You are on page 1of 32

Neuron

Review

Reactive Gliosis and the Multicellular


Response to CNS Damage and Disease
Joshua E. Burda1 and Michael V. Sofroniew1,*
1
Department of Neurobiology and Brain Research Institute, University of California Los Angeles, Los Angeles, CA 90095-1763, USA
*Correspondence: sofroniew@mednet.ucla.edu
http://dx.doi.org/10.1016/j.neuron.2013.12.034

The CNS is prone to heterogeneous insults of diverse etiologies that elicit multifaceted responses. Acute
and focal injuries trigger wound repair with tissue replacement. Diffuse and chronic diseases provoke
gradually escalating tissue changes. The responses to CNS insults involve complex interactions among
cells of numerous lineages and functions, including CNS intrinsic neural cells, CNS intrinsic nonneural
cells, and CNS extrinsic cells that enter from the circulation. The contributions of diverse nonneuronal cell
types to outcome after acute injury, or to the progression of chronic disease, are of increasing interest as
the push toward understanding and ameliorating CNS afflictions accelerates. In some cases,
considerable information is available, in others, comparatively little, as examined and reviewed here.

Introduction cells that


A major goal of contemporary neuroscience is to understand
and ameliorate a wide range of CNS disorders. Toward this
end, there is increasing interest in cellular and molecular
mechanisms of CNS responses to damage, disease, and
repair. Neurons are the principal cells executing neural
functions and have long dominated investigations into
mechanisms underlying CNS dis- orders. Nevertheless,
mounting evidence indicates that treating all types of CNS
disorders will require a deeper understanding of how
multicellular responses to injury and disease are triggered,
evolve, resolve (or not), and impact on neuronal function.
The ability to repair tissue damaged by injury is fundamental
to vertebrate biology and central to survival. Evolutionary
pressure is likely to have forged certain fundamental cellular
and molecu- lar responses to damage that are common
across different tis- sues. The wound or injury response in skin
has long served as a model system for dissecting mechanisms
of tissue repair after acute focal tissue damage and has
provided insight into core cellular and molecular interactions
(Greaves et al., 2013; Gurtner et al., 2008; Singer and Clark,
1999). In addition, organ-specific features exist. Organ-intrinsic
cells that are specialized in inflam- matory regulation and tissue
repair are emerging as critical elements in organ-specific
responses to insults. Organ-specific features apply particularly
in the CNS, where glial cells, which maintain the
cytoarchitecture and homeostatic regulation without which
neurons could not function normally in healthy tis- sue, are also
principal responders to CNS insults. Changes in glial cell
function during responses to insults have the potential to
impact markedly on neuronal interactions and CNS functions.
CNS insults are caused by diverse etiologies that can elicit a
wide range of responses. For example, acute and focal injuries
trigger wound repair with tissue replacement, whereas diffuse
and chronic diseases can trigger gradually escalating tissue
changes. Analysis of similarities and differences in such re-
sponses can provide valuable insights. Cellular responses to
CNS insults involve complex interactions among cells of
numerous lineages and functions, including CNS intrinsic
neural cells, CNS intrinsic nonneural cells, and CNS extrinsic
Neuron 81, January 22, 2014 ª2014 Elsevier Inc. 1
Neuron

Review
enter from the circulation. The biology of cell types that
partici- pate in CNS responses to injury and disease models
has gener- ally been studied in isolation. There is increasing
need to study interplay of different cells to understand
mechanisms. This Re- view examines and reviews the
multiple cell types involved in, and contributing to, different
types of CNS insults. In some cases, extensive information
is available, in others, compara- tively little.
Terminology
Various terms used in discussions of CNS injury and
disease can be subject to different interpretations. In this
Review, we will define and use certain specific terms as
follows. ‘‘Reactive gliosis’’ will refer not only to microglia and
astroglia, but also to glial cells that have come to be known
as NG2-positive oligoden- drocyte progenitor cells (NG2-
OPCs). Glial cells in healthy CNS tissue will not be referred
to as ‘‘resting’’ or ‘‘quiescent.’’ This is an antiquated
concept. Glia are highly active in healthy CNS and
dynamically exert complex functions that play critical roles in
normal CNS functions (Barres, 2008; Sofroniew and Vinters,
2010). For example, astrocytes exhibit physiological
activation in the form of transient, ligand-evoked elevations
in intracellular calcium ([Ca2+]i) that represent a type of
astrocyte excitability, which is under intense investigation as
a potential means of mediating dynamic astrocyte functions,
including interactions with synapses and regulation of blood
flow (Attwell et al., 2010; Halassa and Haydon, 2010; Tong et
al., 2013; Verkhratsky et al., 1998). Microglia perform
essential roles in synapse devel- opment and turnover
(Stephan et al., 2012; Stevens et al., 2007). The term
‘‘activated’’ is often used in a binary all-or-none fashion to
define glial cells that have responded to insults. We feel that
use of the term in this manner is inaccurate in two ways.
First, it does not recognize that glia are continually being
‘‘activated’’ in physiological contexts. Second, as discussed
throughout this Review, glial cell responses to CNS insults
are not binary and are highly diverse and specifically
regulated. To differentiate physiological activation of glial
cells in healthy contexts from re- sponses associated with
injury or disease, we will use the term ‘‘reactive,’’ which is
also meant denote a broad spectrum of

2 Neuron 81, January 22, 2014 ª2014 Elsevier Inc.


Table 1. Diverse Cell Types in CNS Responses to Damage and progenitors and cells of different types that migrate to sites of
Disease injury (Benner et al., 2013; Lagace, 2012; Meletis et al., 2008;
CNS Intrinsic Cells Blood-Borne Nonneural Cells
Ohab and Carmichael, 2008).
CNS Intrinsic Nonneural Cells
CNS Intrinsic Neural Cells Leukocytes
Various nonneural-lineage cells intrinsic to CNS play critical
Neurons Monocyte/macrophage
roles in CNS damage and disease (Table 1). Microglia are well
Oligodendrocytes Neutrophils
docu- mented as highly sensitive early responders that
Astrocytes Eosinophils stimulate and recruit other cells, as well phagocytose debris
NG2-OPC NK cells (Hanisch and Kettenmann, 2007; Kreutzberg, 1996; Nimmerjahn
Neural stem/progenitor cells T cells et al., 2005; Ransohoff and Perry, 2009). Fibroblast-related
Ependyma B cells cells, including perivascular fibroblasts, meningeal fibroblasts,
and pericytes, contribute to tissue replacement by forming
CNS Intrinsic Nonneural Cells Other Bone Marrow-Derived Cells
fibrotic scar tissue after severe damage (Go¨ ritz et al., 2011;
Microglia Platelets (thrombocytes)
Klapka and Mu¨ ller, 2006; Logan and Berry, 2002; Soderblom et
Perivascular fibroblasts Fibrocytes
al., 2013; Winkler et al., 2011). Endothelia and endothelial
Pericytes Mesenchymal (bone marrow
progenitors are promi- nent during tissue replacement after
stromal) stem cells
CNS injury (Loy et al., 2002; Oudega, 2012) and are of
Endothelia and progenitors
increasing interest in CNS repair in light of their emerging roles
as trophic support cells in CNS development (Dugas et al.,
2008) and their ability to produce and present laminin (Davis
and Senger, 2005), a preferred growth
potential responses of glial cells to CNS insults. Lastly, we will substrate for many migrating cells and axons.
avoid use of the term ‘‘scar’’ on its own and will instead distin- Blood-Borne Nonneural Cells
guish between ‘‘astrocyte scars’’ that form compact borders Blood-borne immune and inflammatory cells of different kinds
around tissue lesions and ‘‘fibrotic scars’’ that are formed by (Table 1) play prominent roles in CNS responses to damage
multiple nonneural cell types and extracellular matrix within and disease and have been studied and reviewed (Perry, 2010;
lesion cores (as discussed in detail below). We will equate ‘‘glial Popovich and Longbrake, 2008). In addition to well-known roles
scar’’ with ‘‘astrocyte scar.’’ in phagocytosis and removal of debris, there is also now
increasing evidence that subtypes of leukocytes play active
Multicellular Response to CNS Insults roles in tissue repair (Derecki et al., 2012; London et al., 2013;
Before discussing the different types of responses to CNS dam- Popo- vich and Longbrake, 2008). Platelets aggregate rapidly
age and disease, it is useful briefly to introduce different cell after damage for clot formation and haemostasis. Other blood-
borne,
types involved in these responses. For ease of consideration, bone marrow-derived cell types that home in to tissue injury,
we have groupe d cells according to lineages as (1) neural and including in CNS, include fibrocytes (Aldrich and Kielian, 2011;
nonneural cells intrinsic to CNS and (2) blood-borne nonneural Reilkoff et al., 2011) and bone marrow-derived mesenchymal
cells that derive primarily from bone marrow (Table 1). Some of stem cells (Askari et al., 2003; Bianco et al., 2001; Jaerve et al.,
these cell types have been studied extensively in CNS 2012), but their functions are not well understood.
disorders,
others comparatively little. Most often they have been studied in Extracellular Matrix
isolation. In subsequent sections, we will endeavor to examine Extracellular matrix (ECM) generated by different cells plays
their interactions. crit- ical roles in tissue repair and replacement after acute
CNS Intrinsic Neural Cells insults, as well as in chronic tissue remodeling during chronic
The principal neural-lineage cells in the CNS, neurons, disease (Mid-
oligoden- drocytes and astrocytes, have been studied and wood et al., 2004). Molecules that modify ECM matrix, such as
reviewed exten- sively as regards their individual responses to metaloproteases (MMPs), are also important in this regard
CNS injury and disease (Barres, 2008; Franklin and Ffrench- (Mid- wood et al., 2004). In the CNS, ECM generated by
Constant, 2008; Mattson, 2000; Sofroniew and Vinters, 2010). different cell types responding to damage or disease can
Less well studied include a wide vari- ety of molecules such as laminin,
collagens, and glycoproteins
are glial cells that have come to be known as NG2-positive oligo- such as chondroitin or heparan sulfate proteoglycans (CSPGs
dendrocyte progenitor cells (NG2-OPCs). Like other glia, NG2- or HSPGs) that are implicated both in supporting tissue repair
OPC tile the CNS and respond to CNS insults ( Nishiyama as well as in contributing to the failure of axonal regeneration
et al., 2009). Features of the NG2-OPC response include (Davis and Senger, 2005; Klapka and Mu¨ ller, 2006; Logan and
(Franklin and Ffrench- Berry, 2002; Silver and Miller, 2004).
migra- tion toward injury and cell proliferation
Constant, 2008; Hughes et al., 2013). Besides replacing lost
oligodendrocytes (Sun et al., 2010), other roles of NG2-OPC in Response to Focal Insults
CNS injury and disease await future study and elucidation. In Two principal types of acute focal insults in the CNS are focal
addition, the adult CNS harbors neural stem cells (NSCs) of traumatic injury and ischemic stroke. In addition to being major
different potencies that reside in the periependymal regions clinical problems, they have for decades served as prototypical
along the ventricles and central canal in adult brain and spinal experimental models with which to study CNS mechanisms of
cord (Garcia et al., 2004; Kriegstein and Alvarez-Buylla, 2009). response to damage and repair. In this section, we will examine
These adult NSCs can also respond to CNS injury and basic features of CNS multicellular responses to acute focal
generate
Figure 1. Phases and Time Course of Multicellular Responses to Acute Focal CNS Damage
During periods of cell death (A), cell replacement (B), and tissue remodeling (C), numerous overlapping events occur (D) that involve interactions among CNS
intrinsic neural cells, CNS intrinsic nonneural cells, and cells infiltrating from the circulation.

injuries and how these change over time, using information We will first examine CNS responses in a context in which the
from response to trauma or stroke as models. In this regard, it acute insults remain uninfected and resolve. Below, we will
is note- worthy that acute focal trauma caused by contusion or also briefly consider more chronic focal CNS insults, including
crush re- sults in severe vascular damage and therefore focal infections with abscess formation, primary and secondary
exhibits many
sequelae similar to stroke. We will make comparisons with the tumors, and chronic focal autoimmune lesions, such as multiple
acute wound response in skin as a model system for cellular sclerosis plaques, which exhibit various features and cellular re-
and molecular mechanisms of wound repair (Greaves et al., sponses similar to traumatic and ischemic injury. In order to pre-
2013; Gurtner et al., 2008; Shechter and Schwartz, 2013; (3) tissue remodeling (Figure 1).
Singer and Clark, 1999). As in skin repair, CNS responses to
acute focal damage can also be divided broadly into three
overlapping but distinct phases: (1) cell death and
inflammation, (2) cell prolifer- ation for tissue replacement, and
sent succinct summaries of cellular responses and
interactions, discussion of information about underling
molecular signals is deferred to a later section below. It
deserves mention that the time courses given for response
phases and events are general- ized approximations (Figure
1), and there is much potential for
Figure 2. Lesions Exhibit Tissue
Compartments and Signaling Gradients
(A and B) Focal damage gives rise to lesions
with distinct tissue compartments in the form of
nonneural lesion cores (LC), newly proliferated
compact astrocyte scars (AS), and perilesion pe-
rimeters (PLP) that exhibit diminishing gradients
of reactive gliosis that transition gradually to
healthy tissue (HT). AS abruptly demarcate the
transition between nonneural LC and PLP with
viable neural cells. Size of the lesion core and the
location of AS, and this transition, are regulated
by opposing molecular signals. Gradients of
molecular signals emanating from LC impact on
cells in PLP.

et al., 2005). Astrocytes, in contrast,


remain in situ and do not migrate either
to or away from injury sites but can
swell osmotically and, depending on the
severity of injury or ischemia, can die
in the center of severe lesions or can
overlap of events as well as variations in duration or timing in become reactive and hypertrophy and in some cases
specific situations. proliferate (Bardehle et al., 2013; Zheng et al., 2010). Different
Phase I: Cell Death and Inflammation aspects of this first phase of response occur in overlapping
sequences dur-
After focal CNS tissue damage from insults such as trauma or ing the first few days and then begin gradually diminishing
ischemia that cause acute local cell death, the first phase of (Figure 1D), provided that the insult is a single acute event not
response in the injury center or core includes both very rapid complicated by bony compression, infection, or other exacerba-
events that occur over timescales of seconds to hours and tions that might prolong the damaging insult. As acute
more gradually progressing events that develop over days (Fig- diminish, therresponses e is overlap with the onset of different
ures 1A and 1D ). Very rapid events include haemostasis with proliferation types of cell associated with the next phase of
coagulation cas cade, platelet aggregation, and clot formation. ures 1B and responses (Fig- 1D).
These rapid events are followed by overlapping sequences of Phase II: Cell Proliferation and Tissue Replacement
re- sponses e intrinsic cells, of
recruitment of inflammatory
tissu and The second phase of response to acute CNS tissue damage oc-
immune cells, subacute death of parenchymal cells, and curs from about 2 to 10 days after the insult and is characterized
initiation l (Figures 1A and 1D). In healthy
CNS
of debris remova tissue, by the proliferation and local migration of cells that implement tis-
large
and polar molecules in the circulation are excluded from sue repair and replacement. These cells include endothelial pro-
diffusion into CNS parenchyma by the blood-brain barrier genitors, fibroblast-lineage cells, different types of inflammatory
(BBB) (Abbott
et al., 2006; Zlokovic, 2008). After focal insults with BBB damage, cells, and various types of glia and neural-lineage progenitor
cells,
blood-borne molecules normally excluded from the CNS influx including scar-forming astrocytes and their progenitors (Figures
and signal to local cells, as discussed in more detail below in 1B and 1D). Some of these cellular proliferative phenomena,
the section on molecular signaling. Platelets influx and rapidly such as proliferation of endothelia for neovascularization
form aggregates for haemostasis and also signal to local cells. (Casella et al., 2002) or proliferation of certain fibroblast-
Fibrin and collagen matrices begin to form over a period from lineage cells or in- flammatory cells, reflect classic wound
hours to days, which serve as a scaffold for neutrophil and then responses (Gurtner et al., 2008; Singer and Clark, 1999), while
macrophage and other leukocyte infiltration. Leukocytes then others, such as proliferation of scar-forming astrocytes, are
infiltrate heavily to monitor for pathogens, remove debris, and specific and unique to CNS. This phase is also characterized
provide molecular signals involved in wound repair over a throughout its duration by the absence of a BBB in the lesion
variable number of days or longer depending on severity of core during the period in which damaged blood vessels are
tissue damage or presence of exacerbating factors (Figures 1A replaced by new ones (Figure 1D). As a consequence,
and 1D) (Perry, 2010; Popovich and Longbrake, 2008). endogenous (as well as exogenous) proteins and other
Endogenous mesen- chymal stem cells may also enter the charged molecules can freely diffuse into the surround- ing
lesion core, but their roles neural parenchyma (Bush et al., 1999). These molecules can
are not well understood (Askari et al., 2003; Bianco et al., 2001; include serum proteins (e.g., thrombin, albumin) that signal to
Jaerve et al., 2 wound 12). These basic cellular events reflect classic local
2013;cells,
Gurtner et al., 2008; Singer and Clark, 1999).
im
respon es as found in other tissues (Greaves et al., as Certain
discussedCNS intrinsic cells also respond rapidly to CNS tissue
damage or BBB leak. Live imaging studies in CNS in vivo show munoglobulins, or pathogen-associated
that microglia and NG2-OPC immediately migrate to sites of tis- molecules, below. This leaky BBB creates the
sue damage and BBB leak (Hughes et al., 2013; Nimmerjahn potential for the
lesion core to serve as source of circulating molecules that
generate gradients of molecular signaling that can extend for
considerable distances into neural tissue around the lesion until
the BBB is repaired (Figures 1B, 1D, 2A, and 2B). This
signaling may contribute to the perimeter of tapering gradient
of reactive
Neuron

Review
gliosis and other changes observed in tissue around mature Phase III: Tissue Remodeling
lesions as discussed below. The third phase of response to acute tissue damage generally
During the proliferative phase, cellular elements are generated begins toward the end of the first week after the insult and is
that will in mature lesions form two of the three major tissue distinguished by tissue remodeling that includes events that
com- partments of the mature lesion, (1) the central lesion core are completed within weeks, such as BBB repair and scar orga-
of non- neural tissue and (2) the compact astrocyte scar that nization, as well as chronic events that can continue over many
surrounds
the lesion core (Figures 1B and 1D). Major cellular components months (Figures 1C and 1D). BBB repair or restoration along
of the nonneural lesion core derive from expansion of CNS newly formed blood vessels in the lesion vicinity is generally
intrinsic perivascular fibroblasts and pericytes (Go¨ ritz et al., completed within several weeks after acute uncomplicated in-
2011; Soderblom et al., 2013) and from proliferating endothelial sults and requires the presence of functional astrocytes (Bush
progenitors. The compact astrocyte scar is formed primarily et al., 1999) and pericytes (Daneman et al., 2010; Winkler et al.,
from newly proliferated elongated astrocytes generated by local 2011). The lesion core of nonneural tissue becomes
astroglial progenitors that gather around the edges of severely surrounded by a well-organized, interdigitating compact
damaged tissue containing inflammatory and fibroblast-lineage astrocyte scar by 2– 3 weeks after acute insults not
cells (Figures 1B and 1D) (Faulkner et al., 2004; Sofroniew and exacerbated by ongoing tissue damage (Figures 1C and 1D)
Vinters, 2010; Wanner et al., 2013). It is noteworthy that during (Wanner et al., 2013). These scar- forming astrocytes appear
this proliferative period, the location of compact astrocyte scar actively to corral and surround inflam- matory and fibroblast-
borders is being determined. In mature lesions, these astrocyte lineage cells during tissue remodeling and scar formation
scar borders will precisely demarcate and separate persisting (Wanner et al., 2013). This compact astrocyte scar forms a
areas of nonfunctional, nonneural lesion core tissue from imme- structural and functional border between nonneural tissue in
diately surrounding and potentially functional neural tissue (Fig- the central lesion core and the immediately adjacent and
ures 1B, 2A, and 2B). Cellular and molecular mechanisms that surrounding neural tissue that contains viable cells of all
underlie the determination of precisely where such astrocyte three neural-lineage cell types, astrocytes, oligodendrocytes,
scar borders are formed are incompletely understood but are and neurons (Figures 1C and 2) (Wanner et al., 2013). This
likely to involve a complex interplay and balance of molecular astro- cyte scar border serves as a protective barrier that
signals that on the one hand foster phagocytosis and debris restricts the migration of inflammatory cells from the
clearance and signals that on the other hand foster protection nonneural lesion core into surrounding viable neural tissue.
and preservation of healthy self (Figure 2B), as disc ussed in models lead
Disruption of astrocyte scar formation in different kinds of
more detail below in the section on molecular signals of cell transgenic loss-of-function s to increased lesion size,
damage and death. Several decades of experimental studies increased death of local
on ischemic infarcts show that final lesion size can be neurons, increased demyelination, and decreased recovery of
influenced by subacute metabolic events that continue for function after traumatic or ischemic focal insults (Bush et al.,
hours to days within penumbral zones around the lesions 1999; Faulkner et al., 2004; Herrmann et al., 2008; Li et al., 2008;
(Astrup et al., 1981; Dirnagl et al., 1999; Lo, 2008). In addition, Wanner et al., 2013). After compact astrocyte scar forma- tion,
astrocyte intrinsic signaling cascades have been identified that the second period of tissue remodeling is long lasting, with
play critical roles (Herrmann et al., 2008; Okada et al., 2006; gradual remodeling of lesion core, astrocyte scar border, and
Wanner et al., 2013). A better understanding of the multicellular surrounding perimeter regions (Figures 1D and 2). For
and molecular interactions that determine the location of example, with time there is gradual contraction of lesion cores
astrocyte scar borders around areas of compromised tissue and astro- cyte scars. As transected axons continue to die back
may lead to novel thera- peutic strategies for reducing lesion away from lesions, oligodendrocyte death can continue, leading
size (Figure 2B). These to local tis- sue responses and remodeling. There is long-term
remodeling of ECM in the lesion core, with modulation of
collagen and glyco-
events occur over a time frame of days after the insult protein components (Klapka and Mu¨ ller, 2006; Logan and
(Figure 1D), during which intervention is a realistic possibility. Berry, 2002; Silver and Miller, 2004). In addition, there is
Recent findings also indicate that during the proliferative gradual and chronically ongoing tissue remodeling in reactive
phase after acute CNS damage in the forebrain, neural stem perimeter regions that surround mature lesions, as discussed
cells in periventricular germinal zones give rise to next.
considerable Mature Lesions Exhibit Three Tissue Compartments
numbers of neural progenitors that migrate to the injury sites Astrocyte scar formation around central lesion cores is essen-
in cortex or s triatum (Carmichael, 2006; Kokaia et al., 2012; tially compprogenitor cells, such as contributions to immune
migrating
; Lindvall and Kokaia, 2006). These cells form complicated
Lagace, 201 part regulation, are still in the process of being defined (Kokaia et al.,
of cells in perilesion perimeters (Figures 1B and at this time
of the mi 1C). 2012; Lagace, 2012).
various lines of evidence suggesting that these (Figures 1 an
There are cells tribute beneficially to tissue remodeling in perile- very distinc
may co
sion tissue, but the nature of the contributions made by these
cells remains nebulous. Under normal, unmanipulated circum-
stances, very few newly generated neurons mature and
survive, and other potential contributions by these intriguing,
Neuron 81, January 22, 2014 ª2014 Elsevier Inc. 233
Neuron

Review
lete by 2–4 weeks after acute insults that are
not by prolonged tissue damage, and the lesion
can be considered mature and entering its
chronic stage d 2). In mature lesions, it is useful
to recognize three t lesion compartments: (1) the
central lesion core of
nonneural tissue comprised of locally derived fibroblast-
lineage cells, blood vessels, infiltrating fibrocytes and
inflammatory cells, and extracellular matrix; (2) the compact
astrocyte scar that immediately surrounds the lesion core
and consists of densely packed astrocytes with few if any
neurons or oligoden- drocytes; and (3) the perilesion
perimeter of viable neural tissue that is adjacent to the
compact astrocyte scar and contains all

234 Neuron 81, January 22, 2014 ª2014 Elsevier Inc.


three types of neural-lineage cells (neurons, oligodendrocytes, prevented or attenuated in vivo, inflammation spreads, lesion
and astrocytes) and exhibits a tapering reactive gliosis that size increases, neuronal loss and demyelination are exacer-
grad- ually transitions to healthy tissue (Figure 2). All of these bated, and functional recovery is diminished (Bush et al., 1999;
compart- ments will exhibit further tissue remodeling for weeks Dro¨ gemu¨ ller et al., 2008; Faulkner et al., 2004; Wanner et al.,
to months. Some of this remodeling has the potential to impact 2013). Scar-forming astrocytes are also widely regarded as a,
on functional outcome and may provide targets for therapeutic if not the, major impediment to axon regeneration after CNS
interventions, particularly in the perimeter of neural tissue that injury, based in part on in vitro studies showing astrocyte pro-
exhibits diffuse reactive gliosis (Figures 1D and 2). It is useful to duction of certain proteoglycans that can inhibit elongation
consider separately these compartments and their impact on (Silver et al., 1993). Nevertheless, other studies show that axon
functional outcome. regeneration in vivo occurs along astrocyte bridges and that, in
Lesion Core. Tissue in the mature lesion core contains few the absence of such bridges, axon regeneration does not occur
or no neural-lineage cells and is nonfunctional in neurological and seems more to be impeded by confrontation with
terms. Initially, the main cell types in mature lesion cores nonneural lesion core tissue (Kawaja and Gage, 1991; Williams
include fibroblast-lineage cells, endothelia, fibrocytes, and et al., 2013; Zukor et al., 2013). Transgenic loss-of-function
inflammatory cells (Figure 2A). With time and remodeling, and studies conduct- ed in vivo may help to clarify the precise
in the absence of exacerbating damage or infection, nature and scale of effects exerted by scar-forming astrocytes
inflammatory elements will gradually withdraw, although some on axon regrowth after injury.
may persist for long times. Elements of lesion core will persist Perilesion Perimeters. All mature lesion cores and astrocyte
permanently as fibrotic scar containing various nonneural cells scars are surrounded by perimeters of viable neural tissue that
and ECM. In some areas of lesion core, cells and ECM will exhibits a gradient of tapering reactive gliosis that gradually
recede to leave fluid-filled cysts of quite variable size (Figure tran- sitions to healthy tissue (Figure 2A) (Wanner et al., 2013).
2A) (Tuszynski and Steward, 2012). Functionally, cells and ECM The tissue in these perimeter zones contains all neural-lineage
of lesion core serve as substrates for rapid wound repair and cell types including neurons and oligodendrocytes, and these
tissue replacement. The rapidity of wound closure allowed by cells are intermingled with reactive gliosis that can extend for
fibrotic replacement as compared with parenchymal renewal consid- erable distances (Figure 2A). Reactive perimeter tissue
has been suggested to afford advan- tages in other tissues begins immediately adjacent to compact astrocyte scars, where
(Greaves et al., 2013; Gurtner et al., 2008; Singer and Clark, the elongated cell processes of scar-forming astrocytes overlap
1999). Nevertheless, this mechanism can come with the cost of with hypertrophic reactive astrocytes and reactive microglia
lost tissue functions. In CNS, lesion core tissue has since the that are intermingled with surviving viable neurons and other
19th century been thought to have a negative long-term elements of neural tissue (Figure 2A). It is somewhat
functional impact as an impediment to axon regener- ation, remarkable that viable neurons can be present within a few
confirmed by recent studies as well (Zukor et al., 2013). hundred micro-
Molecular evaluations show that lesion core ECM contains colla- meters of compact astrocyte scars and the lesion core tissue
gens and proteoglycans that poorly support or overtly inhibit filled with potentially cytotoxic inflammatory elements just
regrowth of damaged axons (Hermanns et al., 2006; Kimura- beyond (Bush et al., 1999; Wanner et al., 2013). The reactive
Klapka and Mu¨ ller, 2006; Yoshioka et al., 2010).
Kur- oda et al., 2010; sis in perimetglio- ers includes hypertrophic reactive astrocytes
Compact Astr ocyte Scars (Glial Scars). re astro (Fig and ure 2A). Both the cause and functions of
Matu
consist of relatively narrow zones of newly generated astrocytes of tapering reperimeters active gliosis are not clear.
with elongated processes that intermingle and intertwine exten- Regarding cause, it
sively and that immediately abut and surround on all sides re- from the lesio
is possible that gradients of signaling molecules diffusing out
gions of lesion core tissue (Figure 2A) (Sofroniew and Vinters, leak or prod n core, either entering during the period of BBB
2010; Wanner et al., 2013). These narrow zones of densely inter- may trigger uced there by infiltrating and proliferating cells,
twined elongated scar-forming astrocytes are generally not more among local such a gradient of gradually tapering responses
than a millimeter across and are generally devoid of other neural- tive, the degrglia (Figures 2A and 2B). From a functional
lineage cells (n eurons or oligodendrocytes) (Wanner et al.,
beneficial
are perspec- ee to which specific aspects of this
2013). These sc ar-forming astrocytes are newly proliferated in determined. reactive gliosis or harmful (or some mixture of
response to the insult and the packing density of astrocyte cell Reactive peboth) remains to be

rimeter areas around lesion cores in brain and spi-


bodies in astrocyte scars is up to double that in healthy tissue nal cord are likely to exhibit long-term, and potentially intense,
(Wanner et al.,013). The densely packed elongated scar-form- tissue remode
tions by restricting the spread of inflammatory cells away from
2 ing astrocytesansition seamlessly and rapidly to less densely for formation
tissue lesions caused by trauma, stroke, and autoimmune
tr packed ands densely intertwined hypertrophic reactive as- chael, 2006; When astrocyte scar formation is experimentally
inflammation.
les trocytes thate intermingled with viable neurons and other et al., 2010;
ar elements ofral tissue in the surrounding perimeter of reactive potential to
neu neuralcussed below (Figure 2A) (Wanner et al., 2013). including extr
tissue dis
Transgenic loss-of-function studies show that newly proliferated
compact astrocyte scars exert essential neuroprotective func-
ling, with new synapse formation and the
potential of new relay circuits (Bareyre et al.,
2004; Carmi- Clarkson et al., 2010; Courtine et
al., 2008; Li Overman et al., 2012). Numerous
factors have the impact on this synapse and
circuit remodeling, acellular matrix molecules
such as proteoglycans
in the perineuronal (Garcı´a-Alı´as et al., 2009). Reactive glia
may play important roles in this remodeling, but this topic is
under- studied. In the healthy CNS, astrocytes interact with
neurons on multiple spatial and temporal scales that can
influence or modulate neural function in various ways
(Halassa and Haydon,
Neuron

Review
surrounded by astrocyte scars that
2010). Both astrocytes and microglia have the potential to play
fundamental roles in the synapse remodeling in the perilesion
perimeter (Stephan et al., 2012; Stevens et al., 2007) and the
impact of reactive gliosis on such remodeling is not yet under-
stood. Such perilesion perimeter areas are likely to provide
fertile ground for investing new strategies for interventions that
may influence neural plasticity and relay circuit formation, which
in combination with appropriate rehabilitative training (van den
Brand et al., 2012) have the potential to beneficially influence
functional outcome.
Chronic Focal Insults
In addition to acute focal trauma and stroke, which often serve
as models to study mechanisms of CNS damage as just
discussed, other more chronic forms of damage warrant
consideration. Important chronic insults include focal infections
with abscess formation, tumors, and autoimmune lesions. All of
these condi- tions invoke reactive gliosis and multicellular
responses that have strong similarities with those that occur
after acute focal damage. This multicellular response plays
critical roles in pro- gression, severity, and resolution (or not) of
the insults.
Infection. Focal infections or abscesses can form when
traumatic wounds become infected or can be seeded
spontane- ously, particularly in immune-compromised
individuals. Such infections elicit reactive gliosis with astrocyte
scar formation similar to that after acute focal trauma
(Sofroniew and Vinters, 2010). The time course of lesion
persistence and tissue remodel- ing is protracted and
dependent on resolution of the infection. Transgenic loss-of-
function studies show that astrocyte scar formation is critical to
focal containment of the infection. When astrocyte scars are
disrupted, infection and inflammation spread rapidly through
adjacent neural tissue with devastating effects (Dro¨ gemu¨ ller et
al., 2008).
Cancer. Both primary and metastatic tumors elicit reactive
gliosis and multicellular responses that have similarities to other
forms of focal tissue damage. Interactions of tumor cells with
CNS glia and infiltrating inflammatory cells are complex and
heavily dependent on tumor cell type. Noninvasive tumors are
surrounded by reactive gliosis and encapsulating scar similar
to that seen around traumatic tissue damage. Aggressively
inva- sive tumors are not encapsulated or surrounded by well-
defined astrocyte scars but evoke other forms of reactive gliosis
and multicellular responses. Successful infiltration and spread
of certain tumor cell types is thought to be associated with their
ability to create an environment for growth, vascularization,
and spread, which may include successfully neutralizing bar-
rier-forming functions of local CNS cells and co-opting
programs for creating proliferative niches (Louis, 2006; Silver et
al., 2013; Watkins and Sontheimer, 2012).
Autoimmune Disease. Focal autoimmune lesions are inter-
esting to consider from the perspective of being a response
to CNS damage. Although CNS autoimmune disease is often
viewed as diffuse, chronic, and caused primarily by
dysfunctions of the peripheral immune system, individual
autoimmune lesions bear many similarities to acute focal
traumatic wounds. For example, active multiple sclerosis
plaques are filled with inflam- matory cells and chronic plaques
exhibit central lesion cores that are devoid of all neural-lineage
cell types (no neurons, oligoden- drocytes, or astrocytes)
Neuron 81, January 22, 2014 ª2014 Elsevier Inc. 235
Neuron

Review
synaptic in- teractions and neural circuit functions (Figure 3B).
separate nonneural tissue from perimeters of reactive gliosis A better
in tissue with all three neural cell types (Frohman et al., 2006;
Luc- chinetti et al., 1996) in manners similar to tissue
damage caused by trauma or ischemia (Figures 1 and 2).
Neuromyelitis optic (NMO) is an autoimmune inflammatory
disease in which causal autoantibodies are directed against
aquaporin-4 on the astro- cyte cell membrane, and NMO
lesions are associated with com- plement-mediated, lytic
destruction of astrocytes (Lennon et al., 2005; Popescu and
Lucchinetti, 2012). There is remarkable congruence
between clinical evidence from NMO and experi- mental
loss-of-function studies demonstrating that transgeni- cally
mediated ablation or attenuation of scar-forming astrocytes
results in severe exacerbation of autoimmune CNS
inflammation (Haroon et al., 2011; Voskuhl et al., 2009).
Thus, both clinical and experimental evidence implicate
critical roles for scar-forming astrocytes in limiting the spread
of autoimmune CNS inflamma- tion. These observations
strongly suggest that dysfunction of CNS intrinsic cells such
as astrocytes may contribute causally to the onset or
progression of CNS autoimmune conditions in ways not
generally considered by research focused only on trying to
identify causal mechanisms of the peripheral immune
system. This notion may be further supported by
suggestions that a subgroup of patients with multiple
sclerosis have dis- ease-related autoantibodies against the
potassium channel Kir4.1, which in the CNS is located on
astrocyte membranes (Sri- vastava et al., 2012). It will be
interesting to determine the extent to which gain or loss of
astrocyte functions contributes to the pathophysiology of
CNS autoimmune conditions.

Response to Diffuse Insults


Diffuse insults to CNS tissue can be associated with
neurode- generative diseases, certain seizure disorders, or
mild traumatic brain injury. Diffuse insults tend initially to be
less intense than acute focal damage caused by trauma or
stroke and may not initially cause overt tissue damage but
instead accumulate grad- ually over chronic periods of time.
As the tissue damage be- comes more severe, small
individual lesions form, each of which invokes reactive
gliosis and multicellular responses similar to those caused
by acute tissue damage with breakdown of the BBB,
inflammation, and recruitment of leukocytes (Figures 1 and 2)
but on a smaller scale. Over time, the diffuse damage
comes to resemble a diffuse collection of many small focal
lesions that are intermingled and can be dispersed over
large areas (Figure 3A). Each of these small lesions gives
rise to its own perilesion perimeter zones of tapering reactive
gliosis, with the consequence that overlapping zones of
severe hyper- trophic reactive gliosis and multicellular
responses can be distributed over large areas of tissue
(Figure 3A). It is important to realize that these many small
lesions are interspersed among viable neurons and neural
circuitry, which become enveloped by these overlapping
perilesion perimeters of reactive gliosis (Fig- ures 3A and
3B). As discussed above, there is now substantive evidence
for the participation of astrocytes and microglia in normal
neural circuit function, but the impact of reactive gliosis on
such functions is not known. In the context of diffuse CNS
insults, large areas of functioning neural tissue may be
exposed to intense reactive gliosis that is likely to impact on

236 Neuron 81, January 22, 2014 ª2014 Elsevier Inc.


Figure 3. Diffuse Tissue Damage Leads to Extended Regions of Reactive Gliosis
(A) Small chronic insults can lead to diffuse tissue damage with formation of dispersed small lesion cores (LC), astrocyte scars (AS), and perilesion perimeters
(PLP) that can overlap and coalesce into regions of contiguous reactive gliosis that can extend over large areas.
(B) Reactive astrocytes and reactive microglia in PLP can impact on synapses and neuronal functions in various ways.

understanding of such effects may open inroads to new thera- astrocytes are
peutic strategies.
Neurodegenerative Disease
Reactive gliosis and multicellular responses are triggered in
different and selective ways in different neurodegenerative dis-
eases. In some cases, the trigger can be accumulation of extra-
cellular toxins, such as b-amyloid in Alzheimer’s disease (AD)
(Prokop et al., 2013; Zlokovic, 2011), which can accumulate
and cause many small areas of focal tissue damage that trigger
gliosis with many similarities to other forms of diffuse insults
(Figure 3A). In other cases, the trigger can be neuronal or
synap- tic damage or death secondary to neuronal intrinsic
changes. Some conditions, such as amyotrophic lateral
sclerosis (ALS) or Huntington’s disease, cause intrinsic cellular
changes in both neurons and glia that can perturb cellular
functions and in- teractions, which can serve as triggers (Boille´
e et al., 2006; Mar- agakis and Rothstein, 2006).
Neurodegenerative insults can also lead to disturbance of the
neurovascular unit, resulting in BBB leak, which can serve as a
further trigger for reactive gliosis and also signal to recruit
blood-borne inflammatory cells (Zlo- kovic, 2008, 2011). In
many cases, reactive gliosis and multicel- lular responses may
not be apparent at onset or in early stages of the
neurodegenerative conditions but appear later and may then
become involved in disease progression. Reactive gliosis trig-
gered in various ways, by accumulation of abnormal proteins,
ischemic or traumatic cellular damage, or microbes, can also
lead to BBB leak via specific molecular signaling cascades as
discussed below, with resultant inflammation that may con-
tribute to disease progression.
The contributions of glial cells and reactive gliosis to
neurode- generative conditions are complex and only beginning
to be understood. Glia not only become reactive in response to
degen- erative cues as just mentioned, but can also participate
in trying to combat them. For example, both microglia and
thought to contribute to clearing b-amyloid (Prokop et al., 2013;
Wyss-Coray et al., 2003), and attenuation of reactive
astrogliosis can increase b-amyloid load in a transgenic mouse
model of AD (Kraft et al., 2013). In addition, glia may be
affected by the dis- ease-causing molecular defects and
become dysfunctional, further complicating efforts to
understand the roles of reactive gliosis in the disease process.
For example, in ALS, glial cell dysfunction is causally
implicated in mediating neuronal degen- eration (Yamanaka et
al., 2008), and ongoing reactive gliosis and inflammation are
also thought to contribute to disease progres- sion (Maragakis
and Rothstein, 2006). Distinctions between disease-induced
dysfunctions of glial cells and the reactive re- sponses that are
triggered in glial cells by cell and tissue damage are
sometimes blurred or equated but should not be. Under-
standing these different phenomena and the ways in which
they interact is likely to have important consequences for
under- standing the mechanisms that underlie cellular
pathophysiology and drive disease progression. It is important
to remember that although some aspects of glial reactivity may
contribute to dis- ease progression, others are likely to be
protective. Defining the molecular basis of such differences
may help to identify novel therapeutic strategies.
In the context of neurodegenerative disease, it is also inter-
esting briefly to consider the potential for non-cell-autonomous
neuronal degeneration precipitated by dysfunction of glia or
other nonneural cells. Loss- or gain-of-function genetic muta-
tions in microglia (Derecki et al., 2012) or astrocytes (Brenner
et al., 2001; Rothstein, 2009; Tao et al., 2011) have the
potential to cause non-cell-autonomous degeneration of
different types of neurons in different human diseases and
animal experimental models. Such observations indicate that
glial cell dysfunction may be the primary causal event in
certain neurological condi- tions. Such observations also raise
the possibility that dysfunc- tion of specific aspects of reactive
gliosis during responses to
CNS insults may exacerbate damage or lead to worse tissue Molecular Signaling and Multicellular Interactions
repair and worse outcome. The potential for genetic polymor- Molecular signaling among the different cell types that respond
phisms to impact on glial cell functions and responses to CNS to CNS insults is complex, combinatorial, and densely inter-
injury is an as yet untapped area that may reveal causal factors woven. Different cells have the capacity to produce and
underlying differences in the responses of different individuals respond to similar sets of molecules, and there are hints that
to seemingly similar types and severities of CNS insults. some mole- cules may coordinate multicellular responses.
Epilepsy Many intercellular signaling molecules have been identified, but
Astrocytes and microglia can be involved in epilepsy in multiple we are in the early stages of determining how these multiple
ways. Astrocytes play multiple critical roles in regulating signals regulate multicellular interactions and the temporal
synapse activity and neuronal excitability and astrocytes may progression from one phase to another during responses to
play critical roles in the generation of seizure activity (Devinsky specific CNS insults. Here we provide a general overview of
et al., 2013; Jabs et al., 2008; Wetherington et al., 2008). In molecular functional classes (Figure 4A) and a few
addition, reactive gliosis of both microglia and astrocytes is a representative examples of the many specific molecules that
prominent feature of chronically epileptic neural tissue, which regulate or influence CNS cellular responses to insults (Table
can also exhibit BBB breakdown and multicellular 2).
inflammation (Devinsky et al., 2013; Jabs et al., 2008; Cell Damage and Death
Wetherington et al., 2008). With increasing loss of neurons, Cells that are dead, dying, or temporarily damaged but recover-
tissue sclerosis sets in, which has similarities with severe able release or express molecules that signal damage. These
diffuse astrogliosis due to other causes as discussed above, molecules fall into many categories including neurotransmitters,
with many smaller tissue lesions sur- rounded by a continuum cytokines, chemokines, neuroimmune-regulators (NI-Regs),
of perilesion tissue with a mix of surviv- ing neurons and and danger-associated molecular patterns (DAMPs) that
increasingly severe hypertrophic gliosis (Figure 3A). The stimulate reactive gliosis and other cellular responses including
impact of reactive gliosis and the multicellular response to debris clearance by immune cells (Figures 4B and 4C) (Table
tissue damage on the surviving neurons in the peril- esion 2). Insults of different types and severities release different
perimeter regions is not well understood (Figure 3B) but may combinations of these molecules, which in turn trigger different
contribute to reduced seizure thresholds. Selective experi- responses. For example, mild cellular insults can subtly elevate
mental induction of astrocyte reactivity is associated with a extracellular concentrations of glutamate or ATP that attract
reduction of inhibition in local hippocampal neurons (Ortinski microglial cell processes without initially unleashing full-blown
et al., 2010). More work in this area is warranted. inflammation (Figure 4B) (Davalos et al., 2005; Liu et al., 2009).
Diffuse Traumatic Brain Injury ATP can also elicit calcium responses in astrocytes and cause
Large focal lesions caused by severe focal traumatic brain con- nexin-dependent ATP release that increases extracellular
injury (TBI) (contusion or crush) have been discussed above. ATP levels and amplify this ‘‘danger’’ signal (Figure 4B). More
Neverthe- less, clinically relevant TBI is often diffuse and widely severe cell damage or death will release additional DAMPs and
distributed. The cellular and molecular mechanisms of diffuse alarmins in the form of cytosolic and nuclear contents such as
TBI that cause functional disturbances without large focal K+, heat shock proteins such as ab-crystallin, S100 family cal-
lesions are incom- pletely understood. Diffuse TBI is generally cium-binding proteins, DNA-binding high mobility group box 1
characterized by small foci of vascular breakdown and tissue (HMGB1), as well of DNA and RNA. All of these molecules can
damage that can be diffusely distributed over large areas of serve as ‘‘danger’’ signals to alert innate immune cells to tissue
CNS (DeKosky et al., 2013; McIntosh et al., 1989). It is injury and promote clearance of ‘‘altered self’’ cellular debris via
interesting to consider that each of these small areas of tissue activation of pattern recognition receptors (PRRs) on
or cellular damage may represent a small focal injury with phagocytic innate immune cells (Figure 4C) (Chan et al., 2012;
compartments equivalent to core, astrocyte scar, and Griffiths et al., 2010; Kono and Rock, 2008). Molecules such as
perimeter, which may be spread over large areas without an beta-amyloid (Ab) produced during neurodegenerative
obvious single large focal region (Figure 3A). As discussed disorders can also act as alarmins (Figure 4C). A single type
above, the interspersed viable neural tissue in the many of alarmin may bind multiple classes of PPRs. For example,
overlapping perilesion perimeter regions will be exposed to HMBG1 elicits proinflammatory signaling through activation of
intense hypertrophic reactive gliosis and inflam- mation, which multiple TLRs, receptor for advanced glycation end products
can impact on neural functions (Figure 3B). Spe- cific cellular (RAGE), and macrophage antigen complex-1 (MAC1) (Chan et
and molecular mechanisms of reactive gliosis and multicellular al., 2012; Gao et al., 2011), and S100 activates multiple TLRs
responses triggered by mild or moderate diffuse TBI are only and RAGE (Chan et al., 2012).
beginning to be characterized. Newly proliferated reactive Healthy neurons and glia near CNS insults express neuroim-
astrocytes exert protective functions essential for neuronal mune regulatory molecules (NI-Regs) and self-defense proteins
survival (Myer et al., 2006). Microglia can exert a broad range that signal ‘‘healthy self’’ and serve to confine potentially
of effects depending on specific molecular stimuli, which either noxious proinflammatory signaling and prevent phagocytosis of
protect or exacerbate tissue loss (Hanisch and Ketten- mann, viable cells (Figure 4C) (Table 2) (Griffiths et al., 2010). These
2007). Understanding and manipulating the reactive glio- sis ‘‘healthy self’’ molecular signals include both membrane-bound
and multicellular sequelae triggered by mild to moderate TBI and soluble NI-Reg molecules. For example, sialic acids (SAs)
has enormous potential to identify new therapeutic targets in a in membranes of healthy cells interact with SA receptors
field of high incidence and importance. Emerging molecular (SARs), including Siglecs, on innate immune cells and serve
mechanisms are discussed in the next section. as ‘‘don’t
Figure 4. Multimolecular Regulation and Graded Responses to Damage
(A) Responses to CNS damage are regulated and influenced by diverse categories of molecular signals derived from many types of CNS intrinsic and
extrinsic cells.
(B)Low levels of ATP released by cells during mild CNS insults can act through (1) P2X receptors to induce rapid chemotaxis of microglial processes and (2)
P2Y receptors on local astrocytes to evoke calcium signaling and connexin-dependent ATP release, which can increase extracellular ATP levels and thereby
amplify this ‘‘danger’’ signal.
(C) After severe injury, signaling molecules from ‘‘altered self’’ and ‘‘healthy self’’ vie to balance debris clearance and preservation of viable tissue. Severely
damaged or dead cells release a number of hydrophobic intracellular molecules (left), or ‘‘alarmins,’’ which express damage-associated molecular patterns
(DAMPs). These ‘‘danger’’ signals alert CNS innate immune cells to tissue injury and promote clearance of debris via activation of pattern recognition
receptors on phagocytic immune cells (center). Viable CNS intrinsic cells at lesion borders (right) express membrane-bound and soluble neuroimmune
regulatory molecules to prevent attack and phagocytosis by immune cells.

eat me’’ signals (Figure 4C) (Varki and Gagneux, 2012). CD200 (Griffiths et al., 2010). Complement regulatory proteins (CRPs)
and CD47 similarly act through receptors CD200R and SIRP-a can reduce complement activation (Figure 4C) (Griffiths
on inflammatory cells to prevent phagocytic attack (Figure 4C) et al., 2010). Thrombomodulin (CD141) binds and sequesters
Table 2. Major Classes and a Few Specific Examples of Intercellular Signaling Molecules that Regulate Cellular Responses to CNS
Damage
Molecule
Molecule Source
Source LesionLesion
Compartment Phase Targets
CompartmentPhaseTargets and Effects
and Effects
Class: Protease and Related
Thrombin S LC, AS I Clot formation; astrocyte proliferation
MMP-9 M, O, S LC, AS, PLP I-III OPC proliferation, remyelination; BBB leak; neovascular remodeling
Kallikreins A, M, N, S LC, AS, PLP I-III Proinflammatory; demyelination
Serpins A, M, O LC, PLP I-III Inhibit deleterious protease

Class: NTs
ATP, GlutamateN, O, ALC, AS, PLPIMicroglia chemotaxis; reactive astrogliosis
Class: DAMPs
Alarmins: HMGB1, Damaged Cells LC, PLP I.II ‘‘Altered self’’ signals; proinflammatory; increase phagocytosis
S100s, DNA
PAMPs: LPS Microbes LC, PLP I.II ‘‘Nonself invader’’ signals; proinflammatory; increase phagocytosis
Class: Growth Factors, Morphogens
FGF A, N, E, LC, AS, PLP I-III Fibrotic scar; ECM; neovascular remodeling; reactive astrogliosis
VEGF E, P, F, A LC, AS I-III BBB permeability; neovascular remodeling
PDGF-B E, A LS, PLP I-III Neovascular remodeling
PDGF-A Remyelination; OPC Proliferation
Endothelin, N, A, O LC, AS II, III Astrocyte proliferation, glial scar formation
EGF, BMP
Class: Cytokines, Chemokines
TGFb A, M, L AS, PLP I-III Fibrotic scar formation; ECM production; astrocyte proliferation
IL-1b, TNFa, INFg A, M, L LC, AS, I, II Proinflammatory regulation
IL-6, CCL2 A, M, L LC, AS I, II Leukocyte instruction, astrocyte scar formation
Class: NIRegs
CD141 A, O, E LC, AS I-III Protection of healthy self; resolution of inflammation
CD200, CD47 N LC, AS, PLP I, II Protection of healthy self
Class: Neural Remodeling
Neurotrophins, N, A PLP III Synapse remodeling
BDNF, etc.
Perineuronal net A, OPC PLP III Restrict terminal sprouting
Thmbs, C1q A, M PLP III Synapse formation and pruning
A, astrocyte; E, endothelia; F, fibroblast; L, leukocyte; M, microglia; N, neuron; NTs, neurotransmitters; O, oligodendrocyte; OPC, O progenitor cell;
S, serum; Thmbs, thrombospondin.

HMGB1, thereby reducing alarmin bioavailability (Figure 4C) described, but also molecules entering via leaky BBB,
(Abeyama et al., 2005). molecules released by infiltrating leukocytes, and molecules
Thus, specific receptor-mediated signals that indicate released by local cells including reactive glia themselves
‘‘altered self’’ or ‘‘healthy self’’ can tailor the nature of the (Figure 5A) (Sofroniew, 2009). In response to these many
reactive gliosis and multicellular responses to the type and different and often combinatorial molecular signals, reactive glia
severity of cellular damage or death (Figures 4B and 4C). It is alter their gene expression, structure, and function in selective
important to note that the final level of cellular damage inflicted and specific ways, depending on the type and severity of the
by insults of all kinds is determined in part by prolonged periods insults and de- pending on the specific combinations of
of different types of secondary events (Figure 1D), including a molecular signals that are impinging on them. It is important to
balancing act among innate immune mechanisms that regulate emphasize that glia do not exist in simple all-or-none
the clear- ance of ‘‘altered-self’’ debris and ‘‘non-self’’ ‘‘quiescent’’ or ‘‘activated’’ states and that there is no single
pathogens, while preserving ‘‘healthy self’’ (Figure 4C) (Griffiths program of ‘‘reactive gliosis’’ that is triggered in an all-or-none
et al., 2010). These events will impact on final size, location of fashion and is similar in all situations. Instead, reactive glia can
glial scar, and signaling to perlesion perimeters (Figure 2). exhibit a vast range of re- sponses as determined by
Reactive Gliosis combinations of specific signaling events. For example,
Reactive gliosis is regulated by a large array of different exposure to PAMPs such as lipopolysac- charide (LPS) can
extracel- lular signals generated after CNS insults. This array markedly skew the transcriptome of reactive astrocytes toward
includes not only molecules released by damaged or dead chemokines, cytotoxicity, and inflammation (Hamby et al.,
cells as just 2012; Zamanian et al., 2012). It is also noteworthy
Figure 5. Multicellular and
Multimolecular Regulation of Reactive
Gliosis and BBB
(A)Reactive gliosis can be induced, regulated,
and influenced by a wide variety of molecular
signals that can derive from many types of CNS
intrinsic and extrinsic sources.
(B) BBB permeability can be regulated by spe-
cific molecular signaling cascades involving in-
teractions among different cell types that
converge on endothelial tight junctions (Argaw et
al., 2009, 2012; Seo et al., 2013).

late (1) local reactive astrocytes to


release VEGF and (2) local NG2-OPCs
to release MMP-9, which in turn
influence tight junc- tions and barrier
properties of local endo- thelial cells and
allow entry of serum proteins (including
IgGs and signaling proteins such as
thrombin, albumin, pro- teases, etc.) and
leukocytes into local CNS parenchyma
(Figure 5B) (Argaw et al., 2009, 2012;
Seo et al., 2013). Entering leukocytes
have the potential to generate more IL-1b
and sustain local BBB permeability,
creating the potential for a feedforward
loop under adverse cir- cumstances.
Understanding the molecu- lar
mechanisms whereby such signaling is
downregulated and the BBB reseals may
help to reveal rational therapeutic in-
terventions for certain conditions. Repair
of the BBB after an uncomplicated acute
injury generally occurs within several
weeks (Figure 1D).
Astrocyte Scar Formation
Formation of compact astrocyte scars is
a specialized aspect of reactive
astrogliosis that occurs in response to
severe tissue damage and leukocyte
infiltration and involves phases of cell
proliferation and cell organization (Figure
1) (Sofroniew and Vinters, 2010; Wanner
that different combinations of stimulatory factors can lead to et al., 2013).
synergistic changes in molecular expression that could not be Molecular signals that regulate astrocyte
predicted from individual effects (Hamby et al., 2012). proliferation signals after CNS damage can derive from serum
Information about the effects mediated by reactive glia in proteins or local cells and include thrombin, endothelin, FGF2,
response to specific signals is also steadily increasing and it is ATP, bone morphogenic proteins (BMPs), sonic hedge hog
clear that many aspects of reactive gliosis are part of coordi- (SHH), and others (Table 2) (Gadea et al., 2008; Neary et al.,
nated multicellular innate and adaptive immune responses to 2003; Sahni et al., 2010; Shirakawa et al., 2010; Sirko et al.,
CNS insults. It is interesting to consider that certain molecules 2013). The location of astrocyte scar borders around lesions
are emerging as potential coordinators of multicellular re- can be influenced by interactions among proliferating or newly
sponses. One interesting set of converging data implicates IL- proliferated scar-forming astroglia, fibroblast-linage cells, and
1b as a key trigger that stimulates different types of glial cells inflammatory cells (Wanner et al., 2013) and can be modulated
to modulate BBB permeability to serum proteins and leukocytes by transgenic manipulation of astrocyte intrinsic signaling cas-
during reactive gliosis in response to CNS insults (Figure 5B). cades involving STAT3, SOCS3, or NFkB, which can increase
Numerous DAMPs and PAMPs generated by CNS insults will or decrease lesion sizes (Brambilla et al., 2005; Herrmann
stimulate IL-1b release from microglia, and this IL-1b will stimu- et al., 2008; Okada et al., 2006; Wanner et al., 2013), providing
targets for potential intervention. Signals for organization of
newly proliferated astrocytes into compact scars include
the
after acute
IL-6 receptor-STAT3 signaling system, which critically regulates
astrocyte scar formation after trauma, infection, and autoim-
mune inflammation (Dro¨ gemu¨ ller et al., 2008; Haroon et al.,
2011; Herrmann et al., 2008; Wanner et al., 2013).
Neural Remodeling in Perilesion Perimeters
Reactive gliosis is not only important in responses to very local
CNS damage, but also is likely to play important roles in the
neu- ral remodeling that continues for prolonged times in
perilesion perimeters after acute focal insults such as stroke
and trauma (Figures 1 and 2), as well as in chronic diffuse CNS
insults (Figures 3) including neurodegenerative conditions such
as Alz- heimer’s disease. Such perilesion perimeter regions can
extend for considerable distances away from large focal lesions
(Figures 1 and 2) or can cover large areas of diffuse tissue
damage (Fig- ures 3). In such cases, the associated reactive
gliosis has the po- tential to impact on substantial territories of
functioning neural tissue. There is mounting evidence that the
diverse changes in molecular signaling associated with this
diffuse reactive gliosis (Table 2) will impact on the ongoing
neural remodeling and changes in neural function. Astrocytes
interact with neurons on multiple spatial and temporal scales
that can influence or modu- late neural function in various direct
and indirect ways, including by modulation of the extracellular
balance of ions, transmitters, and water critical for neural
function, as well as direct influences on synaptic activity directly
via release of ‘‘gliotransmitters’’ (Halassa and Haydon, 2010;
Hamilton and Attwell, 2010; Henne- berger et al., 2010;
Volterra and Meldolesi, 2005). Microglia are also implicated in
participating in the regulation of synaptic turn- over (Stephan et
al., 2012). Perineuronal net molecules such as chondroitin
sulfate proteoglycans generated by astrocytes and NG2-OPC
influence synaptic plasticity (Table 2) (Wang and Faw- cett,
2012). The effects of reactive gliosis on such functions is only
beginning to be understood, but reactive astrogliosis has been
reported to impact on neural functions in various ways,
including (1) by downregulation of astrocyte glutamine
synthase that is associated with reduced inhibitory synaptic
currents in local neurons (Ortinski et al., 2010), (2) increased
expression of xCT (Slc7a11), a cysteine-glutamate transporter
associated with increased glutamate signaling, seizures, and
excitotoxicity (Buckingham et al., 2011; Jackman et al., 2010),
and (3) changes in the expression of multiple GPCRs and G
proteins and calcium signaling evoked by their ligands that
have the potential to alter astrocyte-neuron interactions (Hamby
et al., 2012). There is also a growing body of evidence that in
response to inflammatory mediators and cytokines, reactive
astrocytes can modulate their expression of multiple molecules
such as transmitter and ion transporters, neuromodulators such
as nitric oxide and prosta- glandins, growth factors, and
synapse regulatory proteins, which can impact on synaptic and
neuronal functions including com- plex behaviors such as
sickness behavior, pain, appetite, sleep, and mood, as
reviewed in more detail elsewhere (Sofroniew, 2013). Taken
together, such findings provide evidence that different
molecular mediators and signaling mechanisms associ- ated
with reactive gliosis can modulate functions of reactive as-
trocytes and microglia in different ways that have the potential
to impact on synaptic and other neuronal functions in perilesion
perimeters. Such signaling mechanisms are likely to have
effects on the efficacy of training and functional rehabilitation
lasting neural remodeling that occurs in perimeter tis- sue. The
damage such as trauma or stroke, as well as effectors to mix of molecular signals generated in lesion cores
mitigate functional deterioration during degenerative
conditions. Under- standing and manipulating the signaling
mechanisms and effects of reactive gliosis in perilesion
perimeter tissue has the potential to reveal novel therapeutic
strategies (Gleichman and Carmichael, 2013).
Blood-Brain Barrier
The blood-brain barrier (BBB) is generated and regulated by
a complex multicellular neurovascular unit that involves
critical interactions among endothelia, pericytes, astrocytes,
and other cells (Abbott et al., 2006; Zlokovic, 2008).
Compromise of the BBB can occur in various manners and
to various degrees, ranging from specific molecular signaling
events that open BBB permeability during certain types of
reactive gliosis as dis- cussed above (Figure 5B), to severe
breakdown of the BBB caused by traumatic or ischemic
endothelial destruction result- ing in the need for
neovascularization (Figure 1). In addition to overt trauma
and ischemia, considerable evidence implicates a central
role for BBB dysfunction in aging and various neurode-
generative disorders. For example, both transgenically
induced and aging-related pericyte deficiencies result in
BBB breakdown that precedes neuronal degeneration (Bell
et al., 2010). Pericyte deficiency is also apparent in
postmortem amyotrophic lateral sclerosis tissue (Winkler et
al., 2013) and BBB dysfunction pre- cedes
neurodegeneration in a murine model (Zhong et al., 2008).
Polymorphisms in APOE isoforms are implicated in
increased vulnerability to BBB breakdown in
neurodegenerative disorders and traumatic injury (Bell et al.,
2012).
Regardless of triggering events, BBB leaks lead
immediately to CNS parenchymal entry of serum proteins
such as thrombin, albumin, immunoglobulins, and proteases
(Figure 5B) that can have a wide range of direct and indirect
effects. Thrombin can signal directly via protease-activated
receptors on various CNS cells including microglia and
astrocytes (Shigetomi et al., 2008; Suo et al., 2002).
Immunoglobulins contribute to clearing of path- ogens but
may contribute to autoimmune dysfunctions. Platelets can
enter and form aggregates for clotting and release PDGFs,
important for neovascular remodeling as well as for signaling
to pericytes and NG2 cells. Metaloproteases act on multiple
pro- teins and contribute to extracellular matrix and tissue
remodeling (Table 2). Serum kallikrein proteases activate
inflammation. Tis- sue kallikreins, which can interact with
serum proteases as part of larger proteolytic cascades, are
implicated in oligodendrocyte pathology, demyelination,
axonopathy, and neuronal degenera- tion (Burda et al.,
2013; Radulovic et al., 2013). Serpins inhibit and balance
protease effects (Table 2).
In response to signals associated with BBB leak, local glial
cells produce cytokines and chemokines that attract
leukocytes, which on arrival produce additional cytokines
and chemokines. The result is a rich extracellular mixture of
molecular signals that can instruct or influence multiple cell
types in lesion cores and can also diffuse away into
surrounding CNS parenchyma and influence neural cells
that may not have been involved in the initial insult (Figure
2). These molecular gradients may influ- ence or take part in
shaping cellular events in the perilesion pe- rimeters such as
the long-lasting gradient of reactive gliosis and the long-
changes over time as the injury response progresses through Impact of Peripheral Infections on Responses to
different phases and the BBB leak is repaired and different cell CNS Insults
types occupy this area and produce different effector molecules Many of the cell types responding to CNS damage are exqui-
(Figure 1D; Table 2). Depending on the severity of the insult, sitely sensitive to molecular cues associated with microbial
lesion cores exhibit a BBB leak for at least several days up to infection. This is not surprising because limiting infection spread
2 weeks throughout the initial period (Phase I) of damage is likely to have shaped the evolution of injury responses in all
response (Figure 1D). BBB repair requires the presence of tis- sues including CNS. Various features of the response to
func- tional astrocytes (Bush et al., 1999). Release of the infection such as production of cytotoxins and certain types of
protein HMGB1 by reactive astrocytes promotes endothelial inflamma- tion can damage host cells as well as microbes.
cell neo- vascular remodeling, whereas blockade of HMGB1 Triggering such responses in the absence of overt infection has
release by reactive astrocytes prevents neovascular the potential to cause or exacerbate tissue damage. Thus,
remodeling and worsens neurological deficits (Hayakawa et al., certain responses that may be beneficial in microbial infection
2012). In com- plex, chronic, or diffuse insults, BBB leaks may may be detrimental if triggered during sterile (uninfected) tissue
persist and have long-lasting effects on extended areas of damage after trauma, stroke, degenerative disease, or
neural tissue in perilesion perimeters (Figure 3), and late BBB autoimmune attack. In this regard, it is important to recognize
disruption may lead to secondary tissue injury (Zlokovic, 2008). that reactive glia, both microglia and astrocytes, that are
Even when BBB leaks are repaired, lesion cores continue to responding to a sterile primary CNS insult can be influenced by
contain collec- tions of nonneural cells within the BBB that have peripheral infections that generate high levels of circulating
the potential to continue producing high levels of many different cytokines and other inflammatory mediators such as LPS and
signaling mole- cules for prolonged periods. Thus, lesion core other PAMPs. This signaling can occur through blood-borne
tissue has the po- tential to generate diffusion gradients of cytokines or LPS released at the site of peripheral infection and
many potent molecules that spread into neighboring tissue entering through the leaky BBB in the injury core during phase I
parenchyma and form gradi- ents of molecular signals that of the damage response after focal insults or through BBB
influence host cells (Figure 2). It is also noteworthy that while leaks triggered by neurodegenerative or autoimmune
the BBB is open, there is also the potential for entry of various processes (Figures 1A and 5A). LPS and cytokines have
pathogen-associated molecular patterns (PAMPs) (Table 2) powerful effects both on reactive microglia (Perry, 2010; Perry
that are generated by microbial infections in the periphery and et al., 2007) and on reactive astro- cytes (Hamby et al., 2012;
that can substantively alter the response of local cells to CNS Zamanian et al., 2012) that can drive transcriptome profiles and
damage, even when there is no local infection, as discussed wound response toward proinflam- matory and potentially
below. cytotoxic phenotypes over prolonged times in ways that would
Infiltrating Leukocytes not normally be implemented in sterile CNS insults and that can
Leukocytes that infiltrate from the circulation provide a major exacerbate tissue damage. Congruent with such experimental
source of molecular signaling during responses to CNS observations is clinical epidemiological evidence indicating that
damage. These molecular signals include a diverse mix of peripheral infections have a negative impact on neurological
cytokines, chemokines, and growth factors (Table 2) that can outcome after spinal cord injury (Failli et al., 2012).
instruct and modify the functions of local reactive glia
(Figure 5A) and other intrinsic CNS cells as well as other local Common Features, Differences, and Building Models
leukocytes. It is likely that the molecular signals produced by Given the large number of potential CNS insults and the
leukocytes, and their functional effects, will vary during different consid- erable amount of variation among the multicellular
phases of response to acute or chronic insults (Figure 1) and responses they elicit, looking for common features and clear
range from those mediating classical inflammatory responses differences may provide valuable insights regarding shared
with cytotoxic and phagocytic activities, to those influencing fundamental mechanisms. Understanding basic factors that
tissue repair and remodeling. Although temporal changes drive responses to one type of insult may inform ideas
during wound responses are not yet well studied, different regarding responses to another. In this regard, it is interesting
functional roles for different leukocyte subtypes are gradually to consider that evolu- tionary pressures shaping responses to
coming into focus. For example, in response to stimulation by CNS damage are likely to have favored rapid responses to
different cytokines, monocytes generate macrophages with small CNS injuries that were not functionally incapacitating and
distinct functional phenotypes that produce different effector that kept such injuries small and uninfected or that kept small
molecules (Mosser and Edwards, 2008). In response to INFg, infections from spreading. One efficient means of doing so
classically activated macrophages generate cytotoxic antimi- would be to isolate small focal injuries or small infections with
crobial molecules and phagocytose dead cells and debris cellular barriers that effectively wall off these lesions, allowing a
(Figure 4C), whereas in response to stimulation with IL-4 or IL- robust inflammatory (and antimicrobial) responses in lesion
10, macrophages take part in wound healing and anti-inflam- cores, while preserving as much adjacent neural tissue as
matory activities (Mosser and Edwards, 2008). Similarly, possible (Sofroniew, 2005). In this Review, based on available
T cells stimulated by different cytokines become polarized information, we present a basic model of multicellular
toward different activities, such as T H1, TReg, and TH17 cells, responses to different types of focal CNS tissue damage that is
which can exert very different effects ranging from cell killing compatible with this notion (Figures 1, 2, and 3). Certain
to participation in tissue repair (MacIver et al., 2013; Mills, 2011; common features in this model of cellular re- sponses to CNS
Walsh and Kipnis, 2011). damage could be interpreted as serving the
Figure 6. Lesions as Targets for Therapeutic Strategies
Nonneural lesion cores (LC) represent targets for biomaterial depots (A) or grafts of exogenous neural stem/progenitor cells (B), or mesenchymal stem cells
(C) as potential therapeutic strategies. Molecules delivered by biomaterial depots (A) or certain cell grafts (C) have potential to support and attract axon
growth (arrow a), modulate cells in perilesion perimeter (PLP) (arrow b), modulate cells in LC (arrow c), or modulate astrocyte scar (AS) (arrow d). New
neurons generated by stem/ progenitor cell grafts can receive and send neural connects in PLP and beyond and may be able to form new, functional relay
circuits (B).

basic primitive function of preventing or limiting the spread of potential for identifying ways to manipulate these responses to
focal infection. Viewing certain cellular responses to other CNS improve outcome. Manipulations under intense investigation
insults in this light may provide insights regarding basic mecha- include not only traditional approaches such as development
nisms and how to understand and eventually manipulate re- of drug candidates for parenteral pharmacological delivery, but
sponses safely to improve outcomes. We of course recognize also lifestyle modifications such as reduced energy
that not all responses will be determined or influenced in this consumption and implementation of exercise strategies
manner. Nevertheless, the powerful and fundamental role of intended to reduce inflammatory responses, cellular
response to infection is likely to be ancestral to all responses degeneration, and functional decline after stroke and traumatic
to CNS damage and therefore has the potential to influence all injury (Arumugam et al., 2010; Mattson, 2012; Piao et al., 2013).
types of responses that evolved subsequently. This point is un- In addition, novel inter- ventional strategies in the form of
derscored by the growing recognition of the role of inflammatory biomaterial implants as molecular depots for local molecular
mechanisms in different neurodegenerative diseases, and the delivery, as well as tissue regenerative approaches based on
potential for molecules released by peripheral infections to transplantation of progenitor or stem cells, are showing
influ- ence far-distant reactive gliosis as discussed above. It is considerable promise for future appli- cation in neural repair
impor- tant to realize that the multicellular responses to diffuse and regeneration. The nonneural lesion cores of focal lesions
CNS insults are likely to have been shaped by adapting already may represent particularly useful targets for such approaches
exist- ing responses to focal traumatic damage or focal (Figure 6). For example, biomaterial im- plants, particularly in
infection. This notion is likely to apply not only to degenerative the form of injectable hydrogels, have po- tential to serve both
diseases, but also to autoimmune-mediated damage, where as scaffolds and depots that can influence tissue remodeling
the CNS response to such damage bears striking similarities to and cellular functions (Pakulska et al., 2012). Large lesion
responses to other forms of CNS tissue damage. This cores of nonneural tissue after stroke or severe focal trauma
discussion is meant to highlight the usefulness of constructing may represent useful sites for placement of biomaterial depots
models of how multiple cells interact and how multiple that release gradients of molecules to influence locally
molecules drive these interac- tions during the responses to surrounding neural tissue without inflicting additional damage
different types of CNS damage. The models we have (Figure 6A). For example, injectable hydrogel depots can
presented here are compatible with basic evidence currently provide sustained delivery of different kinds of bioactive
available but no doubt will require modifica- tion as information molecules, including growth factors or small hydrophobic
accrues. We hope that they are flexible enough to molecules to influ- ence gene expression and function of local
accommodate modifications and can serve as useful cells over prolonged subacute times, after which they are
frameworks. degraded without substan- tive tissue damage (Pakulska et al.,
2012; Song et al., 2012; Yang et al., 2009; Zhang et al., 2014).
Manipulating CNS Wound Responses to Promote Repair Such molecular delivery has the potential to influence not only
As the cell biology and molecular signaling that underlie re- the ongoing repair in the subacute period after injury such as
sponses to CNS damage become defined, there is increasing scar formation, but also can influence cells in the large
perimeter zone around CNS lesions that
undergoes continual tissue remodeling for prolonged periods ACKNOWLEDGMENTS
after tissue damage (Figure 6A). Delivery of molecules via
depots placed into lesion cores have the potential to influence The authors are supported by the NIH (NINDS), The Dr. Miriam and Sheldon
G. Adelson Medical Foundation, and Wings for Life.
the neural plasticity and tissue remodeling in this perimeter
region (Clark- son et al., 2010).
REFERENCES
Various cellular regenerative approaches may have potential
for CNS repair (Bliss et al., 2007; Martino and Pluchino, 2006; Abbott, N.J., Ro¨ nnba¨ ck, L., and Hansson, E. (2006). Astrocyte-endothelial
Snyder and Teng, 2012). Delivery of neural-lineage progenitor interactions at the blood-brain barrier. Nat. Rev. Neurosci. 7, 41–53.
or stem cells into lesion cores (Figure 6B) can lead to the
Abematsu, M., Tsujimura, K., Yamano, M., Saito, M., Kohno, K., Kohyama,
gener- ation of local neurons that form afferent and efferent J., Namihira, M., Komiya, S., and Nakashima, K. (2010). Neurons derived
connections with host and have the potential to form from transplanted neural stem cells restore disrupted neuronal circuitry in a
mouse model of spinal cord injury. J. Clin. Invest. 120, 3255–3266.
functionally integrated circuits (Abematsu et al., 2010; Lu et al.,
2012). Stromal or mesenchymal stem cells derived from bone Abeyama, K., Stern, D.M., Ito, Y., Kawahara, K., Yoshimoto, Y., Tanaka, M.,
marrow or umbilical cord, delivered by peripheral Uchimura, T., Ida, N., Yamazaki, Y., Yamada, S., et al. (2005). The N-
terminal domain of thrombomodulin sequesters high-mobility group-B1
(intravascular) or CNS injection, can home to injury sites protein, a novel antiinflammatory mechanism. J. Clin. Invest. 115, 1267–
(Figure 6C), where they can release growth factors, cytokines, 1274.
and other factors that may influence the response to damage Aldrich, A., and Kielian, T. (2011). Central nervous system fibrosis is
and repair process (Bliss et al., 2007; Kokaia et al., 2012; associated with fibrocyte-like infiltrates. Am. J. Pathol. 179, 2952–2962.
Lemmens and Steinberg, 2013). Such grafting studies are
Argaw, A.T., Gurfein, B.T., Zhang, Y., Zameer, A., and John, G.R. (2009).
being intensely pursued but are still in the early stages of VEGF-mediated disruption of endothelial CLN-5 promotes blood-brain barrier
development. Beneficial effects are reported on cellular events breakdown. Proc. Natl. Acad. Sci. USA 106, 1977–1982.
such as immunomodulation, survival of CNS intrinsic cells Argaw, A.T., Asp, L., Zhang, J., Navrazhina, K., Pham, T., Mariani, J.N., Ma-
including neurons, and growth of axons; however, limited hase, S., Dutta, D.J., Seto, J., Kramer, E.G., et al. (2012). Astrocyte-derived
information is as yet available on overall functional efficacy. VEGF-A drives blood-brain barrier disruption in CNS inflammatory disease.
J. Clin. Invest. 122, 2454–2468.

Concluding Remarks Arumugam, T.V., Phillips, T.M., Cheng, A., Morrell, C.H., Mattson, M.P., and
Wan, R. (2010). Age and energy intake interact to modify cell stress
The study of cellular responses to CNS injury has progressed pathways and stroke outcome. Ann. Neurol. 67, 41–52.
enormously in the last two decades, as have ideas about their
functions and mechanisms. It is now clear that reactive gliosis Askari, A.T., Unzek, S., Popovic, Z.B., Goldman, C.K., Forudi, F., Kiedrowski,
M., Rovner, A., Ellis, S.G., Thomas, J.D., DiCorleto, P.E., et al. (2003). Effect
and multicellular responses to CNS damage are a complex of stromal-cell-derived factor 1 on stem-cell homing and tissue regeneration
mixture of events that balance clearance of dead cells, patho- in ischaemic cardiomyopathy. Lancet 362, 697–703.
gens, and debris with maximal preservation of adjacent local Astrup, J., Siesjo¨ , B.K., and Symon, L. (1981). Thresholds in cerebral ischemia
tis- sue. These events are regulated by a vast number of - the ischemic penumbra. Stroke 12, 723–725.
molecular signals that control specific cellular functions as
Attwell, D., Buchan, A.M., Charpak, S., Lauritzen, M., Macvicar, B.A., and
dictated by spe- cific situations. Reactive gliosis is not all-or- Newman, E.A. (2010). Glial and neuronal control of brain blood flow. Nature
none or stereotypic; instead, it is highly variable and context 468, 232–243.
specific. Loss-of- function studies show that reactive gliosis and Bardehle, S., Kru¨ ger, M., Buggenthin, F., Schwausch, J., Ninkovic, J.,
multicellular re- sponses to CNS damage exert essential Clevers, H., Snippert, H.J., Theis, F.J., Meyer-Luehmann, M., Bechmann, I.,
beneficial functions without which tissue damage (and function) et al. (2013). Live imaging of astrocyte responses to acute injury reveals
selective juxtavascular proliferation. Nat. Neurosci. 16, 580–586.
would increase and tissue repair would not occur.
Nevertheless, over activity of certain functions may in some Bareyre, F.M., Kerschensteiner, M., Raineteau, O., Mettenleiter, T.C., Wein-
mann, O., and Schwab, M.E. (2004). The injured spinal cord spontaneously
cases exacerbate tissue dam- age. Interventions to block such forms a new intraspinal circuit in adult rats. Nat. Neurosci. 7, 269–277.
activities will need to target spe- cific molecular events rather
than attempt to block the process of gliosis per se. Barres, B.A. (2008). The mystery and magic of glia: a perspective on their
roles in health and disease. Neuron 60, 430–440.
Understanding the ways in which dysfunction of reactive glio-
sis and multicellular responses to CNS damage can occur, for Bell, R.D., Winkler, E.A., Sagare, A.P., Singh, I., LaRue, B., Deane, R., and
Zlo- kovic, B.V. (2010). Pericytes control key neurovascular functions and
example, through genetic mutations or polymorphisms, has neuronal phenotype in the adult brain and during brain aging. Neuron 68,
the potential to identify new mechanisms that may underlie 409–427.
specific CNS disorders or that may underlie population vari-
Bell, R.D., Winkler, E.A., Singh, I., Sagare, A.P., Deane, R., Wu, Z.,
ability in responses to similar insults such as stroke or trauma. Holtzman, D.M., Betsholtz, C., Armulik, A., Sallstrom, J., et al. (2012).
There is a compelling need to deepen our understanding of Apolipoprotein E controls cerebrovascular integrity via cyclophilin A. Nature
485, 512–516.
specific cellular interactions during responses to CNS insults,
and how these change over time and are governed by complex Benner, E.J., Luciano, D., Jo, R., Abdi, K., Paez-Gonzalez, P., Sheng, H.,
molecular signaling mechanisms. Such information is funda- Warner, D.S., Liu, C., Eroglu, C., and Kuo, C.T. (2013). Protective
astrogenesis from the SVZ niche after injury is controlled by Notch modulator
mental to understanding most CNS disorders and for devel- Thbs4. Nature 497, 369–373.
oping rationally targeted therapeutic strategies that can safely
Bianco, P., Riminucci, M., Gronthos, S., and Robey, P.G. (2001). Bone
manipulate or modify cellular responses to CNS insults in marrow stromal stem cells: nature, biology, and potential applications. Stem
ways that improve outcome. Cells 19, 180–192.

Bliss, T., Guzman, R., Daadi, M., and Steinberg, G.K. (2007). Cell
transplanta- tion therapy for stroke. Stroke Suppl. 38, 817–826.
Boille´ e, S., Vande Velde, C., and Cleveland, D.W. (2006). ALS: a disease of Failli, V., Kopp, M.A., Gericke, C., Martus, P., Klingbeil, S., Brommer, B.,
motor neurons and their nonneuronal neighbors. Neuron 52, 39–59. Lagi- nha, I., Chen, Y., DeVivo, M.J., Dirnagl, U., and Schwab, J.M. (2012).
Func- tional neurological recovery after spinal cord injury is impaired in
Brambilla, R., Bracchi-Ricard, V., Hu, W.H., Frydel, B., Bramwell, A., patients with infections. Brain 135, 3238–3250.
Karmally, S., Green, E.J., and Bethea, J.R. (2005). Inhibition of astroglial
nuclear factor kappaB reduces inflammation and improves functional Faulkner, J.R., Herrmann, J.E., Woo, M.J., Tansey, K.E., Doan, N.B., and
recovery after spinal cord injury. J. Exp. Med. 202, 145–156. Sofroniew, M.V. (2004). Reactive astrocytes protect tissue and preserve
func- tion after spinal cord injury. J. Neurosci. 24, 2143–2155.
Brenner, M., Johnson, A.B., Boespflug-Tanguy, O., Rodriguez, D., Goldman,
J.E., and Messing, A. (2001). Mutations in GFAP, encoding glial fibrillary Franklin, R.J., and Ffrench-Constant, C. (2008). Remyelination in the CNS:
acidic protein, are associated with Alexander disease. Nat. Genet. 27, 117– from biology to therapy. Nat. Rev. Neurosci. 9, 839–855.
120.
Frohman, E.M., Racke, M.K., and Raine, C.S. (2006). Multiple sclerosis—the
Buckingham, S.C., Campbell, S.L., Haas, B.R., Montana, V., Robel, S., plaque and its pathogenesis. N. Engl. J. Med. 354, 942–955.
Ogun- rinu, T., and Sontheimer, H. (2011). Glutamate release by primary
brain tumors induces epileptic activity. Nat. Med. 17, 1269–1274. Gadea, A., Schinelli, S., and Gallo, V. (2008). Endothelin-1 regulates
astrocyte proliferation and reactive gliosis via a JNK/c-Jun signaling
Burda, J.E., Radulovic, M., Yoon, H., and Scarisbrick, I.A. (2013). Critical role pathway. J. Neurosci. 28, 2394–2408.
for PAR1 in kallikrein 6-mediated oligodendrogliopathy. Glia 61, 1456–1470.
Gao, H.M., Zhou, H., Zhang, F., Wilson, B.C., Kam, W., and Hong, J.S.
Bush, T.G., Puvanachandra, N., Horner, C.H., Polito, A., Ostenfeld, T., (2011). HMGB1 acts on microglia Mac1 to mediate chronic
Svend- sen, C.N., Mucke, L., Johnson, M.H., and Sofroniew, M.V. (1999). neuroinflammation that drives progressive neurodegeneration. J. Neurosci.
Leukocyte infiltration, neuronal degeneration, and neurite outgrowth after 31, 1081–1092.
ablation of scar-forming, reactive astrocytes in adult transgenic mice. Neuron
23, 297–308. Garcia, A.D.R., Doan, N.B., Imura, T., Bush, T.G., and Sofroniew, M.V.
(2004). GFAP-expressing progenitors are the principal source of constitutive
Carmichael, S.T. (2006). Cellular and molecular mechanisms of neural repair neuro- genesis in adult mouse forebrain. Nat. Neurosci. 7, 1233–1241.
after stroke: making waves. Ann. Neurol. 59, 735–742.
Garcı´a-Alı´as, G., Barkhuysen, S., Buckle, M., and Fawcett, J.W. (2009).
Casella, G.T., Marcillo, A., Bunge, M.B., and Wood, P.M. (2002). New Chon- droitinase ABC treatment opens a window of opportunity for task-
vascular tissue rapidly replaces neural parenchyma and vessels destroyed specific rehabilitation. Nat. Neurosci. 12, 1145–1151.
by a contu- sion injury to the rat spinal cord. Exp. Neurol. 173, 63–76.
Gleichman, A.J., and Carmichael, S.T. (2013). Astrocytic therapies for
Chan, J.K., Roth, J., Oppenheim, J.J., Tracey, K.J., Vogl, T., Feldmann, M., neuronal repair in stroke. Neurosci. Lett. Published online October 31, 2013.
Horwood, N., and Nanchahal, J. (2012). Alarmins: awaiting a clinical http://dx. doi.org/10.1016/j.neulet.2013.10.055.
response. J. Clin. Invest. 122, 2711–2719.
Go¨ ritz, C., Dias, D.O., Tomilin, N., Barbacid, M., Shupliakov, O., and Frise´ n,
Clarkson, A.N., Huang, B.S., Macisaac, S.E., Mody, I., and Carmichael, S.T. J. (2011). A pericyte origin of spinal cord scar tissue. Science 333, 238–242.
(2010). Reducing excessive GABA-mediated tonic inhibition promotes func-
tional recovery after stroke. Nature 468, 305–309. Greaves, N.S., Ashcroft, K.J., Baguneid, M., and Bayat, A. (2013). Current
understanding of molecular and cellular mechanisms in fibroplasia and
Courtine, G., Song, B., Roy, R.R., Zhong, H., Herrmann, J.E., Ao, Y., Qi, J., angio- genesis during acute wound healing. J. Dermatol. Sci. 72, 206–217.
Edgerton, V.R., and Sofroniew, M.V. (2008). Recovery of supraspinal control
of stepping via indirect propriospinal relay connections after spinal cord Griffiths, M.R., Gasque, P., and Neal, J.W. (2010). The regulation of the CNS
injury. Nat. Med. 14, 69–74. innate immune response is vital for the restoration of tissue homeostasis
(repair) after acute brain injury: a brief review. Int. J. Inflamm. 2010, 151097.
Daneman, R., Zhou, L., Kebede, A.A., and Barres, B.A. (2010). Pericytes are
required for blood-brain barrier integrity during embryogenesis. Nature 468, Gurtner, G.C., Werner, S., Barrandon, Y., and Longaker, M.T. (2008). Wound
562–566. repair and regeneration. Nature 453, 314–321.

Halassa, M.M., and Haydon, P.G. (2010). Integrated brain circuits: astrocytic
Davalos, D., Grutzendler, J., Yang, G., Kim, J.V., Zuo, Y., Jung, S., Littman,
networks modulate neuronal activity and behavior. Annu. Rev. Physiol. 72,
D.R., Dustin, M.L., and Gan, W.B. (2005). ATP mediates rapid microglial
response to local brain injury in vivo. Nat. Neurosci. 8, 752–758. 335–355.

Hamby, M.E., Coppola, G., Ao, Y., Geschwind, D.H., Khakh, B.S., and Sofro-
Davis, G.E., and Senger, D.R. (2005). Endothelial extracellular matrix:
niew, M.V. (2012). Inflammatory mediators alter the astrocyte transcriptome
biosynthesis, remodeling, and functions during vascular morphogenesis and
and calcium signaling elicited by multiple G-protein-coupled receptors.
neovessel stabilization. Circ. Res. 97, 1093–1107.
J. Neurosci. 32, 14489–14510.
DeKosky, S.T., Blennow, K., Ikonomovic, M.D., and Gandy, S. (2013). Acute Hamilton, N.B., and Attwell, D. (2010). Do astrocytes really exocytose neuro-
and chronic traumatic encephalopathies: pathogenesis and biomarkers. Nat. transmitters? Nat. Rev. Neurosci. 11, 227–238.
Rev. Neurol. 9, 192–200.
Hanisch, U.K., and Kettenmann, H. (2007). Microglia: active sensor and
Derecki, N.C., Cronk, J.C., Lu, Z., Xu, E., Abbott, S.B., Guyenet, P.G., and versatile effector cells in the normal and pathologic brain. Nat. Neurosci. 10,
Kipnis, J. (2012). Wild-type microglia arrest pathology in a mouse model of 1387–1394.
Rett syndrome. Nature 484, 105–109.
Haroon, F., Dro¨ gemu¨ ller, K., Ha¨ ndel, U., Brunn, A., Reinhold, D., Nishanth,
Devinsky, O., Vezzani, A., Najjar, S., De Lanerolle, N.C., and Rogawski, M.A. G., Mueller, W., Trautwein, C., Ernst, M., Deckert, M., and Schlu¨ ter, D.
(2013). Glia and epilepsy: excitability and inflammation. Trends Neurosci. 36, (2011). Gp130-dependent astrocytic survival is critical for the control of
174–184. autoimmune central nervous system inflammation. J. Immunol. 186, 6521–
6531.
Dirnagl, U., Iadecola, C., and Moskowitz, M.A. (1999). Pathobiology of
ischae- mic stroke: an integrated view. Trends Neurosci. 22, 391–397. Hayakawa, K., Pham, L.D., Katusic, Z.S., Arai, K., and Lo, E.H. (2012).
Astro- cytic high-mobility group box 1 promotes endothelial progenitor cell-
Dro¨ gemu¨ ller, K., Helmuth, U., Brunn, A., Sakowicz-Burkiewicz, M., mediated neurovascular remodeling during stroke recovery. Proc. Natl. Acad.
Gutmann, D.H., Mueller, W., Deckert, M., and Schlu¨ ter, D. (2008). Astrocyte Sci. USA 109, 7505–7510.
gp130 expression is critical for the control of Toxoplasma encephalitis. J.
Immunol. 181, 2683–2693. Henneberger, C., Papouin, T., Oliet, S.H., and Rusakov, D.A. (2010). Long-
term potentiation depends on release of D-serine from astrocytes. Nature
Dugas, J.C., Mandemakers, W., Rogers, M., Ibrahim, A., Daneman, R., and 463, 232–236.
Barres, B.A. (2008). A novel purification method for CNS projection neurons
leads to the identification of brain vascular cells as a source of trophic Hermanns, S., Klapka, N., Gasis, M., and Mu¨ ller, H.W. (2006). The
support for corticospinal motor neurons. J. Neurosci. 28, 8294–8305. collagenous wound healing scar in the injured central nervous system
inhibits axonal regen- eration. Adv. Exp. Med. Biol. 557, 177–190.
Herrmann, J.E., Imura, T., Song, B., Qi, J., Ao, Y., Nguyen, T.K., Korsak, London, A., Cohen, M., and Schwartz, M. (2013). Microglia and monocyte-
R.A., Takeda, K., Akira, S., and Sofroniew, M.V. (2008). STAT3 is a critical derived macrophages: functionally distinct populations that act in concert in
regulator of astrogliosis and scar formation after spinal cord injury. J. CNS plasticity and repair. Front. Cell. Neurosci. 7, 34.
Neurosci. 28, 7231–7243.
Louis, D.N. (2006). Molecular pathology of malignant gliomas. Annu. Rev.
Hughes, E.G., Kang, S.H., Fukaya, M., and Bergles, D.E. (2013). Pathol. 1, 97–117.
Oligodendro- cyte progenitors balance growth with self-repulsion to achieve
homeostasis in the adult brain. Nat. Neurosci. 16, 668–676. Loy, D.N., Crawford, C.H., Darnall, J.B., Burke, D.A., Onifer, S.M., and
Whitte- more, S.R. (2002). Temporal progression of angiogenesis and basal
Jabs, R., Seifert, G., and Steinha¨ user, C. (2008). Astrocytic function and its lamina deposition after contusive spinal cord injury in the adult rat. J. Comp.
alteration in the epileptic brain. Epilepsia 49 (Suppl 2 ), 3–12. Neurol. 445, 308–324.

Jackman, N.A., Uliasz, T.F., Hewett, J.A., and Hewett, S.J. (2010). Lu, P., Wang, Y., Graham, L., McHale, K., Gao, M., Wu, D., Brock, J.,
Regulation of system x(c)(-)activity and expression in astrocytes by Blesch, A., Rosenzweig, E.S., Havton, L.A., et al. (2012). Long-distance
interleukin-1b: impli- cations for hypoxic neuronal injury. Glia 58, 1806–1815. growth and connectivity of neural stem cells after severe spinal cord injury.
Cell 150, 1264–1273.
Jaerve, A., Bosse, F., and Mu¨ ller, H.W. (2012). SDF-1/CXCL12: its role in
spinal cord injury. Int. J. Biochem. Cell Biol. 44, 452–456. Lucchinetti, C.F., Bru¨ ck, W., Rodriguez, M., and Lassmann, H. (1996).
Distinct patterns of multiple sclerosis pathology indicates heterogeneity on
Kawaja, M.D., and Gage, F.H. (1991). Reactive astrocytes are substrates for pathogen- esis. Brain Pathol. 6, 259–274.
the growth of adult CNS axons in the presence of elevated levels of nerve
growth factor. Neuron 7, 1019–1030. MacIver, N.J., Michalek, R.D., and Rathmell, J.C. (2013). Metabolic
regulation of T lymphocytes. Annu. Rev. Immunol. 31, 259–283.
Kimura-Kuroda, J., Teng, X., Komuta, Y., Yoshioka, N., Sango, K.,
Kawamura, K., Raisman, G., and Kawano, H. (2010). An in vitro model of the Maragakis, N.J., and Rothstein, J.D. (2006). Mechanisms of Disease: astro-
inhibition of axon growth in the lesion scar formed after central nervous cytes in neurodegenerative disease. Nat. Clin. Pract. Neurol. 2, 679–689.
system injury. Mol. Cell. Neurosci. 43, 177–187.
Martino, G., and Pluchino, S. (2006). The therapeutic potential of neural stem
Klapka, N., and Mu¨ ller, H.W. (2006). Collagen matrix in spinal cord injury.
cells. Nat. Rev. Neurosci. 7, 395–406.
J. Neurotrauma 23, 422–435.

Kokaia, Z., Martino, G., Schwartz, M., and Lindvall, O. (2012). Cross-talk Mattson, M.P. (2000). Apoptosis in neurodegenerative disorders. Nat. Rev.
between neural stem cells and immune cells: the key to better brain repair? Mol. Cell Biol. 1, 120–129.
Nat. Neurosci. 15, 1078–1087.
Mattson, M.P. (2012). Energy intake and exercise as determinants of brain
Kono, H., and Rock, K.L. (2008). How dying cells alert the immune system to health and vulnerability to injury and disease. Cell Metab. 16, 706–722.
danger. Nat. Rev. Immunol. 8, 279–289.
McIntosh, T.K., Vink, R., Noble, L., Yamakami, I., Fernyak, S., Soares, H.,
Kraft, A.W., Hu, X., Yoon, H., Yan, P., Xiao, Q., Wang, Y., Gil, S.C., Brown, and Faden, A.L. (1989). Traumatic brain injury in the rat: characterization of a
J., Wilhelmsson, U., Restivo, J.L., et al. (2013). Attenuating astrocyte lateral fluid-percussion model. Neuroscience 28, 233–244.
activation accelerates plaque pathogenesis in APP/PS1 mice. FASEB J. 27,
187–198. Meletis, K., Barnabe´ -Heider, F., Carle´ n, M., Evergren, E., Tomilin, N.,
Shuplia- kov, O., and Frise´ n, J. (2008). Spinal cord injury reveals
Kreutzberg, G.W. (1996). Microglia: a sensor for pathological events in the multilineage differen- tiation of ependymal cells. PLoS Biol. 6, e182.
CNS. Trends Neurosci. 19, 312–318.
Midwood, K.S., Williams, L.V., and Schwarzbauer, J.E. (2004). Tissue repair
Kriegstein, A., and Alvarez-Buylla, A. (2009). The glial nature of embryonic and the dynamics of the extracellular matrix. Int. J. Biochem. Cell Biol. 36,
and adult neural stem cells. Annu. Rev. Neurosci. 32, 149–184. 1031–1037.

Lagace, D.C. (2012). Does the endogenous neurogenic response alter Mills, K.H. (2011). TLR-dependent T cell activation in autoimmunity. Nat.
behav- ioral recovery following stroke? Behav. Brain Res. 227, 426–432. Rev. Immunol. 11, 807–822.

Lemmens, R., and Steinberg, G.K. (2013). Stem cell therapy for acute Mosser, D.M., and Edwards, J.P. (2008). Exploring the full spectrum of
cerebral injury: what do we know and what will the future bring? Curr. Opin. macro- phage activation. Nat. Rev. Immunol. 8, 958–969.
Neurol. 26, 617–625.
Myer, D.J., Gurkoff, G.G., Lee, S.M., Hovda, D.A., and Sofroniew, M.V.
Lennon, V.A., Kryzer, T.J., Pittock, S.J., Verkman, A.S., and Hinson, S.R. (2006). Essential protective roles of reactive astrocytes in traumatic brain
(2005). IgG marker of optic-spinal multiple sclerosis binds to the aquaporin- injury. Brain 129, 2761–2772.
4 water channel. J. Exp. Med. 202, 473–477.
Neary, J.T., Kang, Y., Willoughby, K.A., and Ellis, E.F. (2003). Activation of
Li, L., Lundkvist, A., Andersson, D., Wilhelmsson, U., Nagai, N., Pardo, A.C., extracellular signal-regulated kinase by stretch-induced injury in astrocytes
Nodin, C., Sta˚ hlberg, A., Aprico, K., Larsson, K., et al. (2008). Protective role involves extracellular ATP and P2 purinergic receptors. J. Neurosci. 23,
of reactive astrocytes in brain ischemia. J. Cereb. Blood Flow Metab. 28, 2348–2356.
468–481.
Nimmerjahn, A., Kirchhoff, F., and Helmchen, F. (2005). Resting microglial
Li, S., Overman, J.J., Katsman, D., Kozlov, S.V., Donnelly, C.J., Twiss, J.L., cells are highly dynamic surveillants of brain parenchyma in vivo. Science
Giger, R.J., Coppola, G., Geschwind, D.H., and Carmichael, S.T. (2010). An 308, 1314–1318.
age-related sprouting transcriptome provides molecular control of axonal
sprouting after stroke. Nat. Neurosci. 13, 1496–1504. Nishiyama, A., Komitova, M., Suzuki, R., and Zhu, X. (2009).
Polydendrocytes (NG2 cells): multifunctional cells with lineage plasticity. Nat.
Lindvall, O., and Kokaia, Z. (2006). Stem cells for the treatment of Rev. Neurosci. 10, 9–22.
neurological disorders. Nature 441, 1094–1096.
Ohab, J.J., and Carmichael, S.T. (2008). Poststroke neurogenesis: emerging
Liu, G.J., Nagarajah, R., Banati, R.B., and Bennett, M.R. (2009). Glutamate principles of migration and localization of immature neurons. Neuroscientist
induces directed chemotaxis of microglia. Eur. J. Neurosci. 29, 1108–1118. 14, 369–380.

Lo, E.H. (2008). A new penumbra: transitioning from injury into repair after Okada, S., Nakamura, M., Katoh, H., Miyao, T., Shimazaki, T., Ishii, K.,
stroke. Nat. Med. 14, 497–500. Yamane, J., Yoshimura, A., Iwamoto, Y., Toyama, Y., and Okano, H. (2006).
Conditional ablation of Stat3 or Socs3 discloses a dual role for reactive astro-
Logan, A., and Berry, M. (2002). Cellular and molecular determinants of glial cytes after spinal cord injury. Nat. Med. 12, 829–834.
scar formation. Adv. Exp. Med. Biol. 513, 115–158.
Ortinski, P.I., Dong, J., Mungenast, A., Yue, C., Takano, H., Watson, D.J., Silver, D.J., Siebzehnrubl, F.A., Schildts, M.J., Yachnis, A.T., Smith, G.M.,
Hay- don, P.G., and Coulter, D.A. (2010). Selective induction of astrocytic Smith, A.A., Scheffler, B., Reynolds, B.A., Silver, J., and Steindler, D.A.
gliosis generates deficits in neuronal inhibition. Nat. Neurosci. 13, 584–591. (2013). Chondroitin sulfate proteoglycans potently inhibit invasion and serve
as a central organizer of the brain tumor microenvironment. J. Neurosci. 33,
Oudega, M. (2012). Molecular and cellular mechanisms underlying the role of 15603–15617.
blood vessels in spinal cord injury and repair. Cell Tissue Res. 349, 269–
288. Singer, A.J., and Clark, R.A. (1999). Cutaneous wound healing. N. Engl. J. Med.
341, 738–746.
Overman, J.J., Clarkson, A.N., Wanner, I.B., Overman, W.T., Eckstein, I.,
Maguire, J.L., Dinov, I.D., Toga, A.W., and Carmichael, S.T. (2012). A role Sirko, S., Behrendt, G., Johansson, P.A., Tripathi, P., Costa, M., Bek, S.,
for ephrin-A5 in axonal sprouting, recovery, and activity-dependent plasticity Hein- rich, C., Tiedt, S., Colak, D., Dichgans, M., et al. (2013). Reactive glia
after stroke. Proc. Natl. Acad. Sci. USA 109, E2230–E2239. in the injured brain acquire stem cell properties in response to sonic
hedgehog. [corrected]. Cell Stem Cell 12, 426–439.
Pakulska, M.M., Ballios, B.G., and Shoichet, M.S. (2012). Injectable
hydrogels for central nervous system therapy. Biomed. Mater. 7, 024101. Snyder, E.Y., and Teng, Y.D. (2012). Stem cells and spinal cord repair. N.
Engl. J. Med. 366, 1940–1942.
Perry, V.H. (2010). Contribution of systemic inflammation to chronic neurode-
generation. Acta Neuropathol. 120, 277–286. Soderblom, C., Luo, X., Blumenthal, E., Bray, E., Lyapichev, K., Ramos, J.,
Krishnan, V., Lai-Hsu, C., Park, K.K., Tsoulfas, P., and Lee, J.K. (2013). Peri-
Perry, V.H., Cunningham, C., and Holmes, C. (2007). Systemic infections vascular fibroblasts form the fibrotic scar after contusive spinal cord injury.
and inflammation affect chronic neurodegeneration. Nat. Rev. Immunol. 7, J. Neurosci. 33, 13882–13887.
161–167.
Sofroniew, M.V. (2005). Reactive astrocytes in neural repair and protection.
Piao, C.S., Stoica, B.A., Wu, J., Sabirzhanov, B., Zhao, Z., Cabatbat, R., Neuroscientist 11, 400–407.
Loane, D.J., and Faden, A.I. (2013). Late exercise reduces
neuroinflammation and cognitive dysfunction after traumatic brain injury. Sofroniew, M.V. (2009). Molecular dissection of reactive astrogliosis and glial
Neurobiol. Dis. 54, 252–263. scar formation. Trends Neurosci. 32, 638–647.

Popescu, B.F., and Lucchinetti, C.F. (2012). Pathology of demyelinating Sofroniew, M.V. (2013). Multiple roles for astrocytes as effectors of cytokines
diseases. Annu. Rev. Pathol. 7, 185–217. and inflammatory mediators. Neuroscientist. Published online October 8,
2013. http://dx.doi.org/10.1177/1073858413504466.
Popovich, P.G., and Longbrake, E.E. (2008). Can the immune system be
harnessed to repair the CNS? Nat. Rev. Neurosci. 9, 481–493. Sofroniew, M.V., and Vinters, H.V. (2010). Astrocytes: biology and pathology.
Acta Neuropathol. 119, 7–35.
Prokop, S., Miller, K.R., and Heppner, F.L. (2013). Microglia actions in
Alzheimer’s disease. Acta Neuropathol. 126, 461–477. Song, B., Song, J., Zhang, S., Anderson, M.A., Ao, Y., Yang, C.Y., Deming,
T.J., and Sofroniew, M.V. (2012). Sustained local delivery of bioactive nerve
Radulovic, M., Yoon, H., Larson, N., Wu, J., Linbo, R., Burda, J.E., growth factor in the central nervous system via tunable diblock copolypeptide
Diamandis, E.P., Blaber, S.I., Blaber, M., Fehlings, M.G., and Scarisbrick, hydrogel depots. Biomaterials 33, 9105–9116.
I.A. (2013). Kallikrein cascades in traumatic spinal cord injury: in vitro
evidence for roles in axonopathy and neuron degeneration. J. Neuropathol. Srivastava, R., Aslam, M., Kalluri, S.R., Schirmer, L., Buck, D., Tackenberg,
Exp. Neurol. 72, 1072–1089. B., Rothhammer, V., Chan, A., Gold, R., Berthele, A., et al. (2012).
Potassium channel KIR4.1 as an immune target in multiple sclerosis. N.
Ransohoff, R.M., and Perry, V.H. (2009). Microglial physiology: unique Engl. J. Med. 367, 115–123.
stimuli, specialized responses. Annu. Rev. Immunol. 27, 119–145.
Stephan, A.H., Barres, B.A., and Stevens, B. (2012). The complement
Reilkoff, R.A., Bucala, R., and Herzog, E.L. (2011). Fibrocytes: emerging system: an unexpected role in synaptic pruning during development and
effector cells in chronic inflammation. Nat. Rev. Immunol. 11, 427–435. disease. Annu. Rev. Neurosci. 35, 369–389.

Rothstein, J.D. (2009). Current hypotheses for the underlying biology of Stevens, B., Allen, N.J., Vazquez, L.E., Howell, G.R., Christopherson, K.S.,
amyo- trophic lateral sclerosis. Ann. Neurol. 65 (Suppl 1 ), S3–S9. Nouri, N., Micheva, K.D., Mehalow, A.K., Huberman, A.D., Stafford, B., et al.
(2007). The classical complement cascade mediates CNS synapse elimina-
Sahni, V., Mukhopadhyay, A., Tysseling, V., Hebert, A., Birch, D., Mcguire, tion. Cell 131, 1164–1178.
T.L., Stupp, S.I., and Kessler, J.A. (2010). BMPR1a and BMPR1b signaling
exert opposing effects on gliosis after spinal cord injury. J. Neurosci. 30, Sun, F., Lin, C.L., McTigue, D., Shan, X., Tovar, C.A., Bresnahan, J.C., and
1839–1855. Beattie, M.S. (2010). Effects of axon degeneration on oligodendrocyte
lineage cells: dorsal rhizotomy evokes a repair response while axon
Seo, J.H., Miyamoto, N., Hayakawa, K., Pham, L.D., Maki, T., Ayata, C., Kim, degeneration rostral to spinal contusion induces both repair and apoptosis.
K.W., Lo, E.H., and Arai, K. (2013). Oligodendrocyte precursors induce early Glia 58, 1304– 1319.
blood-brain barrier opening after white matter injury. J. Clin. Invest. 123,
782–786. Suo, Z., Wu, M., Ameenuddin, S., Anderson, H.E., Zoloty, J.E., Citron, B.A.,
Andrade-Gordon, P., and Festoff, B.W. (2002). Participation of protease-acti-
Shechter, R., and Schwartz, M. (2013). CNS sterile injury: just another vated receptor-1 in thrombin-induced microglial activation. J. Neurochem. 80,
wound healing? Trends Mol. Med. 19, 135–143. 655–666.

Shigetomi, E., Bowser, D.N., Sofroniew, M.V., and Khakh, B.S. (2008). Two Tao, J., Wu, H., Lin, Q., Wei, W., Lu, X.H., Cantle, J.P., Ao, Y., Olsen, R.W.,
forms of astrocyte calcium excitability have distinct effects on NMDA recep- Yang, X.W., Mody, I., et al. (2011). Deletion of astroglial Dicer causes non-
tor-mediated slow inward currents in pyramidal neurons. J. Neurosci. 28, cell-autonomous neuronal dysfunction and degeneration. J. Neurosci. 31,
6659–6663. 8306–8319.

Shirakawa, H., Sakimoto, S., Nakao, K., Sugishita, A., Konno, M., Iida, S., Tong, X., Shigetomi, E., Looger, L.L., and Khakh, B.S. (2013). Genetically
Kusano, A., Hashimoto, E., Nakagawa, T., and Kaneko, S. (2010). Transient encoded calcium indicators and astrocyte calcium microdomains. Neurosci-
receptor potential canonical 3 (TRPC3) mediates thrombin-induced astrocyte entist 19, 274–291.
activation and upregulates its own expression in cortical astrocytes.
J. Neurosci. 30, 13116–13129. Tuszynski, M.H., and Steward, O. (2012). Concepts and methods for the study
of axonal regeneration in the CNS. Neuron 74, 777–791.
Silver, J., and Miller, J.H. (2004). Regeneration beyond the glial scar. Nat.
Rev. Neurosci. 5, 146–156. van den Brand, R., Heutschi, J., Barraud, Q., DiGiovanna, J., Bartholdi, K.,
Huerlimann, M., Friedli, L., Vollenweider, I., Moraud, E.M., Duis, S., et al.
Silver, J., Edwards, M.A., and Levitt, P. (1993). Immunocytochemical demon- (2012). Restoring voluntary control of locomotion after paralyzing spinal cord
stration of early appearing astroglial structures that form boundaries and injury. Science 336, 1182–1185.
path- ways along axon tracts in the fetal brain. J. Comp. Neurol. 328, 415–
436.
Varki, A., and Gagneux, P. (2012). Multifarious roles of sialic acids in Yamanaka, K., Chun, S.J., Boillee, S., Fujimori-Tonou, N., Yamashita, H.,
immunity. Ann. N Y Acad. Sci. 1253, 16–36. Gut- mann, D.H., Takahashi, R., Misawa, H., and Cleveland, D.W. (2008).
Astro- cytes as determinants of disease progression in inherited amyotrophic
Verkhratsky, A., Orkand, R.K., and Kettenmann, H. (1998). Glial calcium: lateral sclerosis. Nat. Neurosci. 11, 251–253.
homeostasis and signaling function. Physiol. Rev. 78, 99–141.
Yang, C.Y., Song, B., Ao, Y., Nowak, A.P., Abelowitz, R.B., Korsak, R.A.,
Volterra, A., and Meldolesi, J. (2005). Astrocytes, from brain glue to
Havton, L.A., Deming, T.J., and Sofroniew, M.V. (2009). Biocompatibility of
communi- cation elements: the revolution continues. Nat. Rev. Neurosci. 6, amphiphilic diblock copolypeptide hydrogels in the central nervous system.
626–640. Biomaterials 30, 2881–2898.
Voskuhl, R.R., Peterson, R.S., Song, B., Ao, Y., Morales, L.B., Tiwari-Wood-
ruff, S., and Sofroniew, M.V. (2009). Reactive astrocytes form scar-like peri- Yoshioka, N., Hisanaga, S., and Kawano, H. (2010). Suppression of fibrotic
vascular barriers to leukocytes during adaptive immune inflammation of the scar formation promotes axonal regeneration without disturbing blood-brain
CNS. J. Neurosci. 29, 11511–11522. barrier repair and withdrawal of leukocytes after traumatic brain injury. J.
Comp. Neurol. 518, 3867–3881.
Walsh, J.T., and Kipnis, J. (2011). Regulatory T cells in CNS injury: the
simple, the complex and the confused. Trends Mol. Med. 17, 541–547. Zamanian, J.L., Xu, L., Foo, L.C., Nouri, N., Zhou, L., Giffard, R.G., and
Barres,
Wang, D., and Fawcett, J. (2012). The perineuronal net and the control of B.A. (2012). Genomic analysis of reactive astrogliosis. J. Neurosci. 32, 6391–
CNS plasticity. Cell Tissue Res. 349, 147–160. 6410.
Wanner, I.B., Anderson, M.A., Song, B., Levine, J., Fernandez, A., Gray-
Thompson, Z., Ao, Y., and Sofroniew, M.V. (2013). Glial scar borders are Zhang, S., Anderson, M.A., Ao, Y., Khakh, B.S., Fan, J., Deming, T.J., and
formed by newly proliferated, elongated astrocytes that interact to corral Sofroniew, M.V. (2014). Tunable diblock copolypeptide hydrogel depots for
inflammatory and fibrotic cells via STAT3-dependent mechanisms after spinal local delivery of hydrophobic molecules in healthy and injured central
cord injury. J. Neurosci. 33, 12870–12886. nervous system. Biomaterials 35, 1989–2000.

Watkins, S., and Sontheimer, H. (2012). Unique biology of gliomas: Zheng, W., Watts, L.T., Holstein, D.M., Prajapati, S.I., Keller, C., Grass, E.H.,
challenges and opportunities. Trends Neurosci. 35, 546–556. Walter, C.A., and Lechleiter, J.D. (2010). Purinergic receptor stimulation
reduces cytotoxic edema and brain infarcts in mouse induced by photothrom-
Wetherington, J., Serrano, G., and Dingledine, R. (2008). Astrocytes in the bosis by energizing glial mitochondria. PLoS ONE 5, e14401.
epileptic brain. Neuron 58, 168–178.

Williams, R.R., Henao, M., Pearse, D.D., and Bunge, M.B. (2013). Zhong, Z., Deane, R., Ali, Z., Parisi, M., Shapovalov, Y., O’Banion, M.K., Sto-
Permissive Schwann Cell Graft/Spinal Cord Interfaces for Axon janovic, K., Sagare, A., Boillee, S., Cleveland, D.W., and Zlokovic, B.V.
Regeneration. Cell Trans- plant. Published online October 22, 2013. (2008). ALS-causing SOD1 mutants generate vascular changes prior to
http://dx.doi.org/10.3727/ 096368913X674657. motor neuron degeneration. Nat. Neurosci. 11, 420–422.

Winkler, E.A., Bell, R.D., and Zlokovic, B.V. (2011). Central nervous system Zlokovic, B.V. (2008). The blood-brain barrier in health and chronic neurode-
pericytes in health and disease. Nat. Neurosci. 14, 1398–1405. generative disorders. Neuron 57, 178–201.
Winkler, E.A., Sengillo, J.D., Sullivan, J.S., Henkel, J.S., Appel, S.H., and
Zlokovic, B.V. (2011). Neurovascular pathways to neurodegeneration in
Zlokovic, B.V. (2013). Blood-spinal cord barrier breakdown and pericyte
Alzheimer’s disease and other disorders. Nat. Rev. Neurosci. 12, 723–738.
reductions in amyotrophic lateral sclerosis. Acta Neuropathol. 125, 111–120.

Wyss-Coray, T., Loike, J.D., Brionne, T.C., Lu, E., Anankov, R., Yan, F., Zukor, K., Belin, S., Wang, C., Keelan, N., Wang, X., and He, Z. (2013).
Silver- stein, S.C., and Husemann, J. (2003). Adult mouse astrocytes Short hairpin RNA against PTEN enhances regenerative growth of
degrade amy- loid-beta in vitro and in situ. Nat. Med. 9, 453–457. corticospinal tract axons after spinal cord injury. J. Neurosci. 33, 15350–
15361.

You might also like