [138]. Colloquially, (rather than univariate statistics) [157–159], for exam-
a multilayer network is a network that contains different ple by building on tools developed in the field of func-
layers, and in which the edges in a given layer represent tional data analysis [160–162]. In addition to compar-
a different type of relationship than the edges in another ing networks, one often wishes to understand whether
layer. Perhaps the simplest type of multilayer network is the network structure or dynamics that one observes
a temporal network, where each layer is a time window in empirical data is expected or unexpected. Answer-
and the edges within that layer represent relationships ing these questions depends on the development of ap-
that are true in that time window [139]. By tying each propriate static and dynamic network null models (see
layer to the next using identity links (edges between node [159, 163, 164] and [141], respectively). Statistical consid-
i in layer l and node i in layer l ± 1), the static graph rep- erations also extend to estimating the connectivity itself
resentation as an adjacency matrix can be expanded to [32, 165], assessing its significance [157], and measuring
a dynamic graph representation as an adjacency tensor its relationship to behavior or symptomatology [166]. Fi-
[140, 141], providing important mathematical advantages nally, a nascent area of inquiry lies in building statistical
to common statistical challenges present in these data. models of networks [31, 167, 168] in order to understand
9

their generative principles [23–25, 30].

Algebraic topology

While extremely powerful, network science is largely


built on the tools of graph theory, which inherently treat
the dyad (a single connection between two nodes) as the
fundamental unit of interest. Recent evidence, however,
points to the fact that sensor networks, technological net-
works, and even neural networks display higher-order in-
teractions that simply cannot be reduced to pairwise rela-
tionships [169, 170]. To address this growing realization,
we can turn to recent advances in applied algebraic topol- FIG. 4. Tools for Higher-Order Interactions from Al-
ogy [171], which reframes the problem of relational data gebraic Topology. (a) The human connectome is a com-
in terms of simplices or collections – rather than pairs – plex network architecture that contains both dyadic and
of vertices [172] (Fig. 4). This added sensitivity enables higher-order interactions. Graph representations of the hu-
algebro-topological tools to offer mechanisms for neural man connectome only encode dyadic relationships, and leave
coding [173, 174], distinguish disparate classes of graph the higher-order interactions unaccounted for. A natural way
models [175], and separate healthy from clinical popula- in which to encode higher-order interactions is in the language
tions [176]. The framework also offers useful tools to con- of algebraic topology, which defines building blocks called sim-
sider the evolution of simplices over time drawing on the plices [172]: a 0-simplex is a node, a 1-simplex is an edge
between two nodes, a 2-simplex is a filled triangle, etc. (b)
notion of a filtration, and tools to identify and track hol-
These building blocks enable the desciption of two distinct
low cavities in networks – structures that are otherwise structural motifs that are thought to play very different roles
invisible to common graph metrics [172]. We anticipate in neural computations [174]: (i) cliques, which are all-to-
that the next few years will see an increasing interest in all connected subgraphs, are thought to facilitate integrated
better understanding the role of these higher order in- codes and computations, and (ii) cycles or cavities, which are
teractions in healthy cognition versus disease, and their collection of n-simplices arranged to have an empty geometric
sensitivity as biomarkers for tracking effects of training boundary, are thought to facilitate segregated codes and com-
and rehabilitation. putations. (c) Additional tools available to the investigator
include filtrations and persistent homology. Filtrations repre-
sent weighted simplicial complexes as a series of unweighted
simplicial complexes, and can be used to study networks that
Network control theory
change over time, or that display hierarchical structure across
edge weights. Filtrations allow one to follow cycles from one
A final exciting frontier that we will mention – which complex to another and quantify how long they live (via the
bridges both computational science and systems engi- number of complexes in which they are consecutively present).
neering – is the development and application of explic- Because this is a study of the persistence of a cycle, it is re-
itly network control theory [177, 178] to neural systems ferred to as the persistent homology of the weighted simplicial
(Fig. 5). Indeed, neural control engineering [179] is slowly complex.
evolving into neuro-network control engineering, as the
control problems become tuned to the underlying graph
architecture of the dynamical processes [180]. In general,
these applications take one of two forms: either seeking
to understand how neural systems control themselves, brain into difficult-to-reach states, far away on an en-
or how one can exogeneously control a neural system, ergy landscape [181]. Moreover, the brain’s densely in-
steering it away from pathological dynamics and towards terconnected rich-club is poised to form the ground state
healthy dynamics. of the system, being the least energetically costly target
In the first case, we seek to understand how neural state [182]. Interestingly, these control principles of the
systems control themselves. To address this question, brain, built on the organization of white matter tracts,
we can write down a model of brain dynamics where are significantly altered in individuals who have experi-
the current brain state depends on (i) the previous brain enced traumatic brain injury [183], suggesting their util-
state, (ii) the wiring pattern that structurally connects ity in clinical applications. However, it is also impor-
network nodes (brain regions), and (iii) the control in- tant to be cautious; these predictions are based on linear
put. Assuming this is a linear, time-invariant, noise-free, network control while the brain is a nonlinear dynamical
and discrete-time model, we can infer which brain regions system, and therefore interpretations should be validated
are predisposed to affect the system, and in what ways. in additional studies [184]. For example, demonstrating
Early efforts along these lines revealed that regions in that individual differences in cognitive control function
the brain’s executive system are well-poised to push the are correlated with individual differences in network con-
10

trol statistics will be an important first step [185], as will


demonstrating that these statistics change over develop-
mental time scales in which cognitive control emerges
in children [186]. Moreover, exploring the applicability
of nonlinear control strategies, including linearization of
nonlinear systems, will be an important avenue of inquiry
for future work [184].
The second context in which network control theory
offers a powerful toolset for neuroengineers is in address-
ing the question of how to exogeneously control a neu-
ral system and accurately predict the outcome on neu-
rophysiological dynamics – and, by extention, cognition
and behavior. Indeed, how to target, tune, and opti-
mize stimulation interventions is one of the most press- FIG. 5. Brain network regulation and control can help
navigate dynamical states. To accomplish behavioral and
ing challenges in the treatment of Parkinson’s disease and
cognitive goals, brain networks internally navigate a complex
epilepsy, to name a couple [7]. More broadly, this ques- space of dynamical states. Putative brain states may be sit-
tion directly impacts the targeting of optogentic stimu- uated in various peaks and troughs of an energy landscape
lation in animals [187] and the use of invasive and non- – requiring the brain to expend metabolic energy to move
invasive stimulation in humans [188] (e.g., deep brain, from the current state to the next state. Within the space
grid, transcranial magnetic, transcranial direct current, of possible dynamical states, there are easily accessible states
and transcranial alternating current stimulation). As a and harder-to-reach states; in some cases, the accessible states
case study, consider medically refractory epilepsy, where are healthy while in other cases they may contribute to dys-
network techniques can be used to identify seizure onset function, and similarly for the harder-to-reach states. Two
zones [77, 88, 189] and where network control theory can commonly observed control strategies used by brain networks
be used to detect drug-resistant seizures [190], inform the are average control and modal control. In average control,
highly central nodes navigate the brain towards easy-to-reach
development of a distributed control algorithm to quiet
states. In contrast, modal control nodes tend to be isolated
seizures using grid stimulation [191], and identify areas brain regions that navigate the brain towards hard-to-reach
of the network to target during resective surgery [78]. states that may require additional energy expenditure [181].
As a self-regulation mechanism for preventing transitions to-
wards damaging states, the brain may employ cooperative and
antagonistic, push-pull, strategies [78]. In such a framework,
the propensity for the brain to transition towards a damag-
ing state might be competitively limited by opposing modal
TOWARDS CLINICAL TRANSLATION and average controllers whose goal would be to pull the brain
towards less damaging states.
Together, these exciting computational frontiers have
the potential to directly inform clinical practice. Indeed,
several of the main translational challenges of neuroengi-
neering are ripe for the incorporation of network data.
These opportunities begin at the earliest stages of clin-
ical diagnosis and monitoring, where variation between
EXTENSIONS BEYOND NEUROENGINEERING
individuals – and even indeed variation within a single
individual – stymie progress in tuning medication, stim-
ulation, brain-machine interfaces, neuroprosthetics, and
physical or cognitive-behavioral therapy to offer individ- Before concluding, it is important to point out that the
uals a better quality of life (Fig. 6). Concerted efforts in mathematical methods and conceptual frameworks that
mapping relational architectures in neural and behavioral we have been discusing in this review are more generally
data in the form of graphs and networks will be critical to applicable beyond the specific realm of brain connectiv-
obtaining a more holistic understanding of mental health, ity. From genes [194] to the musculo-skeletal system,
as well as greater insights into optimizing interventions. from central to peripheral nervous systems [1], and from
Such mappings could occur in the traditional sense using injured neural tissue in brains causing cognitive deficits
empirical measurements performed in research or hospi- to neural tissue in muscles causing pain [16], network sci-
tal settings; but perhaps the most tantalizing possibilities ence offers an approach that spurns reductionism in favor
currently being discussed include the use of digital data of wholistic maps and models of complex interconnected
from smart phones to accurately phenotype individuals, systems. Indeed, future work may benefit from consider-
and the health of their nervous system, with the goal ing the nervous system as embedded or embodied in the
of better guiding intervention strategies for the clinically broader context, as only one part of an interconnected
unwell [192, 193]. web of networks supporting human life [195, 196].
11

CONCLUSION AND FUTURE OUTLOOK

In this review, we have sought to introduce an exciting


and emerging frontier in neuroengineering: a network
science of brain connectivity. In addition to outlining
the mathematical underpinnings of the field, we have
briefly described some marked initial successes in which
the tools of network neuroscience have been brought to
bear on neural mapping and connectivity estimation,
diagnosis and monitoring, and rehabilitation and treat-
ment. However, we are also careful to describe common
pitfalls and associated limitations, in an effort to offer
a balanced guide in incorporating these techniques into
FIG. 6. Clinical translation of network neuroscience
tools. Network neuroscience offers a natural framework for one’s own research practices. We took the liberty to
improving tools to diagnose and treat brain network disorders. speculate in the later sections about some important
(A) For drug-resistant epilepsy patients, invasive monitoring frontiers that we believe will become increasingly critical
of brain activity to localize brain tissue where seizures orig- to the questions posed by neuroengineering in the near
inate and plan resective surgery is challenging, because the future, both from a computational point of view and
neural processes generating seizures are poorly understood. from a view towards clinical impact. In closing, we
Epileptic brain signals, electrical fields produced by the firing underscore yet again that the strength and novelty of
of neuron populations, are sensed by electrodes that rest on network neuroscience lies in its brazen grasp on the full
the surface of the brain, beneath the dura, and are recorded complexities of relational data, facilitating transforma-
by a digital acquisition system. A three-dimensional recon- tive approaches to understanding, fixing, and building
struction of a patient’s brain (red) with electrodes co-localized
(green) to anatomical features is shown here. (B) Recorded
brains.
brain signals are studied by clinical practitioners to charac-
terize spatial and temporal behavior of the patient’s seizure Acknowledgements. We thank Chad Giusti, Jason
activity. In the plot, each line represents time-varying volt- Kim, and Matthew Hemphill for helpful comments on an
age fluctuation from each electrode sensor. (C) Inferred func- earlier version of this manuscript. DSB would also like to
tional connections from a single time-slice during the patient’s acknowledge support from the John D. and Catherine T.
recorded seizure demonstrates rich relationships in neural dy- MacArthur Foundation, the Alfred P. Sloan Foundation,
namics between brain regions and are not visually evident the Army Research Laboratory and the Army Research
from B (blue circles are nodes, red links are strong connec- Office through contract numbers W911NF-10-2-0022
tions, yellow links are weak connections). Functional connec- and W911NF-14-1-0679,the National Institute of Mental
tivity patterns demonstrate strong interactions around brain
Health (2-R01-DC-009209-11), the National Institute of
regions in which seizures begin and weak projections to brain
regions where seizures spread. Objective tools in network
Child Health and Human Development (1R01HD086888-
neuroscience can usher in an era of personalized algorithms 01), the Office of Naval Research, and the National
capable of mapping epileptic network architecture from neural Science Foundation (BCS-1441502, BCS-1430087, BCS-
signals and pinpointing implantable, neurostimulation devices 1631550, and CAREER PHY-1554488). The content
to specific brain regions for intervention [77, 78, 188]. is solely the responsibility of the authors and does not
necessarily represent the official views of any of the
funding agencies.

[1] Chen, H. I. et al. Neural substrate expansion for the [4] Putze, F. & Schultz, T. Adaptive cognitive technical
restoration of brain function. Front Syst Neurosci 10, systems. J Neurosci Methods 234, 108–115 (2014).
1 (2016). [5] Krusienski, D. J. et al. Critical issues in state-of-the-
[2] Haynes, J. D. & Rees, G. Decoding mental states from art brain-computer interface signal processing. J Neural
brain activity in humans. Nat Rev Neurosci 7, 523–534 Eng 8, 025002 (2011).
(2006). [6] DiLorenzo, D. J. & Bronzino, J. D. Neuroengineering
[3] Christophel, T. B., Hebart, M. N. & Haynes, J. D. (CRC Press, 2007).
Decoding the contents of visual short-term memory [7] Johnson, M. D. et al. Neuromodulation for brain disor-
from human visual and parietal cortex. J Neurosci 32, ders: challenges and opportunities. IEEE Trans Biomed
12983–12989 (2012). Eng 60, 610–624 (2013).
12

[8] Glaser, J. I. & Kording, K. P. The development and [30] Pavlovic, D. M., Vértes, P. E., Bullmore, E. T., Schafer,
analysis of integrated neuroscience data. Front Comput W. R. & Nichols, T. E. Stochastic blockmodeling of the
Neurosci 10, 11 (2016). modules and core of the caenorhabditis elegans connec-
[9] Valiant, L. G. What must a global theory of cortex tome. PloS one 9, e97584 (2014).
explain? Curr Opin Neurobiol 25, 15–19 (2014). [31] Simpson, S. L., Hayasaka, S. & Laurienti, P. J. Expo-
[10] Craver, C. F. Beyond reduction: mechanisms, multifield nential random graph modeling for complex brain net-
integration and the unity of neuroscience. Stud Hist works. PLoS One 6, e20039 (2011).
Philos Biol Biomed Sci 36, 373–395 (2005). [32] Lindquist, M. A., Xu, Y., Nebel, M. B. & Caffo, B. S.
[11] Bullmore, E. et al. Generic aspects of complexity in Evaluating dynamic bivariate correlations in resting-
brain imaging data and other biological systems. Neu- state fMRI: a comparison study and a new approach.
roimage 47, 1125–1134 (2009). Neuroimage 101, 531–546 (2014).
[12] Stevenson, I. H. & Kording, K. P. How advances in [33] Misic, B. et al. Cooperative and competitive spreading
neural recording affect data analysis. Nat Neurosci 14, dynamics on the human connectome. Neuron 86, 1518–
139–142 (2011). 1529 (2015).
[13] Bassett, D. S. & Sporns, O. Network neuroscience. Na- [34] Misic, B., Sporns, O. & McIntosh, A. R. Communica-
ture Neuroscience In Press (2016). tion efficiency and congestion of signal traffic in large-
[14] Long, B., Zhang, Z. & Yu, P. S. Relational Data Cluster- scale brain networks. PLoS Comput Biol 10, e1003427
ing: Models, Algorithms, and Applications (CRC Press, (2014).
2010). [35] Patel, T. P., Ventre, S. C., Geddes-Klein, D., Singh,
[15] Conaco, C. et al. Functionalization of a protosynaptic P. K. & Meaney, D. F. Single-neuron NMDA receptor
gene expression network. Proc Natl Acad Sci U S A phenotype influences neuronal rewiring and reintegra-
109, 10612–10618 (2012). tion following traumatic injury. J Neurosci 34, 4200–
[16] Zhang, S., Bassett, D. S. & Winkelstein, B. A. Stretch- 4213 (2014).
induced network reconfiguration of collagen fibres in the [36] Gratton, C., Lee, T. G., Nomura, E. M. & D’Esposito,
human facet capsular ligament. J R Soc Interface 13, M. The effect of theta-burst TMS on cognitive control
20150883 (2016). networks measured with resting state fMRI. Front Syst
[17] van den Heuvel, M. P., Bullmore, E. T. & Sporns, O. Neurosci 7, 124 (2013).
Comparative connectomics. Trends in Cognitive Sci- [37] Kaiser, M. A tutorial in connectome analysis: topolog-
ences 20, 345–361 (2016). ical and spatial features of brain networks. Neuroimage
[18] Shih, C. T. et al. Connectomics-based analysis of in- 57, 892–907 (2011).
formation flow in the Drosophila brain. Curr Biol 25, [38] Fornito, A., Zalesky, A. & Breakspear, M. Graph anal-
1249–1258 (2015). ysis of the human connectome: promise, progress, and
[19] Bassett, E. T., D Sand Bullmore. Human brain net- pitfalls. Neuroimage 80, 426–444 (2013).
works in health and disease. Curr Opin Neurol 22, [39] van Diessen, E. et al. Opportunities and methodological
340–347 (2009). challenges in EEG and MEG resting state functional
[20] Fornito, A. & Bullmore, E. T. Connectomics: a new brain network research. Clin Neurophysiol 126, 1468–
paradigm for understanding brain disease. Eur Neu- 1481 (2015).
ropsychopharmacol 25, 733–748 (2015). [40] Reijneveld, J. C., Ponten, S. C., Berendse, H. W. &
[21] Medaglia, J. D., Lynall, M. E. & Bassett, D. S. Cogni- Stam, C. J. The application of graph theoretical analysis
tive network neuroscience. J Cogn Neurosci 27, 1471– to complex networks in the brain. Clin Neurophysiol
1491 (2015). 118, 2317–2331 (2007).
[22] Misic, B. & Sporns, O. From regions to connections [41] Bullmore, E. & Sporns, O. Complex brain networks:
and networks: new bridges between brain and behavior. graph theoretical analysis of structural and functional
Curr Opin Neurobiol 40, 1–7 (2016). systems. Nat Rev Neurosci 10, 186–198 (2009).
[23] Vertes, P. E. et al. Simple models of human brain func- [42] Bassett, D. S. & Bullmore, E. Small-world brain net-
tional networks. Proc Natl Acad Sci U S A 109, 5868– works. Neuroscientist 12, 512–523 (2006).
5873 (2012). [43] Bullmore, E. T. & Bassett, D. S. Brain graphs: graph-
[24] Vertes, P. E., Alexander-Bloch, A. & Bullmore, E. T. ical models of the human brain connectome. Annu Rev
Generative models of rich clubs in Hebbian neuronal Clin Psychol 7, 113–140 (2011).
networks and large-scale human brain networks. Philos [44] Butts, C. T. Revisiting the foundations of network anal-
Trans R Soc Lond B Biol Sci 369, 1653 (2014). ysis. Science 325, 414–416 (2009).
[25] Betzel, R. F. et al. Generative models of the human [45] Zalesky, A. et al. Whole-brain anatomical networks:
connectome. Neuroimage 124, 1054–1064 (2016). does the choice of nodes matter? Neuroimage 50, 970–
[26] Pilosof, S., Porter, M. A., Pascual, M. & Kefi, S. The 983 (2010).
multilayer nature of ecological networks. arXiv 1511, [46] Bollobas, B. Random Graphs (1985).
04453 (2016). [47] Bollobas, B. Graph Theory: An Introductory Course
[27] Proulx, S. R., Promislow, D. E. & Phillips, P. C. Net- (Springer-Verlag, 1979).
work thinking in ecology and evolution. Trends Ecol [48] Watts, D. J. & Strogatz, S. H. Collective dynamics of
Evol 20, 345–353 (2005). ’small-world’ networks. Nature 393, 440–442 (1998).
[28] Newman, M. E. J. Networks: An Introduction (MIT [49] Kitzbichler, M. G., Henson, R. N., Smith, M. L.,
Press, 2010). Nathan, P. J. & Bullmore, E. T. Cognitive effort drives
[29] Rubinov, M. & Sporns, O. Complex network measures workspace configuration of human brain functional net-
of brain connectivity: uses and interpretations. Neu- works. J Neurosci 31, 8259–8570 (2011).
roimage 52, 1059–1069 (2010).
13

[50] Rubinov, M. & Bassett, D. S. Emerging evidence of [70] van den Heuvel, M. P., Scholtens, L. H. & de Reus,
connectomic abnormalities in schizophrenia. J Neurosci M. A. Topological organization of connectivity strength
31, 6263–6265 (2011). in the rat connectome. Brain Struct Funct 221, 1719–
[51] Bassett, D. S. & Lynall, M.-E. Cognitive Neurosciences: 1736 (2016).
The Biology of the Mind, chap. Network methods to [71] Kaiser, M. Neuroanatomy: connectome connects fly and
characterize brain structure and function (MIT Press, mammalian brain networks. Curr Biol 25, R416–R418
2015). (2015).
[52] Li, Y. et al. Brain anatomical network and intelligence. [72] Hagmann, P. et al. Mapping the structural core of hu-
PLoS Comput Biol 5, e1000395 (2009). man cerebral cortex. PLoS Biol 6, e159 (2008).
[53] Bassett, D. S. & Bullmore, E. T. Small world brain [73] Deng, Z. D., McClinctock, S. M. & Lisanby, S. H.
networks revisited. The Neuroscientist Commissioned Brain network properties in depressed patients receiving
(2016). seizure therapy: A graph theoretical analysis of peri-
[54] Sporns, O. & Betzel, R. F. Modular brain networks. treatment resting EEG. Conf Proc IEEE Eng Med Biol
Annu Rev Psychol 67 (2016). Soc 2015, 2203–2206 (2015).
[55] Porter, M. A., Onnela, J.-P. & Mucha, P. J. Commu- [74] Toppi, J. et al. Graph theory in brain-to-brain con-
nities in networks. Notices of the AMS 56, 1082–1097 nectivity: A simulation study and an application to an
(2009). EEG hyperscanning experiment. Conf Proc IEEE Eng
[56] Fortunato, S. Community detection in graphs. Physics Med Biol Soc 2015, 2211–2214 (2015).
reports 486, 75–174 (2010). [75] Zhang, Y., Xu, P., Guo, D. & Yao, D. Prediction
[57] Everett, M. G. & Borgatti, S. P. Peripheries of cohesive of SSVEP-based BCI performance by the resting-state
subsets. Social Networks 21, 397–407 (1999). EEG network. J Neural Eng 10, 066017 (2013).
[58] Borgatti, S. P. & Everett, M. G. Models of [76] Bassett, D. S. et al. Cognitive fitness of cost-efficient
core/periphery structures. Social Networks 21, 375–395 brain functional networks. Proc Natl Acad Sci U S A
(1999). 106, 11747–11752 (2009).
[59] Bassett, D. S. et al. Task-based core-periphery organi- [77] Khambhati, A. N. et al. Dynamic network drivers
zation of human brain dynamics. PLoS Comput Biol 9, of seizure generation, propagation and termination in
e1003171 (2013). human neocortical epilepsy. PLoS Comput Biol 11,
[60] van den Heuvel, M. P. & Sporns, O. Rich-club organi- e1004608 (2015).
zation of the human connectome. The Journal of neu- [78] Khambhati, A., Davis, K., Lucas, T., Litt, B. & Bas-
roscience 31, 15775–15786 (2011). sett, D. S. Virtual cortical resection reveals push-pull
[61] Bassett, D. S. & Siebenhuhner, F. Multiscale Analy- network control preceding seizure evolution. Neuron In
sis and Nonlinear Dynamics: From Genes to the Brain, Press (2016).
chap. Multiscale network organization in the human [79] Niu, H., Wang, J., Zhao, T., Shu, N. & He, Y. Revealing
brain (Wiley, 2013). topological organization of human brain functional net-
[62] Bassett, D. S. et al. Efficient physical embedding of works with resting-state functional near infrared spec-
topologically complex information processing networks troscopy. PLoS One 7, e45771 (2012).
in brains and computer circuits. PLoS Comput Biol 6, [80] Zhang, J. et al. Mapping the small-world properties
e1000748 (2010). of brain networks in deception with functional near-
[63] Bassett, D. S., Brown, J. A., Deshpande, V., Carlson, infrared spectroscopy. Sci Rep 6, 25297 (2016).
J. M. & Grafton, S. T. Conserved and variable architec- [81] Bassett, D., Brown, J., Deshpande, V., Carlson, J. &
ture of human white matter connectivity. Neuroimage Grafton, S. Conserved and variable architecture of hu-
54, 1262–1279 (2011). man white matter connectivity. Neuroimage 54, 1262–
[64] Muldoon, S. F., Soltesz, I. & Cossart, R. Spatially clus- 1279 (2011).
tered neuronal assemblies comprise the microstructure [82] Zalesky, A. et al. Whole-brain anatomical networks:
of synchrony in chronically epileptic networks. Proc Natl does the choice of nodes matter? Neuroimage 50,
Acad Sci USA 110, 3567–3572 (2013). 970–83 (2010). URL http://www.ncbi.nlm.nih.gov/
[65] Bettencourt, L. M., Stephens, G. J., Ham, M. I. & pubmed/20035887.
Gross, G. W. Functional structure of cortical neuronal [83] Jones, D. K. Studying connections in the living human
networks grown in vitro. Phys Rev E Stat Nonlin Soft brain with diffusion mri. Cortex; a journal devoted
Matter Phys 75, 021915 (2007). to the study of the nervous system and behavior 44,
[66] Fulcher, B. D. & Fornito, A. A transcriptional signature 936–52 (2008). URL http://www.ncbi.nlm.nih.gov/
of hub connectivity in the mouse connectome. Proc Natl entrez/query.fcgi?db=pubmed&cmd=Retrieve&dopt=
Acad Sci U S A 113, 1435–1440 (2016). AbstractPlus&list_uids=18635164.
[67] Hilgetag, C. C. & Kaiser, M. Clustered organization [84] Bassett, D. S. et al. Hierarchical organization of hu-
of cortical connectivity. Neuroinformatics 2, 353–360 man cortical networks in health and schizophrenia. J
(2004). Neurosci 28, 9239–9248 (2008).
[68] Markov, N. T. et al. Cortical high-density counter- [85] Lynall, M. E. et al. Functional connectivity and brain
stream architectures. Science 342, 1238406 (2013). networks in schizophrenia. J Neurosci 30, 9477–9487
[69] Rubinov, M., Ypma, R., Watson, C. & Bullmore, E. (2010).
Wiring cost and topological participation of the mouse [86] Nomi, J. S. & Uddin, L. Q. Developmental changes in
brain connectome. Proceedings of the National Academy large-scale network connectivity in autism. Neuroimage
of Sciences of the USA doi/10.1073/pnas.1420315112 Clin 7, 732–741 (2015).
(2015). [87] Menon, V. Large-scale brain networks and psy-
chopathology: a unifying triple network model. Trends
14

Cogn Sci 15, 483–506 (2011). [106] Christopoulos, V. N. et al. A network analysis of devel-
[88] Burns, S. P. et al. Network dynamics of the brain and oping brain cultures. J Neural Eng 9 (2012).
influence of the epileptic seizure onset zone. Proc Natl [107] Kafashan, M. & Ching, S. Optimal stimulus scheduling
Acad Sci U S A 111, E5321–E5330 (2014). for active estimation of evoked brain networks. J Neural
[89] He, Y., Chen, Z., Gong, G. & Evans, A. Neuronal net- Eng 12, 066011 (2015).
works in Alzheimer’s disease. Neuroscientist 15, 333– [108] Kanagasabapathi, T. T. et al. Functional connectivity
350 (2009). and dynamics of cortical-thalamic networks co-cultured
[90] Poza, J. et al. Characterization of the spontaneous elec- in a dual compartment device. J Neural Eng 9, 036010
troencephalographic activity in Alzheimer’s disease us- (2012).
ing disequilibria and graph theory. Conf Proc IEEE Eng [109] Chang, J. C., Brewer, G. J. & Wheeler, B. C. Neuronal
Med Biol Soc 2013, 5990–5993 (2013). network structuring induces greater neuronal activity
[91] Tijms, B. M. et al. Alzheimer’s disease: connecting find- through enhanced astroglial development. J Neural Eng
ings from graph theoretical studies of brain networks. 3, 217–226 (2006).
Neurobiol Aging 34, 2023–2036 (2013). [110] Viallon, M. et al. State-of-the-art mri tech-
[92] Stam, C. J. Modern network science of neurological niques in neuroradiology: principles, pitfalls, and
disorders. Nat Rev Neurosci 15, 683–695 (2014). clinical applications. Neuroradiology 57, 441–467
[93] Zhu, H. et al. Changes of intranetwork and internetwork (2015). URL http://link.springer.com/article/10.
functional connectivity in Alzheimer’s disease and mild 1007/s00234-015-1500-1.
cognitive impairment. J Neural Eng 13, 046008 (2016). [111] Beckmann, C. F., DeLuca, M., Devlin, J. T. & Smith,
[94] Petti, M. et al. Aged-related changes in brain activity S. M. Investigations into resting-state connectivity using
classification with respect to age by means of graph in- independent component analysis. Philos Trans R Soc
dexes. Conf Proc IEEE Eng Med Biol Soc 2013, 4350– Lond B Biol Sci 360, 1001–13 (2005). URL http://
4353 (2013). www.ncbi.nlm.nih.gov/pubmed/16087444.
[95] Sacchet, M. D., Prasad, G., Foland-Ross, L. C., Thomp- [112] De Luca, M., Beckmann, C. F., De Stefano, N.,
son, P. M. & Gotlib, I. H. Elucidating brain con- Matthews, P. M. & Smith, S. M. fmri resting state
nectivity networks in major depressive disorder us- networks define distinct modes of long-distance inter-
ing classification-based scoring. Proc IEEE Int Symp actions in the human brain. Neuroimage 29, 1359–67
Biomed Imaging 2014, 246–249 (2014). (2006). URL http://www.ncbi.nlm.nih.gov/pubmed/
[96] Toppi, J. et al. Investigating statistical differences in 16260155.
connectivity patterns properties at single subject level: [113] Greicius, M. D., Supekar, K., Menon, V. & Dougherty,
a new resampling approach. Conf Proc IEEE Eng Med R. F. Resting-state functional connectivity reflects
Biol Soc 2014, 6357–6360 (2014). structural connectivity in the default mode network.
[97] Zhao, C. et al. The reorganization of human brain net- Cereb Cortex 19, 72–8 (2009). URL http://www.ncbi.
works modulated by driving mental fatigue. IEEE J nlm.nih.gov/pubmed/18403396.
Biomed Health Inform Epub ahead of print (2016). [114] Aurich, N. K. & Filho, J. A. Evaluating the
[98] Toppi, J. et al. Time varying effective connectivity for reliability of different preprocessing steps to esti-
describing brain network changes induced by a memory mate graph theoretical measures in resting state fmri
rehabilitation treatment. Conf Proc IEEE Eng Med Biol data. Frontiers in Neuroscience 9, 1–10 (2015).
Soc 2014, 6786–6789 (2014). URL http://journal.frontiersin.org/article/10.
[99] Ge, R., Zhang, H., Yao, L. & Long, Z. Motor imagery 3389/fnins.2015.00048/full.
learning induced changes in functional connectivity of [115] Brooks, J. C. W., Faull, O. K., Pattinson, K. T. S.
the default mode network. IEEE Trans Neural Syst Re- & Jenkinson, M. Physiological noise in brainstem
habil Eng 23, 138–148 (2015). fmri. Frontiers in human neuroscience 7, 623 (2013).
[100] Stoeckel, L. E. et al. Optimizing real time fMRI neu- URL http://journal.frontiersin.org/article/10.
rofeedback for therapeutic discovery and development. 3389/fnhum.2013.00623/abstract.
Neuroimage Clin 5, 245–255 (2014). [116] Marchitelli, R. et al. Test-retest reliability of the default
[101] Banca, P., Sousa, T., Duarte, I. C. & Castelo-Branco, mode network in a multi-centric fmri study of healthy
M. Visual motion imagery neurofeedback based on the elderly: Effects of data-driven physiological noise
hMT+/V5 complex: evidence for a feedback-specific correction techniques. Human brain mapping 37, 2114–
neural circuit involving neocortical and cerebellar re- 2132 (2016). URL http://eutils.ncbi.nlm.nih.gov/
gions. J Neural Eng 12, 066003 (2015). entrez/eutils/elink.fcgi?dbfrom=pubmed&amp;id=
[102] Fels, M., Bauer, R. & Gharabaghi, A. Predicting work- 26990928&amp;retmode=ref&amp;cmd=prlinks.
load profiles of brain-robot interface and electromy- [117] Birn, R. M., Diamond, J. B., Smith, M. A. & Ban-
graphic neurofeedback with cortical resting-state net- dettini, P. A. Separating respiratory-variation-related
works: personal trait or task-specific challenge? J Neu- fluctuations from neuronal-activity-related fluctuations
ral Eng 12, 046029 (2015). in fmri. NeuroImage 31, 1536–1548 (2006).
[103] Andersen, R. A., Kellis, S., Klaes, C. & Aflalo, T. To- [118] Shmueli, K. et al. Low-frequency fluctuations in the
ward more versatile and intuitive cortical brain-machine cardiac rate as a source of variance in the resting-state
interfaces. Curr Biol 24, R885–R897 (2014). fmri bold signal. Neuroimage 38, 306–20 (2007). URL
[104] Zhang, R. et al. Efficient resting-state EEG network http://www.ncbi.nlm.nih.gov/pubmed/17869543.
facilitates motor imagery performance. J Neural Eng [119] Glover, G. H., Li, T. Q. & Ress, D. Image-based method
12, 066024 (2015). for retrospective correction of physiological motion ef-
[105] Sporns, O. The human connectome: origins and chal- fects in fmri: Retroicor. Magn Reson Med 44, 162–7
lenges. Neuroimage 80, 53–61 (2013). (2000). URL http://www.ncbi.nlm.nih.gov/pubmed/
15

10893535. http://eutils.ncbi.nlm.nih.gov/entrez/eutils/
[120] Beall, E. B. & Lowe, M. J. The non-separability of elink.fcgi?dbfrom=pubmed&amp;id=26416652&amp;
physiologic noise in functional connectivity mri with retmode=ref&amp;cmd=prlinks.
spatial ica at 3t. Journal of Neuroscience Methods 191, [132] Hagmann, P. et al. Understanding diffusion mr imaging
263–276 (2010). URL http://eutils.ncbi.nlm.nih. techniques: from scalar diffusion-weighted imaging to
gov/entrez/eutils/elink.fcgi?dbfrom=pubmed&amp; diffusion tensor imaging and beyond. Radiographics : a
id=20600313&amp;retmode=ref&amp;cmd=prlinks. review publication of the Radiological Society of North
[121] Salimi-Khorshidi, G. et al. Automatic denoising America, Inc 26 Suppl 1, S205–23 (2006). URL http:
of functional mri data: combining independent //pubs.rsna.org/doi/abs/10.1148/rg.26si065510.
component analysis and hierarchical fusion of clas- [133] Pestilli, F., Yeatman, J. D., Rokem, A., Kay, K. N.
sifiers. NeuroImage 90, 449–468 (2014). URL & Wandell, B. A. Evaluation and statistical inference
http://eutils.ncbi.nlm.nih.gov/entrez/eutils/ for human connectomes. Nat Methods 11, 1058–63
elink.fcgi?dbfrom=pubmed&amp;id=24389422&amp; (2014). URL http://www.ncbi.nlm.nih.gov/pubmed/
retmode=ref&amp;cmd=prlinks. 25194848.
[122] Beall, E. B. & Lowe, M. J. Isolating physiologic [134] Yeh, C. H., Smith, R. E., Liang, X., Calamante, F.
noise sources with independently determined spatial & Connelly, A. Correction for diffusion mri fibre track-
measures. NeuroImage 37, 1286–1300 (2007). URL ing biases: The consequences for structural connectomic
http://eutils.ncbi.nlm.nih.gov/entrez/eutils/ metrics. Neuroimage (2016). URL http://www.ncbi.
elink.fcgi?dbfrom=pubmed&amp;id=17689982&amp; nlm.nih.gov/pubmed/27211472.
retmode=ref&amp;cmd=prlinks. [135] Hutchison, R. M. et al. Dynamic functional connectiv-
[123] Chang, C. & Glover, G. H. Time-frequency dynamics ity: promise, issues, and interpretations. Neuroimage
of resting-state brain connectivity measured with fmri. 80, 360–378 (2013).
Neuroimage 50, 81–98 (2010). URL http://www.ncbi. [136] Calhoun, V. D., Miller, R., Pearlson, G. & Adal, T.
nlm.nih.gov/pubmed/20006716. The chronnectome: time-varying connectivity networks
[124] Fox, M. D., Zhang, D., Snyder, A. Z. & Raichle, M. E. as the next frontier in fMRI data discovery. Neuron 84,
The global signal and observed anticorrelated resting 262–274 (2014).
state brain networks. J Neurophysiol 101, 3270–83 [137] Kopell, N. J., Gritton, H. J., Whittington, M. A. &
(2009). URL http://www.ncbi.nlm.nih.gov/pubmed/ Kramer, M. A. Beyond the connectome: the dynome.
19339462. Neuron 83, 1319–1328 (2014).
[125] Murphy, K., Birn, R. M., Handwerker, D. A., Jones, [138] Kivel, M. et al. Multilayer networks. J. Complex Netw.
T. B. & Bandettini, P. A. The impact of global 2, 203–271 (2014).
signal regression on resting state correlations: are [139] Holme, P. & Saramaki, J. Temporal networks. Phys.
anti-correlated networks introduced? Neuroimage 44, Rep. 519, 97–125 (2012).
893–905 (2009). URL http://www.ncbi.nlm.nih.gov/ [140] Mucha, P. J., Richardson, T., Macon, K., Porter,
entrez/query.fcgi?cmd=Retrieve&db=PubMed&dopt= M. A. & Onnela, J.-P. Community structure in time-
Citation&list_uids=18976716. dependent, multiscale, and multiplex networks. science
[126] Andellini, M., Cannat, V., Gazzellini, S., Bernardi, 328, 876–878 (2010).
B. & Napolitano, A. Test-retest reliability of graph [141] Bassett, D. S. et al. Robust detection of dynamic com-
metrics of resting state mri functional brain networks: munity structure in networks. Chaos 23, 013142 (2013).
A review. Journal of Neuroscience Methods 253, [142] Reinkensmeyer, D. J. et al. Computational neuroreha-
183–192 (2015). URL http://eutils.ncbi.nlm.nih. bilitation: modeling plasticity and learning to predict
gov/entrez/eutils/elink.fcgi?dbfrom=pubmed&amp; recovery. J Neuroeng Rehabil 13, 42 (2016).
id=26072249&amp;retmode=ref&amp;cmd=prlinks. [143] Bassett, D. S. et al. Dynamic reconfiguration of human
[127] Satterthwaite, T. D. et al. An improved framework for brain networks during learning. Proc Natl Acad Sci U
confound regression and filtering for control of motion S A 108, 7641–7646 (2011).
artifact in the preprocessing of resting-state functional [144] Mantzaris, A. V. et al. Dynamic network centrality
connectivity data. Neuroimage 64, 240–56 (2013). URL summarizes learning in the human brain. Journal of
http://www.ncbi.nlm.nih.gov/pubmed/22926292. Complex Networks 1, 83–92 (2013).
[128] Ciric, R. et al. Benchmarking confound regression [145] Bassett, D. S., Wymbs, N. F., Porter, M. A., Mucha,
strategies for the control of motion artifact in studies P. J. & Grafton, S. T. Cross-linked structure of network
of functional connectivity. Neuroimage In Submission evolution. Chaos 24, 013112 (2014).
(2016). [146] Bassett, D. S., Yang, M., Wymbs, N. F. & Grafton,
[129] Friston, K. J., Williams, S., Howard, R., Frackowiak, S. T. Learning-induced autonomy of sensorimotor sys-
R. S. & Turner, R. Movement-related effects in fmri tems. Nat Neurosci 18, 744–751 (2015).
time-series. Magn Reson Med 35, 346–55 (1996). URL [147] Heitger, M. H. et al. Motor learning-induced changes
http://www.ncbi.nlm.nih.gov/pubmed/8699946. in functional brain connectivity as revealed by means
[130] Lemieux, L., Salek-Haddadi, A., Lund, T. E., Laufs, H. of graph-theoretical network analysis. Neuroimage 61,
& Carmichael, D. Modelling large motion events in fmri 633–650 (2012).
studies of patients with epilepsy. Magn Reson Imaging [148] Mattar, M. G., Thompson-Schill, S. L. & Bassett, D. S.
25, 894–901 (2007). URL http://www.ncbi.nlm.nih. The network architecture of value learning. arXiv 1607,
gov/pubmed/17490845. 04169 (2016).
[131] Tierney, T. M. et al. Fiach: A biophysical [149] Karuza, E. A., Thompson-Schill, S. L. & Bassett, D. S.
model for automatic retrospective noise control in Local patterns to global architectures: Influences of net-
fmri. NeuroImage 124, 1009–1020 (2016). URL work topology on human learning. Trends Cogn Sci
16

S1364-6613, 30071–30077 (2016). [170] Ganmor, E., Segev, R. & Schneidman, E. Sparse low-
[150] Muldoon, S. F. & Bassett, D. S. Network and multilayer order interaction network underlies a highly correlated
network approaches to understanding human brain dy- and learnable neural population code. Proc Natl Acad
namics. Philosophy of Science In Press (2016). Sci U S A 108, 9679–9684 (2011).
[151] Nicosia, V., Skardal, P. S., Latora, V. & Arenas, A. [171] Ghrist, R. Elementary applied topology (Createspace,
Spontaneous synchronization driven by energy trans- 2014).
port in interconnected networks. arXiv 1405, 5855v2 [172] Giusti, C., Ghrist, R. & Bassett, D. S. Two’s company,
(2014). three (or more) is a simplex : Algebraic-topological tools
[152] Brookes, M. J. et al. A multi-layer network approach to for understanding higher-order structure in neural data.
MEG connectivity analysis. Neuroimage 132, 425–438 J Comput Neurosci 41, 1–14 (2016).
(2016). [173] Giusti, C., Pastalkova, E., Curto, C. & Itskov, V. Clique
[153] De Domenico, M., Sasai, S. & Arenas, A. Mapping mul- topology reveals intrinsic geometric structure in neural
tiplex hubs in human functional brain network. Front. correlations. Proc Natl Acad Sci U S A 112, 13455–
Neurosci. 10, 326 (2016). 13460 (2015).
[154] Kolaczyk, E. D. Statistical Analysis of Network Data: [174] Curto, C. What can topology tell us about the neural
Methods and Models (Springer, 2009). code? arXiv 1605, 01905 (2016).
[155] Simpson, S. L., Lyday, R. G., Hayasaka, S., Marsh, A. P. [175] Sizemore, A., Giusti, C. & Bassett, D. S. Classifica-
& Laurienti, P. J. A permutation testing framework tion of weighted networks through mesoscale homolog-
to compare groups of brain networks. Front Comput ical features. Journal of Complex Networks In Press
Neurosci 7, 171 (2013). (2016).
[156] Winkler, A. M., Webster, M. A., Vidaurre, D., Nichols, [176] Kim, E. et al. Morphological brain network assessed
T. E. & Smith, S. M. Multi-level block permutation. using graph theory and network filtration in deaf adults.
Neuroimage 123, 253–268 (2015). Hearing Research 315, 88–98 (2014).
[157] Ginestet, C. E. & Simmons, A. Statistical paramet- [177] Liu, Y.-Y., Slotine, J.-J. & Barabási, A.-L. Controllabil-
ric network analysis of functional connectivity dynamics ity of complex networks. Nature 473, 167–173 (2011).
during a working memory task. Neuroimage 55, 688– [178] Pasqualetti, F., Zampieri, S. & Bullo, F. Controlla-
704 (2011). bility metrics, limitations and algorithms for complex
[158] Bassett, D. S., Nelson, B. G., Mueller, B. A., Camchong, networks. IEEE Transactions on Control of Network
J. & Lim, K. O. Altered resting state complexity in Systems 1, 40–52 (2014).
schizophrenia. Neuroimage 59, 2196–2207 (2012). [179] Schiff, S. J. Neural Control Engineering: The Emerging
[159] Betzel, R. F. et al. The modular organization of human Intersection between Control Theory and Neuroscience
anatomical brain networks: Accounting for the cost of (MIT Press, 2011).
wiring. arXiv 1608, 01161 (2016). [180] Motter, A. E. Networkcontrology. Chaos 25, 097621
[160] Ramsay, J. & Silverman, B. W. Functional Data Anal- (2015).
ysis (Springer, 2005). [181] Gu, S. et al. Controllability of structural brain networks.
[161] Ramsay, J. O. & Silverman, B. W. Applied functional Nat Commun 6, 8414 (2015).
data analysis: methods and case studies, vol. 77 (Cite- [182] Betzel, R. F., Gu, S., Medaglia, J. D., Pasqualetti, F. &
seer, 2002). Bassett, D. S. Optimally controlling the human connec-
[162] Ramsay, J. O. Functional data analysis (Wiley Online tome: the role of network topology. Scientific Reports
Library, 2006). In Press (2016).
[163] Papadopoulos, L., Puckett, J., Daniels, K. E. & Bassett, [183] Gu, S. et al. Optimal trajectories of brain state transi-
D. S. Evolution of network architecture in a granular tions. arXiv 1607, 01706 (2016).
material under compression. arXiv 1603, 08159 (2016). [184] Cornelius, S. P., Kath, W. L. & Motter, A. E. Realistic
[164] Bassett, D. S., Owens, E. T., Porter, M. A., Manning, control of network dynamics. Nat Commun 4, 1942
M. L. & Daniels, K. E. Extraction of force-chain network (2013).
architecture in granular materials using community de- [185] Medaglia, J. D. et al. Cognitive control in the control-
tection. Soft Matter 11, 2731–2744 (2015). lable connectome. arXiv 1606, 09185 (2016).
[165] Lepage, K. Q., Ching, S. & Kramer, M. A. Inferring [186] Tang, E. et al. Structural drivers of diverse neural dy-
evoked brain connectivity through adaptive perturba- namics and their evolution across development. arXiv
tion. J Comput Neurosci 34, 303–318 (2013). 1607 (2016).
[166] Shehzad, Z. et al. A multivariate distance-based ana- [187] Ching, S. & Ritt, J. T. Control strategies for underac-
lytic framework for connectome-wide association stud- tuated neural ensembles driven by optogenetic stimula-
ies. Neuroimage 93, 74–94 (2014). tion. Front Neural Circuits 7, 54 (2013).
[167] Simpson, S. L., Moussa, M. N. & Laurienti, P. J. An [188] Muldoon, S. F. et al. Stimulation-based control of dy-
exponential random graph modeling approach to creat- namic brain networks. PLoS Comp Biol In Press
ing group-based representative whole-brain connectivity (2016).
networks. Neuroimage 60, 1117–1126 (2012). [189] Hao, S. et al. Computing network-based features
[168] Klimm, F., Bassett, D. S., Carlson, J. M. & Mucha, P. J. from intracranial EEG time series data: Application to
Resolving structural variability in network models and seizure focus localization. Conf Proc IEEE Eng Med
the brain. PLoS Computational Biology 10, e1003491 Biol Soc 2014, 5812–5815 (2014).
(2014). [190] Santaniello, S. et al. Quickest detection of drug-resistant
[169] Ganmor, E., Segev, R. & Schneidman, E. The architec- seizures: an optimal control approach. Epilepsy Behav
ture of functional interaction networks in the retina. J 22, S49–S60 (2011).
Neurosci 31, 3044–3054 (2011).
17

[191] Ching, S., Brown, E. N. & Kramer, M. A. Distributed [194] Beagan, J. A. et al. Local genome topology can exhibit
control in a mean-field cortical network model: impli- an incompletely rewired 3d-folding state during somatic
cations for seizure suppression. Phys Rev E Stat Nonlin cell reprogramming. Cell Stem Cell 18, 611–624 (2016).
Soft Matter Phys 86, 021920 (2012). [195] Baldassano, S. N. & Bassett, D. S. Topological distor-
[192] Onnela, J. P. & Rauch, S. L. Harnessing smartphone- tion and reorganized modular structure of gut microbial
based digital phenotyping to enhance behavioral and co-occurrence networks in inflammatory bowel disease.
mental health. Neuropsychopharmacology 41, 1691– Sci Rep 6, 26087 (2016).
1696 (2016). [196] Steinway, S. N., Biggs, M. B., Loughran, T. P. J., Papin,
[193] Torous, J., Kiang, M. V., Lorme, J. & Onnela, J. P. New J. A. & Albert, R. Inference of network dynamics and
tools for new research in psychiatry: A scalable and cus- metabolic interactions in the gut microbiome. PLoS
tomizable platform to empower data driven smartphone Comput Biol 11, e1004338 (2015).
research. JMIR Ment Health 3, e16 (2016).

You might also like