Você está na página 1de 16

Journal of Cerebral Blood Flow & Metabolism (2011) 31, 18361851

& 2011 ISCBFM All rights reserved 0271-678X/11 $32.00


www.jcbfm.com

Review Article

Heterogeneity in the penumbra


Gregory J del Zoppo1,2*, Frank R Sharp3,4, Wolf-Dieter Heiss5 and Gregory W Albers6
1

Department of Medicine (Division of Hematology), University of Washington School of Medicine, Seattle,


Washington, USA; 2Department of Neurology, University of Washington School of Medicine, Seattle,
Washington, USA; 3Department of Neurology, MIND Institute, University of California at Davis Medical
Center, Sacramento, California, USA; 4Department of Genetics, MIND Institute, University of California
at Davis Medical Center, Sacramento, California, USA; 5Max Planck Institute for Neurological Research,
Cologne, Germany; 6Department of Neurology and Neurological Sciences, Stanford University Medical
Center, Palo Alto, California, USA

Original experimental studies in nonhuman primate models of focal ischemia showed flow-related
changes in evoked potentials that suggested a circumferential zone of low regional cerebral blood
flow with normal K + homeostasis, around a core of permanent injury in the striatum or the cortex.
This became the basis for the definition of the ischemic penumbra. Imaging techniques of the time
suggested a homogeneous core of injury, while positing a surrounding penumbral region that
could be salvaged. However, both molecular studies and observations of vascular integrity indicate
a more complex and dynamic situation in the ischemic core that also changes with time. The
microvascular, cellular, and molecular events in the acute setting are compatible with heterogeneity
of the injury within the injury center, which at early time points can be described as multiple minicores associated with multiple mini-penumbras. These observations suggest the progression of
injury from many small foci to a homogeneous defect over time after the onset of ischemia. Recent
observations with updated imaging techniques and data processing support these dynamic
changes within the core and the penumbra in humans following focal ischemia.
Journal of Cerebral Blood Flow & Metabolism (2011) 31, 18361851; doi:10.1038/jcbfm.2011.93; published online 6 July 2011
Keywords: focal ischemia; imaging; ischemic penumbra; metabolic characteristics; microvessel characteristics;

molecular characteristics

Introduction
Occlusion of a brain-supplying artery for an extended period of ischemia leads to permanent injury if
return of flow is inadequate. Astrup et al (1977, 1981)
posited the development of a core of injury destined
for tissue destruction (infarction), surrounded by a
penumbra of metabolically metastable tissue that
has the potential for full recovery (Figure 1A)
(Branston et al, 1974, 1977). This general depiction
*
Correspondence: Dr GJ del Zoppo, MD, Department of Medicine (in
Hematology), Department of Neurology, University of Washington
School of Medicine, Box 359756 at Harborview Medical Center, 325
Ninth Avenue, Seattle, WA 98104, USA.
E-mail: grgdlzop@u.washington.edu
The work reported here has been supported in part by NIH grants
NS 053716, NS 038710, and NS 036945 to Dr del Zoppo, NIH
grants NS066845 and NS054652 to Dr Sharp, and NIH grants
NS39325 and NS044848 to Dr Albers, and funding by the WDH
Foundation to Dr Heiss.
Received 7 January 2011; revised 8 May 2011; accepted 9 May
2011; published online 6 July 2011

of the penumbra has greatly influenced the concept


of ischemic stroke following thrombotic and thromboembolic occlusion of brain-supplying arteries, and
our attempts to enhance tissue recovery clinically.
However, evolving data regarding cellular and
molecular responses to ischemia, the interrelationships of vascular and neuronal activation (the
neurovascular unit), experience with models of focal
ischemia, and progress in acute imaging techniques
in both animals and patients have suggested important refinements to the penumbra concept.
Although the original conceptualization of the
penumbra remains solid, newer data that take into
account the time course of injury development,
potential interactions of boundary zones to the core,
and the vascular territory at risk within the stricken
hemisphere may provide even greater relevance
to patient outcome than indicated by many acute
intervention clinical trials. Recent work is beginning
to address these issues and could point the way to
further constructive developments in brain injury
management.

Heterogeneity in the penumbra


GJ del Zoppo et al
1837

Figure 1 The penumbra. (A) Depiction of the penumbra based


on rCBF and electrophysiological studies on the nonhuman
primate (from Symon (1980), with permission). (B) Depiction of
the relationship among rCBF, electrical failure in the dependent
cerebral tissue, and ischemia (taken from a figure in the study by
Astrup et al (1977), with permission). rCBF, regional cerebral
blood flow.

Evolving Definitions of the Penumbra


Early approaches to modeling focal ischemia in
mammals sought to develop discrete regions of neuron
injury by manipulating the cerebral vascular supply.
As model development moved toward the more
clinically relevant occlusion of the middle cerebral
artery (MCA) in the nonhuman primate, descriptions of
neuron function and integrity were linked to regional
changes in regional cerebral blood flow (rCBF). By
necessity, the concept of the penumbra has depended
in a large part on the techniques used to visualize it.
From the notion of a circumferential region of
metabolic disturbance that has not reached the
threshold of permanent injury, the concept has
evolved to include (1) characteristic electrophysiological changes, (2) biochemical/molecular alterations, (3) responses of the microvasculature, (4)
metabolic changes defined by imaging methods,

and (5) observed differences between regions of


abnormal tissue perfusion and diffusion regions on
magnetic resonance imaging (MRI) studies.
In the original electrophysiological concept, the
ischemic penumbra was an area that became
electrically silent, but could recover function if the
rCBF was restored in time (Heiss, 1992; Hossmann,
1994). An operational definition states that the
penumbra is ischemic tissue which is functionally
impaired and is at risk for infarction, but has the
potential to be salvagedy. If not salvaged this tissue
is progressively recruited into the infarct core, which
will expand with time into the maximal volume
originally at risk (Baron, 1999). Imaging of the
penumbra, using positron emission tomography
(PET) has allowed measurement of rCBF, cerebral
metabolic rate for oxygen (CMRO2), rCMRglc (regional
cerebral metabolic rate of glucose), and oxygen
extraction fraction (OEF). Infarction usually corresponds to rCBF decreased below 12 mL/100 g per min
and rCMRO2 below 65 mm/100 g per min, whereas the
penumbra has been defined as rCBF decreased to
12 to 22 mL/100 mg per min, the rCMRO2 above
65 mm/100 g per min, and OEF increased to 50% to
90% (Heiss, 2000). The concept of a mismatch
between areas of blood perfusion and H2O diffusion
in the injured territory as revealed by high fieldstrength magnetic MRI has provided a possible way
to image the penumbra in humans.
Using animal models and methods with increased
resolution, alterations in protein synthesis occur
within and around the regions of injury. In regions
of decreased rCBF, molecular correlates of the
penumbra are: (1) decreased protein synthesis that
can recover, (2) preserved ATP, (3) synthesis of heatshock proteins (Hsps), and possibly (4) a successful
unfolded protein response (Sharp et al, 2000). If
these molecular criteria are met, the tissue has the
capacity to be salvaged if blood flow is restored.
Once considered to be inert conduits, the microvasculature has been shown to respond rapidly and
dynamically within the territory at risk, containing
the ischemic core and regions peripheral to the core.
Microvessel adhesion receptor and basal lamina
matrix expression can present as mini-cores and
mini-penumbras within the early evolving core of
neuron injury, after occlusion of a brain-supplying
artery, in a characteristic time-dependent manner
(Tagaya et al, 2001). Given that the microvasculature,
which in capillaries consists of the endothelium
basal lamina matrixastrocyte end-feet complex,
serves neurons (as part of a hypothetical neurovascular unit), changes in microvessels and neurons
must to be connected locally.
Each of these definitions describes the events/
conditions in regions of focal ischemic injury within
the territory at risk. Not considered by these
definitions is whether the processes that determine
the penumbra or core occur uniformly in adjacent
areas over time and whether the active conversion of
the penumbra to core occurs at the interface of the
Journal of Cerebral Blood Flow & Metabolism (2011) 31, 18361851

Heterogeneity in the penumbra


GJ del Zoppo et al
1838

core. Recent data have suggested that the core of


ischemic injury develops heterogeneously, and with
time coalesces dynamically into a homogenous core.
These data are consistent with the original concepts
of the penumbra and core, but recognize the dynamic
complex heterogeneous processes involved. This
refinement has potential therapeutic implications.

The Ischemic Penumbra and the


Ischemic Core
The ischemic penumbra was initially defined by
physiologic experiments performed in large animals
subjected to focal cerebral ischemia as (1) a region
around a central core of low rCBF and physiologic
silence, (2) a boundary region with release of
intracellular K + , and (3) a region of metabolic
instability that could be recovered.
Symon et al (1980) described the penumbra as a
tissue of evolving injury around a homogeneous
central core destined for infarction. Symon et al
(1974) showed that 3 years after the rapid reduction
in rCBF in the MCA territory in the nonhuman
primate (Papio sp), a residual core of low rCBF
remained that was congruent with the acute CBF
reduction. Regions of CBF peripheral to the core
had returned to normal levels. Branston et al (1974),
using the same nonhuman primate model, related
the loss in evoked potential (EP) amplitude to
significant reductions in local CBF. Here, the rCBF
threshold for 50% reduction in EP amplitude was
B18 mL/100 g per min, when regional flow was
reduced by phlebotomy and induced hypotension.
The functional threshold for neurologic dysfunction progressed from mild paresis at 22 mL/100 g per
min to complete paralysis at 8 mL/100 g per min.
Hence, the presence of a boundary zone in which the
EP amplitude was sensitive to blood pressure/flow
could be defined. Reduction in EP amplitude
correlated with cellular K + release into the surrounding tissue. When measured with H2 clearance
electrodes, rCBF reduction could be observed at
which the EP amplitude remained at 50% normal
and K + extravasation occurred in the core, but not
in the peripheral zones (Symon et al, 1974; Branston
et al, 1977). Further studies have shown a differential
sensitivity of K + and Ca2 + extravasation when rCBF
ranged from 8 to 15 mL/100 g per min (Harris and
Symon, 1984); EP amplitude decreased when rCBF
dropped below 15 mL/100 g per min even when
K + extravasation was not observed (Branston et al,
1977). On the basis of model studies, flow rates of
12 mL/100 g per min lasting for 2 to 3 hours led to
large infarctions, but individual cells became necrotic after shorter intervals of time and at higher levels
of residual flow. The potential for irreversible
damage or recovery then seemed determined not
only by the level of residual flow but also by the
duration of flow disturbance for a given region.
Journal of Cerebral Blood Flow & Metabolism (2011) 31, 18361851

Figure 2 Response of tissue to the duration of rCBF reduction,


and development of the penumbra. Diagram of CBF thresholds
required for the preservation of function and morphology of brain
tissue. The activity of individual neurons is blocked when flow
decreases below a certain threshold (dashed line) and returns
when flow is increased again above this threshold. The upper
recordings are from a single neuron before, during, and after
reversible MCA occlusion. The fate of a single cell depends on
the duration for which CBF is impaired below a certain level. The
curved solid line demarcates conditions for structurally damaged
from a functionally impaired, but morphologically intact tissue,
the penumbra. The dashed line distinguishes tissue not at risk
from the functionally impaired tissue. MCA, middle cerebral
artery; rCBF, regional cerebral blood flow.

These observations form the basis for the familiar


temporal relationship among rCBF, electrical function,
and brain tissue integrity characterizing regions of
permanent injury and of reversible injury from
ischemia onset (Figure 1B) (Jones et al, 1981). This
synthesis implies that the core is homogeneous, and
expands as a wave front with time into a penumbra of
tissue that is at least initially not doomed to infarction.
The range of perfusion between those limitsa rCBF
level below which neuronal function is impaired and a
lower threshold below which irreversible membrane
failure and morphologic damage occurtypifies the
ischemic penumbra (Astrup et al, 1981).
Separately, experiments using multiple electrodes in
cats subjected to focal cerebral ischemia, which simultaneously assessed single-cell activity and local flow,
showed pockets of low flow and cortical neuron
vulnerability in closely adjacent cortical areas (Rosner
et al, 1986). The altered single-cell activity with grouped
or regular discharges at flow levels just above the
threshold correlated with the gradual appearance of
functional deficits (Figure 2). Spontaneous neuronal
activity and EPs were restored when blood flow was
reestablished (Heiss et al, 1976; Heiss and Rosner, 1983).

Metabolic Characteristics of the


Penumbra and Core
Energy demands of the central nervous system are
high and almost entirely provided by oxidative

Heterogeneity in the penumbra


GJ del Zoppo et al
1839

glucose metabolism. Increases in glucose consumption (and rCBF) evoked by functional activation are
most prominent in synapse-rich regions, i.e., which
contain axonal terminals, dendritic processes, and
astrocytic processes that envelope the synapses and
microvasculature. Energy requirements for functional activation are mostly caused by stimulation
of Na + K + -ATPase activity to restore the ionic
gradients across neuron cell membranes and membrane potentials that are altered by the spike activity
(Sokoloff, 1999).
From its conceptual foundation in experiments in
the nonhuman primate, further work on metabolic
aberrations in the at-risk tissue proceeded in
smaller animal models of focal cerebral ischemia.
Loss of bioluminescent ATP and tissue acidosis
occur in the ischemic core and penumbra (Astrup
et al, 1977; Hossman and Mies, 2007) (AJ Strong,
personal communication). The relationships among
ischemic perfusion, functional impairment, biochemical disturbances, tissue damage, and duration
of critical perfusion were shown in PET imaging
studies of cats following MCA occlusion (Heiss et al,
1994, 1997). The PET imaging studies have suggested
three phases of injury progression:
(1) At flows below the threshold of energy metabolism
(B20% to 30% of preocclusion values), acute
ischemic injury is usually established within
minutes after the onset of ischemia (Branston
et al, 1974, 1977; Astrup et al, 1977, 1981).
(2) During the subsequent subacute phase (flows
ranging from 25% to 50% of preocclusion values),
the core expands into the penumbra, defined as
areas of decreased CBF and O2 metabolism, but
increased OEF. In most models, the core has
consumed the entire penumbra within 4 to
6 hours, whereas in some animal models and in
some humans (Heiss et al, 1992), the extension of
the core into the penumbra can require > 24 hours.
This expansion is postulated to be attributed in
part to peri-infarct spreading depression, and other
factors related to the failure to restore CBF. Periinfarct spreading depressions are initiated at the
border of the infarct core and spread over the
ipsilateral hemisphere. During spreading depression, the metabolic rate of the tissue markedly
increases in response to the activated ion exchange
pumps (Somjen, 2001), but is not associated with
an increased rCBF in ischemic areas of the brain
(review in the study by Strong (2009)). Thus,
the increased metabolic workload coupled with
limited O2 supply leads to transient episodes of
worse hypoxia and stepwise increases in lactate
with each depolarization, loss of ionic gradients,
and other molecular disturbances that increase
cell death (Mies et al, 1993; Hossmann, 2006).
(3) A delayed phase of injury evolution occurs that
can last for several days to weeks. Using multiparametric imaging techniques for differentiation
between the core and the penumbra, by 1 hour

after proximal MCA occlusion in the cat, the


penumbra approximately predicts the size of the
final infarct if flow is not restored (Heiss et al,
1994). However, after 3 hours, > 50% of the
penumbra progresses to infarction, and between
6 and 8 hours, almost all of the penumbra
disappears and is converted into the irreversibly
damaged infarct core (see Figure 5 below).
The flow values for irreversible damage and for
functional impairment from experimental models of
ischemia in cats correspond to those observed in
humans. Considerable variability exists (damage
below 4.8 to 17.3 mL/100 g per min, penumbra below
14.1 to 35.4 mL/100 g per min), which depends on
the definition of damage/penumbra, the methods
used for flow determination, the time of measurement after stroke, and whether occlusion was
permanent or temporary (for review see the study
by Heiss (2000)).

Molecular Characteristics of the


Ischemic Penumbra and the Core
During focal ischemia, the molecular events that
correspond best to the physiologic and imaging
definitions of the penumbra are: (1) regions of
decreased protein synthesis with preserved ATP
and (2) regions of Hsp70 induction in neurons. Many
molecular events occur well outside the clinically
defined penumbra of ischemia-induced injury that
does not lead to detectable infarction. These are
associated with vascular changes during noninjurious decreases in blood flow, inflammation with loss
of cells and their processes in the core, and synaptic/
brain reorganization.
Kleihues and Hossmann (1971) and Hossmann
(1993) first described decreased cerebral protein
synthesis following focal cerebral ischemia. As rCBF
decreases to B30% to 50% of baseline (Mies et al,
1990), blockade of translation and decreased protein
synthesis are observed (Dienel et al, 1980; Kiessling
et al, 1986; Bergstedt et al, 1993; Hossmann, 1994).
Within the core region of ischemic injury, protein
synthesis decreases early and is associated with ATP
loss and irreversible translation blockade, whereas
in the penumbra, protein synthesis is initially
depressed, ATP remains normal, and then protein
synthesis recovers over time (Hossmann, 1993, 1994).
The Unfolded Protein Response

The decrease in protein synthesis in the ischemic


core is mediated in part by the unfolded protein
response within the cell endoplasmic reticulum.
Translation blockade proceeds through initiation,
maintenance, and termination (DeGracia and Hu,
2007). Initiation of the translation blockade in part
involves the PKR-like endoplasmic reticulum kinase
that phosphorylates eukaryotic initiation factor-2a
Journal of Cerebral Blood Flow & Metabolism (2011) 31, 18361851

Heterogeneity in the penumbra


GJ del Zoppo et al
1840

and blocks translation in the endoplasmic reticulum.


If eukaryotic initiation factor-2a is phosphorylated,
the initiation complex is disrupted and translation
is slowed or blocked (Hu and Wieloch, 1993). This
is associated with disaggregation of polysomes,
followed by the appearance of stress granules
(DeGracia and Hu, 2007). The PKR-like endoplasmic
reticulum kinase knockouts only show early translation blocks after ischemia, so that eukaryotic
initiation factor-2a is related to the early, but not
prolonged, blockade of protein synthesis following
ischemia. After the initial translation blockade, other
transcription factors, including eukaryotic initiation
factor-4(E,F,G), likely maintain decreased protein
synthesis for 4 to 24 hours and perhaps longer. This
maintenance phase is followed either by (1) recovery
of translation or by (2) permanent translation blockade and cell death, which constitute the termination
phase (DeGracia et al, 2008). The proximate stimuli
for the unfolded protein response in cerebral ischemia are still unknown, but may include increased
Ca + 2 in the endoplasmic reticulum, impaired aminoacid or glucose delivery, abnormal glycosylation
of proteins, and other factors (Paschen, 1996, 1998;
Kaufman et al, 2002, 2004; Schroder and Kaufman,
2005). It has been suggested by several groups that
protein synthesis is arrested following ischemia
to prevent formation of partially denatured or
improperly processed proteins, which is probably
protective. How protein synthesis resumes in the
penumbra that will be salvaged and why protein
synthesis remains suppressed in regions of penumbra that infarct are still not understood, but are under
study.

Heat-Shock Protein Induction

Another major molecular responder to ischemia is


the family of Hsps. The Hsps, expressed in every
living cell and organism, are induced in cells during
periods of ischemia and other stresses that produce
intercellular denatured proteins (Lindquist and
Craig, 1988; Craig et al, 1993; Lindquist and Kim,
1996; Massa et al, 1996; Yenari, 2002).
Now, Hsp70, the major inducible Hsp, is expressed
at almost undetectable levels in normal brain cells.
After heat stress, ischemia, and other stresses, Hsp70
is massively induced and becomes the most abundant protein in the cell because it binds denatured
proteins directly and attempts to refold them
(Beckmann et al, 1990, 1992; Welch, 1993; Welch
and Brown, 1996). Denatured proteins induce heatshock transcription factors that bind to heat-shock
elements in the Hsp genes and induce Hsp40, Hsp70,
Hsp90, and others. At this point, the injured cell
appears to make a decision: Hsp70 can promote
protein refolding and cell survival or allow protein
degradation and progression to cell death (Hohfeld
et al, 2001). Importantly, cells that synthesize Hsp70
mRNA but cannot synthesize Hsp70 protein likely
Journal of Cerebral Blood Flow & Metabolism (2011) 31, 18361851

die. Most cells that express both Hsp70 mRNA and


Hsp70 protein appear to survive.
After MCA occlusion, Hsp70 mRNA is expressed
throughout the MCA territory, both within the infarction and in regions adjacent to the infarction (Kinouchi
et al, 1993a, b, 1994). Hata et al (1998) have shown that
Hsp70 transcription (RNA) and Hsp70 translation
(protein) occur in regions of decreased protein synthesis, but preserved ATP. This is the area where blood
flow is 18% to 20% normal and in which infarction
would occur if reperfusion did not occur (Hata et al,
2000a, b, 1998). Under conditions in which Hsp70
transcripts are induced in the MCA distribution, the
Hsp70 protein is only synthesized in neurons peripheral to the ischemic core, whereas there is no or very
little Hsp70 protein synthesized in the core (Kinouchi
et al, 1993a). Thus, the ischemic core corresponds to a
region of either no Hsp70 mRNA induction or induction of Hsp70 mRNA, but without translation into the
Hsp70 protein. This contrasts with the molecular
events within the penumbra: regions of Hsp70 mRNA
induction and Hsp70 protein synthesis in the regions
of denatured proteins where cells can effectively refold
proteins (Figure 3). This also correlates with differential cellular vulnerability to ischemia because blood
vessels that synthesize Hsp70 mRNA and Hsp70
protein may well survive in areas of infraction where
neurons and glia succumb. Although the regions of
Hsp70 mRNA and Hsp70 protein induction at 24 hours
appear to correlate well with the clinical definition of
the penumbra, it is not known whether the volume of
tissue destined to infarction corresponds exactly to that
represented by the Hsp70 protein, because there is no
known independent molecular marker of tissue at risk
for infarction.
The degree and duration of ischemia affect
the regional expression of Hsp70. For example,
10 minutes of focal ischemia in the MCA distribution
(using the suture model), which does not infarct rat
brain, induces Hsp70 protein in neurons throughout
the MCA distribution 4 to 24 hours later. Thus, the
regions of Hsp70 protein induction indicate the
penumbra because this is the region at risk for
infarction, but which did not infarct because blood
flow was restored after 10 minutes of focal ischemia
(Zhan et al, 2008). In contrast, at 24 hours after
3-hour (suture-induced) MCA occlusion infarction
occurs in the MCA territory, and the small area of
Hsp70 induction indicates a small penumbra located
between the middle and anterior, and the posterior
cerebral arteries (Zhan et al, 2008). The expression
of Hsp70 in neurons outside areas of infarction is
presumed to protect these cells from further protein
denaturation, as overexpression of the Hsp70 protein
in transgenic mice markedly protects the brain
against infarction (Rajdev et al, 2000).
Heterogeneity of the Molecular Responses

The boundary of the penumbra reflects the


degree and duration of decreased cerebral perfusion.

Heterogeneity in the penumbra


GJ del Zoppo et al
1841

penumbra

*
mini-core

*
mini-penumbra

Figure 3 Development of mini-penumbra and mini-core. (A) Coronal section of the adult rat brain that shows Hsp70 protein
(stain) 24 hours after occlusion of the right middle cerebral artery (MCA) for 10 minutes. Hsp70 immunoreactivity in the cortex, basal
ganglia, and ventral thalamus and dorsal hypothalamus in the MCA distribution must be noted. Hsp70 staining delineates the entire
penumbra, because this region of brain did not infarct, but would have infarcted if MCA occlusion had persisted for 2 hours. (B) The
region of the cortex within the box in panel A shows a central area of little Hsp70 expression surrounded by a large number of
Hsp70-stained cells that are predominantly microglia (two are denoted with green stars). For the purposes of this review, we have
designated the central area that represents a possible microscopic infarct as the mini-core and the surrounding area of Hsp70stained glia and some neurons as the mini-penumbra. This figure is adapted from the study by Zhan et al (2008). Hsp70, heatshock protein 70.

Outside this region, the hypoperfused ischemic brain


tissue contains neurons, glia, and vascular elements
together with inflammatory cells that are associated
with gliosis, synaptic pruning, and other structural
and molecular changes presaging recovery after
stroke.
In contrast to a central zone of hypoperfused brain
at risk for infarction, the pattern and evolution of the
penumbra can be quite variable. For example, in
some models of cerebral infarction, there are no
zones that correspond to decreased protein synthesis
and Hsp70 proteinand thus there is no penumbra.
In other models, there are small islands of the
infarcted brain (mini-cores) surrounded by neurons
and glial cells that express Hsp70 (mini-penumbras) (Figure 3). The most-stressed cells at the
margins of these islands are sometimes neurons and
sometimes microglia. The features that determine
this heterogeneity are unknown, but likely reflect a
dynamic interplay between adjacent microvascular
beds and complex interactions between the microvessels and the adjacent glia and neurons that differ
over regions of 30 to 40 mm. How this relates to
infarction without the intervening surviving tissue is
still unknown. Indeed, although neurons are more
vulnerable than glia to ischemia, there is eventually
a discrete demarcation between the infarcted brain
and the surviving brain where both viable neurons
and glia exist at the margin and where their
companions have perished on the infarcted side of
the margin. The factors that define such a sharp
margin are unknown, but may reflect interdependent survival growth factors responsible for the
survival of all members of a given neurovascular
unit, or regions lacking adequate microvascular
supply.
The data in Figure 3 strongly support the idea
of mini-penumbras and mini-cores based on the

pattern of stress gene response. Histologic data


obtained from the study by Hughes et al (2010) also
support this concept because they found patchy
neuronal loss and associated microglial activation in
the salvaged cortical penumbra after brief MCA
occlusion. In addition, we have shown very patchy
areas of microinfarction in the striatum and occasionally in the cortex after 5- and 10-minute periods
of focal ischemia produced with the suture technique in rats (Zhan et al, 2008). These mini-cores
were microscopic and defined by contiguous loss
of NeuN-immunostained neurons associated with
contiguous loss of glial fibrillary acidic protein
(GFAP)-stained astrocytes in the same region.

Vascular Characteristics of the


Ischemic Penumbra and the Core
Heterogeneity in the development of cortical neuron
injury has been shown in a rodent model of focal
cerebral ischemia (Dawson and Hallenbeck, 1996).
One provocative view has suggested that vascular
and hemostatic responses vary depending on the
specific organ, although little information was available about the cerebral vasculature (Rosenberg and
Aird, 1999). It is now known that cellular and
molecular responses of cerebral microvessels to focal
ischemia are heterogeneously distributed in space
and time very early in the ischemic territory.
After MCA occlusion in the nonhuman primate,
neuron injury and the microvessel responses are
rapid and are distributed in a characteristic topographical arrangement with regard to the development of the ischemic core. Tagaya et al (1997) have
shown that in the evolving core early after injury
(defined by evidence of DNA strand breaks or repair),
80.0%6.6% of injured cells were neurons at
Journal of Cerebral Blood Flow & Metabolism (2011) 31, 18361851

Heterogeneity in the penumbra


GJ del Zoppo et al
1842

2 hours after MCA occlusion, but there is a much


lower proportion of injured endothelial cells at later
times ( < 2% of cells at 27 hours). This is consistent
with the notion that neurons are most sensitive to
ischemia, whereas vascular endothelial cells are
most resistant. However, microvessels within the
territories of injury are rapidly reactive, participating
in inflammatory cell adhesion, and displaying
changes in barrier permeability and matrix integrity.
These observations indicate heterogeneity in cell
vulnerability to injury within the developing core
and adjacent penumbra. Indeed, in the primate
corpus striatum after proximal MCA occlusion,
microvessel alterations and injury within the regions
of the ischemic core are distributed heterogeneously
(Tagaya et al, 2001).
Microvessel Matrix Receptor Responses

This heterogeneity in the evolution of injury is also


shown by the response of integrin a1b1 expression by
microvessel endothelial cells in regions of the
territory at risk that become the core. Integrin a1b1
is a common receptor for endothelial cell adhesion to
laminin, collagen IV, and perlecan in the microvessel
basal lamina. The b1 integrins on the cerebral endothelium, and the integrin a6b4 as well as ab-dystroglycan on astrocyte end-feet all decrease significantly
within 2 hours after MCA occlusion (Wagner et al,
1997; Abumiya et al, 1999; Tagaya et al, 2001).
Tagaya et al (2001) have shown that the distributions
and density of microvessels suffering loss of a1 and
b1 subunits were heterogeneously distributed, in a
pattern that depended on the vascular supply.
Microvessels in the ischemic striatum expressed
increased b1 subunit transcripts in boundaries
around central regions devoid of b1 integrin transcription. To help explain these observations, in vitro
integrin b1 subunit mRNA expression increased
when murine brain endothelial cells (grown on
collagen type IV, laminin, or perlecan) were exposed
to moderate oxygenglucose deprivation (experimental ischemia) (Milner et al, 2008). Hence, the regions
of endothelial cell response in the striatum of the
nonhuman primate can be interpreted as multiple
mini-penumbras made up of areas of b1 integrin
mRNA upregulation on the edges of each core region
and absent b1 integrin expression in mini-cores
because of the failure of transcription and translation
in the areas of infarction (Figure 4A) (Tagaya et al,
1997, 2001).

responsible for the upregulation of integrin avb3,


and stimulates proliferation, migration, and increased permeability of the endothelium (Senger
et al, 1996). Abumiya et al have shown that during
striatal ischemia, VEGF mRNA was detectable in
noncapillary microvessels in relationship to the avb3
expression. Colocalization studies, involving microvessels reconstructed over 100 mm of their length in
regions of dUTP incorporation (the core), showed
a significant colocalization among proliferating cell
nuclear antigen (PCNA), the integrin avb3, and VEGF
along the length of activated microvessels (Abumiya
et al, 1999). Integrin avb3 was upregulated mainly on
7.5- to 30.0-mm diameter microvessels within the
core region. Microvessel-associated PCNA, VEGF,
and integrin avb3 were all upregulated in this region
(Figure 4B). Using a hierarchical log-linear model for
the categorical data analysis, tests for pairwise
interactions at different times showed that the
interaction with time was not significant. Therefore,
coexpression of PCNA, VEGF, and integrin avb3 by
microvessels within the ischemic core indicated a
highly significant interaction that was independent
of time, and was therefore heterogeneous within the
core regions.
An interesting observation in this respect suggests
that in some settings, cerebral microvessels in a
territory could be variously primed for response,
such that their response is heterogeneous. Ruetzler
et al (2001) have shown segmental heterogeneity of
central nervous system vascular responses in normoxic spontaneously hypertensive stroke-prone rats
exposed to lipopolysaccharide, in which circular
decoration of brain tissue by manganese superoxide
dismutase was seen around some microvessels
and not others. This is possibly relevant to the observations made by Mabuchi et al (2005) of an ordered,
but heterogeneously distributed, neuron injury
within 2 hours of MCA occlusion that is related to
the microvessel supply (the neurovascular unit).
Of importance, these observations are not simply
explained by the dependence of local injury on the
fall-off of O2 diffusion as applied to cerebral
capillaries (the Krogh cylinder), but suggest other
mechanisms (Quistorff et al, 1977; Mabuchi et al,
2005). Whether the coordinated expression of PCNA,
integrin avb3, and VEGF by cerebral microvessels
during focal ischemia is an intrinsic characteristic
of some vessels of a size class, or the heterogeneity of
the injury, is not yet certain.

Heterogeneity of the Microvascular Responses


Angiogenesis

Another example of vascular response heterogeneity


to focal ischemia is the increased expression of
integrin avb3 and vascular endothelial growth factor
(VEGF) by cerebral microvessels within the ischemic
core during MCA occlusion (Abumiya et al, 1999).
Vascular endothelial growth factor expression is
Journal of Cerebral Blood Flow & Metabolism (2011) 31, 18361851

Responses of the microvasculature in the territory at


risk (striatum) to focal ischemia are very rapid,
nearly as rapid as those of the neurons they supply.
Within the core regions, in the first minutes after
ischemia onset, microvessel obstruction, endothelial
cell receptor presentation, matrix degradation, and
detachment of astrocyte end-feet occur initially in

Heterogeneity in the penumbra


GJ del Zoppo et al
1843

Figure 4 Expression of microvessel-related gene products at 2 hours after MCA occlusion in the striatum. (A) Expression of the
integrin b1 subunit mRNA by microvessels in the striatum of three nonhuman primate subjects (ac) after MCA:O (Tagaya et al,
2001). Around mini-cores of absent b1 subunit mRNA, significant upregulation of the b1 subunit gene product was observed. The
cores and boundary zones of b1 subunit upregulation corresponded to the regions where cells incorporated dUTP (i.e., dUTP + )
(Tagaya et al, 1997). (B) Regions of VEGF expression (black), an subunit (gray), and expression of both products (hatched) by
activated microvessels in the striata of nonhuman primates that were subject to various periods of MCA occlusion (each image
represents a separate animal) (Abumiya et al, 1999). The mini-cores of integrin or VEGF expression by microvessels must also be
noted. MCA, middle cerebral artery; VEGF, vascular endothelial growth factor.

a heterogeneous manner in pockets (mini-cores).


The initial microvessel responses are neither simultaneous nor homogeneous. This suggests that in the
early moments after arterial occlusion, the entire

core is studded with pockets of penumbra, and


that these mini-cores and their mini-penumbras
evolve dynamically and heterogeneously depending
on local differences of microvessel perfusion.
Journal of Cerebral Blood Flow & Metabolism (2011) 31, 18361851

Heterogeneity in the penumbra


GJ del Zoppo et al
1844

Imaging the Ischemic Penumbra and


Core
Noninvasive brain imaging modalities have evolved
greatly since the first descriptions of the core
penumbra relationships in large animal models.
The use of PET and MR imaging techniques in
humans is now beginning to confirm and even
extend observations of the core and penumbra
defined in animal models.
Positron Emission Tomography Imaging

Positron emission tomography techniques have


permitted classification of three regions within the
disturbed vascular territory: (1) the core of ischemia
which usually undergoes necrosis with a flow rate
< 12 mL/100 g per min, (2) a penumbra region (12 to
22 mL/100 g per min) of still viable tissue, but
with uncertain chances for infarction or recovery
depending on reflow, and (3) a hypoperfused area
( > 22 mL/100 g per min) not primarily damaged by
decreased blood supply that will not proceed to
infarction (see Figure 5). In humans, the brain tissue
with rCBF < 12 mL/100 g per min and rCMRO2
< 65 mmol/100 g per min (often measured several
hours after stroke) usually displays eventual evidence
of infarction on late computerized tomographic scans
(Ackerman et al, 1981; Baron et al, 1981a; Lenzi et al,
1982; Powers et al, 1985). Relatively preserved CMRO2
in regions of severely reduced CBF has been taken as
an indicator of maintained neuronal function in
regions with severely reduced CBF. This pattern,
coined misery perfusion, has served as a PET-derived
definition of the penumbra (Baron et al, 1981b). It is
characterized as the area of increased OEF ( > 80%
from the normal value of B40%). Positron emission
tomography investigations imply that the extent of the
penumbra depends on the time of measurement
relative to the onset of ischemia. The volume is large
and rCBF is low if the penumbra is defined in the first
hours of ischemia; the penumbra volume is small, if
defined later (Heiss et al, 1992). The heterogeneity in
the core of ischemia and in the penumbra over time
can be seen in the occurrence of OEF especially in
animal studies (Figure 5), but also seen repeatedly in
human PET studies (e.g., Heiss et al, 1992).
As these measurements require arterial blood
sampling and complex logistics, a marker of neuronal integrity is preferable. The central benzodiazepine receptor ligand flumazenil (FMZ) binds to the
GABA receptor that is abundant in the cerebral
cortex. These receptors are sensitive to ischemic
damage and can identify early neuronal loss. Flumazenil, validated in the cat MCA occlusion model,
predicted the size of final infarction in patients with
acute ischemic stroke, and showed the efficacy of
thrombolytic treatment (Heiss, 2000). Another marker of the penumbra, 18F misonidazol is trapped in
viable hypoxic tissue (Takasawa et al, 2007). In early
Journal of Cerebral Blood Flow & Metabolism (2011) 31, 18361851

stroke, increased 18F misonidazol uptake surrounds


the core, and there is a strong association between
the extent of 18F misonidazol-binding tissue that survives and functional outcome (Markus et al, 2003).
18
F misonidazol uptake also assesses hypoxia in the
white matter, where progression of ischemic damage
is slower (Heiss et al, 2000; Spratt et al, 2007).
As PET methodology has improved, higher resolution with shorter time sequences have detected
temporal and spatial heterogeneity of the core and
penumbra (Kidwell et al, 2003), which were especially evident in sequential studies in animal models
of focal and reversible cerebral ischemia (Heiss et al,
1994).

Magnetic Resonance Imaging

Although PET remains the imaging gold standard for


identification of tissue in the penumbra in human
stroke patients, MR studies using diffusion and
perfusion imaging by MR have provided valuable
insights into the relationships between the core and
the penumbra. More than a decade ago, it was
hypothesized that the early diffusion-weighted imaging (DWI) lesion estimates the ischemic core in
ischemic stroke patients and adjacent critically
hypoperfused tissue could be identified with perfusion-weighted imaging (PWI) (Baird et al, 1997;
Barber et al, 1998). Therefore, brain regions with a
PWI lesion that did not show restricted diffusion
(PWI/DWI mismatch) were hypothesized to represent the penumbra. Subsequent studies have
extended this notion by showing that appropriate
quantification of perfusion and diffusion imaging
can provide a reasonable estimate of the penumbra.
Problems with the MRI definition of the penumbra
include the fact that there is no threshold within a
region of oligemia (Kidwell et al, 2003; Kane et al,
2007; Toth and Albers, 2009), and that the PWI
abnormality often overestimates the final infarction
volume, and hence the amount of tissue at risk
(Parsons et al, 2001). In addition, the initial diffusion
lesion does not necessarily define the core of
infarcted tissue because some diffusion lesions can
be transiently or permanently reversed if blood flow
is rapidly restored (Kidwell et al, 2000; Parsons et al,
2002; Chalela et al, 2004).
Kidwell et al (2003) proposed a modified model of
ischemia-compromised tissue in which the penumbra includes the region of perfusiondiffusion mismatch, minus the region of benign oligemia plus a
portion of the initial diffusion abnormality itself.
Although several attempts have been made to
identify perfusion or apparent diffusion coefficient
(ADC) thresholds so as to better differentiate these
regions, consensus on the best method has been
hampered by the lack of methodological standardization of image postprocessing and analysis. These
restrict the pooling of data and cross-comparison of
results across studies (Kane et al, 2007).

Heterogeneity in the penumbra


GJ del Zoppo et al
1845

Figure 5 Sequential PET images of CBF, CMRO2, and OEF of permanent MCA occlusion in cats (left columns) compared with images
of patients 12 hours after stroke (right columns). Two different cat subjects are shown (efficient and nonefficient perfusion) and three
different patients are shown (A: early OEF defect with corresponding infarction on MRI, B: effective reperfusion without infarction,
and C: ineffective/delayed reperfusion with large final partially hemorrhagic infarction). In the cat, the progressive decrease of CMRO2
and the reduction of OEF predict infarction. In the patient, the area with preserved OEF is not infarcted (outside region on late MRI,
upper part of figure A). If reperfusion occurs before OEF is reduced, tissue can be salvaged (left: cat, and left: patient in the lower part
of the figure B). If reperfusion is achieved after this therapeutic window, tissue cannot be salvaged (right: cat, and right: patient
in lower part of the figure C). CBF, cerebral blood flow; CMRO2, cerebral metabolic rate for oxygen; MCA, middle cerebral artery;
MRI, magnetic resonance imaging; OEF, oxygen extraction fraction; PET, positron emission tomography.

Validation of MR signatures with regard to PET


measurements might help in the interpretation of
the respective findings and in the assessment of
the accuracy of the various measures for predicting
tissue outcome. Comparisons of PET and MR imaging
have been performed in small groups of patients to
assess perfusion, and the delineation of tissue in the
penumbra (Figure 5).
To predict irreversible cortical damage, results
from sequential early DWI and 11C-FMZ-PET outcomes were compared with infarct extension 24 to
48 hours later on T2-weighted MRI in 12 acute stroke
patients (Heiss et al, 2004). When the volumes of
tissue beyond the defined thresholds of FMZ binding
and DWI intensity were compared, close correlations
(1) between volumes with FMZ and DWI beyond
threshold and (2) between predicted and final infarct
volumes were obtained, but the volumes did not
completely overlap. Whereas false-positive results
were observed for 25.9% of the total volume of DWI

increase, they were negligible with FMZ-PET. A


time-to-peak delay of 4 seconds correlated with flow
decreases below 20 mL/100 g per min as identified
by H2 15O-PET (Zaro-Weber et al, 2009).
For demarcation of the volume of the penumbra,
the areas of PWI/DWI mismatch were compared with
those of increased OEF. Using PWI/DWI mismatch,
there was considerable variability in penumbra
volume, and all patients (13/13) showed areas of
mismatch. However, PET detected OEF in only 8 of
13 patients. The areas of OEF elevation were always
located within the areas of time-to-peak prolongation, and were significantly smaller and covered only
1% to 75% (median, 33%) of the time-to-peak area
on MRI. These data show a high sensitivity, and a
low specificity of the chosen thresholds to identify
the penumbra as defined by PET. This suggests that
the PWI/DWI volume ratios depicted by time to peak
do not reliably reflect the penumbra as defined
by PET.
Journal of Cerebral Blood Flow & Metabolism (2011) 31, 18361851

Heterogeneity in the penumbra


GJ del Zoppo et al
1846

Advances in Perfusion-Weighted Imaging Techniques

Nonetheless, the spatial correspondence between


early DWI and PWI lesions, in conjunction with
knowledge of whether reperfusion of the supply
artery(ies) occurs, allow a reasonable (but not a
highly accurate) prediction of the final infarction
volume. More advanced PWI techniques, including
deconvolution sequences that account for differences
in arterial input to vessels with normal flow, appear
to produce more accurate estimates of the regions of
hypoperfusion that define the outer boundaries of
the penumbra. Although PWI provides a number of
parameters including mean transit time and cerebral
blood volume, recently there has been focus on the
maximum of the tissue residue function (Tmax)
obtained by deconvolution (Zaro-Weber et al, 2010).
Although the physiologic interpretation of Tmax is
complex, it reflects the degree of lag between the
arterial input and the tissue response, as well as
dispersion and mean transit time (Calamante et al,
2010). This parameter appears to correlate with CBF
values determined by Xenon computerized tomographic scans and has performed well in two
prospective clinical stroke trials (Albers et al, 2006;
Davis et al, 2008).
Thresholding techniques that restrict the volume
of PWI lesions to brain regions where there are
substantial delays in contrast arrival or transit times
have also been used. This reduces the volume of
benign oligemia captured by the PWI map. Prediction of the final infarction volume and salvage of the
penumbra seems to be more accurate using Tmax
maps with higher thresholds. In the DEFUSE (Diffusion and Perfusion Imaging Evaluation for Understanding Stroke Evolution) study, for instance, the
correlation between infarct growth and the volume
of penumbra salvaged was significantly better for
PWI lesions defined by Tmax > 6 seconds compared
with > 2 seconds (Olivot et al, 2009). In addition, in
patients who did not experience early reperfusion,
the > 4-second threshold was a more accurate
predictor of final infarction volume than Tmax
> 2 seconds. Similarly, in EPITHET (Echoplanar
Imaging Thrombolytic Evaluation Trial), both the
size and the severity of PWI lesions correlated with
clinical outcomes (Parsons et al, 2010). Recent data
have suggested a B90% sensitivity and specificity
for Tmax > 5.5 seconds to identify the PET-defined
penumbra rCBF threshold of 20 mL/100 g per min
(Zaro-Weber et al, 2010). These results suggest that
quantification of the severity of PWI lesions can
improve accuracy for identification of tissue that is
in the penumbra.
Advances in Diffusion-Weighted Imaging Techniques

Progress has been made in quantification of DWI


using ADC measurements. Animal studies and
human trials are congruent, both showing correlations between CBF and the severity of ADC decline
Journal of Cerebral Blood Flow & Metabolism (2011) 31, 18361851

(Kohno et al, 1995; Hoehn-Berlage et al, 1995;


Dijkhuizen et al, 1997; Thijs et al, 2002; Lin et al,
2003). With increasing time from symptom onset,
ADC values become progressively reduced in a
critically low CBF environment. Recent data have
shown that only a small volume of the acute DWI
lesion is reversible (Chemmanam et al, 2010).
Diffusion-weighted imaging lesions are most likely
to reverse if they have modest reductions in ADC and
there is early reperfusion (Olivot et al, 2009).
However, observations in both animal models and
in human stroke patients show that reperfusion is
associated with a rapid increase in ADC values, and
that this increase is not necessarily reflective of
tissue salvage. Therefore, an isolated ADC map can
be an unreliable predictor of eventual infarct volume
(Ringer et al, 2001).
The fact that early DWI lesions are not uniformly
incorporated into the final infarction has an important implication: it is likely that some part of the
ischemic penumbra is contained within the acute
DWI lesion. Additional research is required to more
accurately determine which regions of the acute DWI
lesions are most likely to be salvageable, i.e., part of
the penumbra.

The Core and Penumbra are Dynamic

Experimental data from vascular and molecular


modeling studies, as well as recent imaging work
indicate that the penumbra is dynamic. There appear
to be substantial fluctuation in volumes, locations,
and structures of DWI and PWI lesions during the
early hours after stroke onset (Ma et al, 2009).
Coregistration of acute DWI lesions with PET
imaging has shown that DWI lesions are heterogeneous and include regions of variable metabolic
disruption and flow-metabolism coupling (Guadagno
et al, 2006). These findings are compatible with the
observations discussed above regarding the relationship among rCBF, ADC, and DWI. Therefore, predicting the fate of ischemic brain tissue based on data
obtained from any single imaging modality performed at only one early time point may not have
high specificity and sensitivity for prediction of the
core and penumbra.
Visualization of the spatial relationships between
PWI and DWI lesions at different times after stroke
onset can be achieved by coregistration of sequential
MRI scans in stroke patients. Three-dimensional
analyses have shown that a central volume of restricted diffusion (core) surrounded by a hypoperfused
penumbra is not present in the majority of stroke
patients imaged 3 to 6 hours after symptom onset
(before administration of thrombolytic therapy).
Multiple discrete regions of restricted diffusion or
perfusion were documented in one-third of the
patients enrolled in the DEFUSE study (Figure 6).
Surprisingly, a substantial proportion of the acute
DWI lesions (54%) did not have a superimposed

Heterogeneity in the penumbra


GJ del Zoppo et al
1847

Figure 6 Heterogeneity of DWI and PWI lesion structure. (A) Examples of the three-dimensional structure of DWI and PWI lesions in
acute stroke patients. Left, a single PWI lesion; center, a DWI lesion with multiple individual lesion components; right, a PWI lesion
with multiple components. (B) Three-dimensional imaging of DWI and PWI lesions in acute stroke patients reveals intertwined
regions of mismatch, DWI/PWI overlap, and early reperfusion. Regions that contain DWI lesions without superimposed PWI lesions
(early reperfusion) are shown in blue; areas where DWI lesions have superimposed PWI lesions are shown in purple. The red areas
are PWI lesions without superimposed DWI lesions (regions of mismatch). Reprinted from Stroke with permission from Olivot et al
(2009). DWI, diffusion-weighted imaging; PWI, perfusion-weighted imaging.

perfusion lesion (Olivot et al, 2009). These regions


of restricted diffusion without superimposed PWI
lesions are assumed to represent regions where
perfusion deficits have resolved or shifted to adjacent brain regions. The term RADAR (Reversible
Acute Diffusion lesion Already Reperfused) has been
used to describe these regions (Olivot et al, 2009).
The RADAR regions appear to have an increased rate
of DWI reversal. These findings emphasize the
heterogeneity and complexity of the relationships
between acute PWI and DWI lesions.

mini-cores

normal
+
mini-penumbras

normal

core

penumbra

The Penumbra Revisited


The original notion that occlusion of a brainsupplying artery produces a central core of tissue
injury destined for infarction that is surrounded by a
penumbra of metabolically metastable tissue with
the potential for full recovery still has great merit.
This model is strongly supported by the pathologic
findings of stroke that show confluent regions of
tissue infarction in animal models and patients.
Recent experimental and imaging work offer the
refinement that in the early minutes and hours after
ischemia onset, the core contains pockets of injury

Figure 7 A new hypothetical construct of the penumbra. The


architecture and reversibility depend on time and location of
rCBF reduction in the territory at risk after occlusion of a brainsupplying artery. The outside boundary represents the territory at
risk of cerebral tissue that functions normally until rCBF is
reduced for an extended period. Mini-cores coalesce with the
duration of rCBF reduction devouring micro-penumbras. The
period of evolution from normal function to the final state of the
territory at risk may depend on a number of factors, including
tissue location, depth of reduction of rCBF, inflammatory state at
baseline and/or degree of inflammatory response, and other
factors. The location of the mini-cores and mini-penumbras
appear heterogeneously distributed. However, they in fact reflect
the microvascular supply of tissue and cell vulnerabilities. rCBF,
regional cerebral blood flow.
Journal of Cerebral Blood Flow & Metabolism (2011) 31, 18361851

Heterogeneity in the penumbra


GJ del Zoppo et al
1848

which we characterize as mini-cores, surrounded


by mini-penumbras. We hypothesize that unless
salvaged, such embedded mini-penumbras are
consumed by the expanding mini-cores of metabolic
and functional failure to generate a lesion that is
ultimately homogenous and can grow into the surrounding injured tissue (Figure 7). Furthermore, it is
hypothesized that the mini-cores contain tissues in
which neurovascular units have been irreversibly
damaged because of flow cessation and its consequences, whereas the mini-penumbras have a proportion of neurovascular units that are viable. The separate
networks of microvessels, neurons, and glia associate
the mini-cores with the mini-penumbras. In this
way, if unimpeded, these mini-cores can grow into
their respective mini-penumbras to encompass a larger
region of injury.
This refined view has several implications:
1. Multiple mini-penumbras are apparent at the
molecular, cellular, and microvessel levels, such
that higher imaging resolution will be necessary to
follow their fate in patients,
2. these cell and microvessel events are related to
local microvascular flow,
3. serial imaging studies are required to show the
evolution of this dynamic core and penumbra,
4. inflammatory cellendothelial interactions, periinfarct depolarization, flow changes, and other
processes contribute to the engulfment of minipenumbras into the evolving ischemic core,
5. these observations underscore the need for
extremely early interventions, and
6. depending on the vascular territory involved and
the timing and severity of the ischemic event,
injury evolution may occur at different speeds
within different mini-cores/mini-penumbras,
such that certain interventions might have benefit
later.
Future studies will need to determine whether the
presence or absence of mini-cores and minipenumbras might affect the efficacy of different
types of treatment, the timing of treatment, or impact
ultimate prognosis.

Acknowledgement
The authors thank G Berg for manuscript preparation.

Disclosure/Conflict of interest
The authors declare no conflict of interest.

References
Abumiya T, Lucero J, Heo JH, Tagaya M, Koziol JA,
Copeland BR, del Zoppo GJ (1999) Activated microvessels express vascular endothelial growth factor and
Journal of Cerebral Blood Flow & Metabolism (2011) 31, 18361851

integrin alpha(v)beta3 during focal cerebral ischemia.


J Cereb Blood Flow Metab 19:103850
Ackerman RH, Correia JA, Alpert NM, Baron JC, Gouliamos
A, Grotta JC, Brownell GL, Taveras JM (1981) Positron
imaging in ischemic stroke disease using compounds
labeled with oxygen 15. Initial results of clinicophysiologic correlations. Arch Neurol 38:53743
Albers GW, Thijs VN, Wechsler L, Kemp S, Schlaug G,
Skalabrin E, Bammer R, Kakuda W, Lansberg MG,
Shuaib A, Coplin W, Hamilton S, Moseley M, Marks
MP (2006) Magnetic resonance imaging profiles predict
clinical response to early reperfusion: the Diffusion and
Perfusion Imaging Evaluation for Understanding Stroke
Evolution (DEFUSE) study. Ann Neurol 60:50817
Astrup J, Siesjo BK, Symon L (1981) Thresholds in cerebral
ischemiathe ischemic penumbra. Stroke 12:7235
Astrup J, Symon L, Branston NM, Lassen NA (1977)
Cortical evoked potential and extracellular K+ and H+
at critical levels of brain ischemia. Stroke 8:517
Baird AE, Benfield A, Schlaug G, Siewert B, Lovblad KO,
Edelman RR, Warach S (1997) Enlargement of human
cerebral ischemic lesion volumes measured by diffusion-weighted magnetic resonance imaging. Ann Neurol
41:5819
Barber PA, Darby DG, Desmond PM, Yang Q, Gerraty RP,
Jolley D, Donnan GA, Tress BM, Davis SM (1998) Prediction of stroke outcome with echoplanar perfusion- and
diffusion-weighted MRI. Neurology 51:41826
Baron JC (1999) Mapping the ischaemic penumbra with
PET: implications for acute stroke treatment. Cerebrovasc Dis 9:193201
Baron JC, Bousser MG, Comar D, Soussaline F, Castaigne P
(1981a) Noninvasive tomographic study of cerebral
blood flow and oxygen metabolism in vivo. Potentials,
limitations, and clinical applications in cerebral
ischemic disorders. Eur Neurol 20:27384
Baron JC, Bousser MG, Rey A, Guillard A, Comar D,
Castaigne P (1981b) Reversal of focal misery-perfusion
syndrome by extra-intracranial arterial bypass in hemodynamic cerebral ischemia. A case study with 15O
positron emission tomography. Stroke 12:4549
Beckmann RP, Lovett M, Welch WJ (1992) Examining the
function and regulation of hsp 70 in cells subjected to
metabolic stress. J Cell Biol 117:113750
Beckmann RP, Mizzen LE, Welch WJ (1990) Interaction of
Hsp 70 with newly synthesized proteins: implications
for protein folding and assembly. Science 248:8504
Bergstedt K, Hu BR, Wieloch T (1993) Postischaemic
changes in protein synthesis in the rat brain: effects of
hypothermia. Exp Brain Res 95:919
Branston NM, Strong AJ, Symon L (1977) Extracellular
potassium activity, evoked potential and tissue blood
flow. Relationships during progressive ischaemia in
baboon cerebral cortex. J Neurol Sci 32:30521
Branston NM, Symon L, Crockard HA, Pasztor E (1974)
Relationship between the cortical evoked potential and
local cortical blood flow following acute middle
cerebral artery occlusion in the baboon. Exp Neurol
45:195208
Calamante F, Christensen S, Desmond PM, Ostergaard L,
Davis SM, Connelly A (2010) The physiological significance of the time-to-maximum (Tmax) parameter in
perfusion MRI. Stroke 41:116974
Chalela JA, Kang DW, Luby M, Ezzeddine M, Latour LL,
Todd JW, Dunn B, Warach S (2004) Early magnetic
resonance imaging findings in patients receiving tissue
plasminogen activator predict outcome: insights into

Heterogeneity in the penumbra


GJ del Zoppo et al

the pathophysiology of acute stroke in the thrombolysis


era. Ann Neurol 55:10512
Chemmanam T, Campbell BC, Christensen S, Nagakane Y,
Desmond PM, Bladin CF, Parsons MW, Levi CR, Barber PA,
Donnan GA, Davis SM (2010) Ischemic diffusion lesion
reversal is uncommon and rarely alters perfusion-diffusion
mismatch. Neurology 75:10407
Craig EA, Gambill BD, Nelson RJ (1993) Heat shock
proteins: molecular chaperones of protein biogenesis.
Microbiol Rev 57:40214
Davis SM, Donnan GA, Parsons MW, Levi C, Butcher KS,
Peeters A, Barber PA, Bladin C, De Silva DA, Byrnes G,
Chalk JB, Fink JN, Kimber TE, Schultz D, Hand PJ,
Frayne J, Hankey G, Muir K, Gerraty R, Tress BM,
Desmond PM (2008) Effects of alteplase beyond 3 hours
after stroke in the Echoplanar Imaging Thrombolytic
Evaluation Trial (EPITHET): a placebo-controlled
randomised trial. Lancet Neurol 7:299309
Dawson DA, Hallenbeck JM (1996) Acute focal ischemiainduced alterations in MAP2 immunostaining: description of temporal changes and utilization as a marker for
volumetric assessment of acute brain injury. J Cereb
Blood Flow Metab 16:1704
DeGracia DJ, Hu BR (2007) Irreversible translation arrest in
the reperfused brain. J Cereb Blood Flow Metab 27:
87593
DeGracia DJ, Jamison JT, Szymanski JJ, Lewis MK (2008)
Translation arrest and ribonomics in post-ischemic
brain: layers and layers of players. J Neurochem
106:2288301
Dienel GA, Pulsinelli WA, Duffy TE (1980) Regional
protein synthesis in rat brain following acute hemispheric ischemia. J Neurochem 35:121626
Dijkhuizen RM, Berkelbach van der Sprenkel JW,
Tulleken KA, Nicolay K (1997) Regional assessment
of tissue oxygenation and the temporal evolution of
hemodynamic parameters and water diffusion during
acute focal ischemia in rat brain. Brain Res 750:
16170
Guadagno JV, Warburton EA, Jones PS, Day DJ, Aigbirhio
FI, Fryer TD, Harding S, Price CJ, Green HA, Barret O,
Gillard JH, Baron JC (2006) How affected is oxygen
metabolism in DWI lesions? A combined acute stroke
PET-MR study. Neurology 67:8249
Harris RJ, Symon L (1984) Extracellular pH, potassium, and
calcium activities in progressive ischaemia of rat cortex.
J Cereb Blood Flow Metab 4:17886
Hata R, Maeda K, Hermann D, Mies G, Hossmann KA
(2000a) Dynamics of regional brain metabolism and
gene expression after middle cerebral artery occlusion
in mice. J Cereb Blood Flow Metab 20:30615
Hata R, Maeda K, Hermann D, Mies G, Hossmann KA
(2000b) Evolution of brain infarction after transient focal
cerebral ischemia in mice. J Cereb Blood Flow Metab
20:93746
Hata R, Mies G, Wiessner C, Hossmann KA (1998)
Differential expression of c-fos and hsp72 mRNA in
focal cerebral ischemia of mice. Neuroreport 9:2732
Heiss WD (1992) Experimental evidence of ischemic
thresholds and functional recovery. Stroke 23:166872
Heiss WD (2000) Ischemic penumbra: evidence from
functional imaging in man. J Cereb Blood Flow Metab
20:127693
Heiss WD, Graf R, Lottgen J, Ohta K, Fujita T, Wagner R,
Grond M, Weinhard K (1997) Repeat positron emission
tomographic studies in transient middle cerebral artery
occlusion in cats: residual perfusion and efficacy of

postischemic reperfusion. J Cereb Blood Flow Metab


17:388400
Heiss WD, Graf R, Wienhard K, Lottgen J, Saito R, Fujita T,
Rosner G, Wagner R (1994) Dynamic penumbra demonstrated by sequential multitracer PET after middle
cerebral artery occlusion in cats. J Cereb Blood Flow
Metab 14:892902
Heiss WD, Hayakawa T, Waltz AG (1976) Cortical neuronal
function during ischemia. Effects of occlusion of one
middle cerebral artery on single-unit activity in cats.
Arch Neurol 33:81320
Heiss WD, Huber M, Fink GR, Herholz K, Pietryzyk V,
Wagner R, Weinhard K (1992) Progressive derangement
of peri-infarct viable tissue in ischemic stroke. J Cereb
Blood Flow Metab 12:193203
Heiss WD, Kracht L, Grond M, Rudolf J, Bauer B, Wienhard K,
Pawlik G (2000) Early [(11)C]Flumazenil/H(2)O positron
emission tomography predicts irreversible ischemic
cortical damage in stroke patients receiving acute
thrombolytic therapy. Stroke 31:3669
Heiss WD, Rosner G (1983) Functional recovery of cortical
neurons as related to degree and duration of ischemia.
Ann Neurol 14:294301
Heiss WD, Sobesky J, Smekal U, Kracht LW, Lehnhardt FG,
Thiel A, Jacobs AH, Lackner K (2004) Probability of
cortical infarction predicted by flumazenil binding and
diffusion-weighted imaging signal intensity: a comparative positron emission tomography/magnetic resonance
imaging study in early ischemic stroke. Stroke 35:
18928
Hoehn-Berlage M, Norris DG, Kohno K, Mies G, Leibfritz D,
Hossmann KA (1995) Evolution of regional changes in
apparent diffusion coefficient during focal ischemia
of rat brain: the relationship of quantitative diffusion
NMR imaging to reduction in cerebral blood flow and
metabolic disturbances. J Cereb Blood Flow Metab
15:100211
Hohfeld J, Cyr DM, Patterson C (2001) From the cradle to
the grave: molecular chaperones that may choose
between folding and degradation. EMBO Rep 2:88590
Hossman K-A, Mies G (2007) Multimodal mapping of the
ischemic penumbra in animal models. In: The Ischemic
Penumbra: Pathophysiology, Imaging and Therapy
(Donnan GA, Baron J-C, Davis SM, Sharp FR, eds),
New York: Informa Healthcare, 7792
Hossmann KA (1993) Disturbances of cerebral protein
synthesis and ischemic cell death. Prog Brain Res
96:16177
Hossmann KA (1994) Viability thresholds and the penumbra of focal ischemia. Ann Neurol 36:55765
Hossmann KA (2006) Pathophysiology and therapy of
experimental stroke. Cell Mol Neurobiol 26:105783
Hu BR, Wieloch T (1993) Stress-induced inhibition of
protein synthesis initiation: modulation of initiation
factor 2 and guanine nucleotide exchange factor
activities following transient cerebral ischemia in the
rat. J Neurosci 13:18308
Hughes JL, Beech JS, Jones PS, Wang D, Menon DK, Baron JC
(2010) Mapping selective neuronal loss and microglial
activation in the salvaged neocortical penumbra in the
rat. Neuroimage 49:1931
Jones TH, Morawetz RB, Crowell RM, Marcoux FW,
Fitzgibbon SJ, DeGirolami U, Ojemann RG (1981) Thresholds of focal cerebral ischemia in awake monkeys.
J Neurosurg 54:77382
Kane I, Carpenter T, Chappell F, Rivers C, Armitage P,
Sandercock P, Wardlaw J (2007) Comparison of 10

1849

Journal of Cerebral Blood Flow & Metabolism (2011) 31, 18361851

Heterogeneity in the penumbra


GJ del Zoppo et al
1850

different magnetic resonance perfusion imaging processing methods in acute ischemic stroke: effect on lesion
size, proportion of patients with diffusion/perfusion
mismatch, clinical scores, and radiologic outcomes.
Stroke 38:315864
Kaufman RJ (2004) Regulation of mRNA translation by
protein folding in the endoplasmic reticulum. Trends
Biochem Sci 29:1528
Kaufman RJ, Scheuner D, Schroder M, Shen X, Lee K, Liu
CY, Arnold SM (2002) The unfolded protein response in
nutrient sensing and differentiation. Nat Rev Mol Cell
Biol 3:41121
Kidwell CS, Alger JR, Saver JL (2003) Beyond mismatch:
evolving paradigms in imaging the ischemic penumbra
with multimodal magnetic resonance imaging. Stroke
34:272935
Kidwell CS, Saver JL, Mattiello J, Starkman S, Vinuela F,
Duckwiler G, Gobin YP, Jahan R, Vespa P, Kalafut M,
Alger JR (2000) Thrombolytic reversal of acute human
cerebral ischemic injury shown by diffusion/perfusion
magnetic resonance imaging. Ann Neurol 47:4629
Kiessling M, Dienel GA, Jacewicz M, Pulsinelli WA
(1986) Protein synthesis in postischemic rat brain: a
two-dimensional electrophoretic analysis. J Cereb Blood
Flow Metab 6:6429
Kinouchi H, Sharp FR, Chan PH, Koistinaho J, Sagar SM,
Yoshimoto T (1994) Induction of c-fos, junB, c-jun, and
hsp 70 mRNA in cortex, thalamus, basal ganglia and
hippocampus following middle cerebral artery occulsion. J Cereb Blood Flow Metab 14:80817
Kinouchi H, Sharp FR, Hill MP, Koistinaho J, Sagar SM,
Chan PH (1993a) Induction of 70-kDa heat shock protein
and hsp70 mRNA following transient focal cerebral
ischemia in the rat. J Cereb Blood Flow Metab 13:
10515
Kinouchi H, Sharp FR, Koistinaho J, Hicks K, Kamii H,
Chan PH (1993b) Induction of heat shock hsp70 mRNA
and HSP70 kDa protein in neurons in the penumbra
following focal cerebral ischemia in the rat. Brain Res
619:3348
Kleihues P, Hossmann KA (1971) Protein synthesis in the
cat brain after prolonged cerebral ischemia. Brain Res
35:40918
Kohno K, Back T, Hoehn-Berlage M, Hossmann K (1995)
Relationship between diffusion-weighted magnetic resonance images, cerebral blood flow and energy state in
experimental brain infarction. Magn Reson Imaging
13:6571
Lenzi GL, Frackowiak RSJ, Jones T (1982) Cerebral oxygen
metabolism and blood flow in human cerebral ischemic
infarction. J Cereb Blood Flow Metab 2:32135
Lin W, Lee JM, Lee YZ, Vo KD, Pilgram T, Hsu CY (2003)
Temporal relationship between apparent diffusion
coefficient and absolute measurements of cerebral blood
flow in acute stroke patients. Stroke 34:6470
Lindquist S, Craig EA (1988) The heat-shock proteins.
Annu Rev Genet 22:63177
Lindquist S, Kim G (1996) Heat-shock protein 104 expression is sufficient for thermotolerance in yeast. Proc Natl
Acad Sci USA 93:53016
Ma H, Zavala JA, Teoh H, Churilov L, Gunawan M, Ly J,
Wright P, Phan T, Arakawa S, Davis SM, Donnan GA
(2009) Fragmentation of the classical magnetic resonance mismatch penumbral pattern with time. Stroke
40:37527
Mabuchi T, Lucero J, Feng A, Koziol JA, del Zoppo GJ
(2005) Focal cerebral ischemia preferentially affects

Journal of Cerebral Blood Flow & Metabolism (2011) 31, 18361851

neurons distant from their neighboring microvessels.


J Cereb Blood Flow Metab 25:25766
Markus R, Reutens DC, Kazui S, Read S, Wright P,
Chambers BR, Sachinidis JI, Tochon-Danguy HJ,
Donnan GA (2003) Topography and temporal evolution
of hypoxic viable tissue identified by 18F-fluoromisonidazole positron emission tomography in humans after
ischemic stroke. Stroke 34:264652
Massa SM, Swanson RA, Sharp FR (1996) The stress gene
response in brain. Cerebrovasc Brain Metab Rev 8:
95158
Mies G, Iijima T, Hossmann KA (1993) Correlation between
peri-infarct DC shifts and ischaemic neuronal damage in
rat. Neuroreport 4:70911
Mies G, Paschen W, Ebhardt G, Hossmann KA (1990)
Relationship between of blood flow, glucose metabolism, protein synthesis, glucose and ATP content in
experimentally-induced glioma (RG1 2.2) of rat brain.
J Neurooncol 9:1728
Milner R, Hung S, Wang X, Berg GI, Spatz M, del Zoppo GJ
(2008) Responses of endothelial cell and astrocyte
matrix-integrin receptors to ischemia mimic those
observed in the neurovascular unit. Stroke 39:1917
Olivot JM, Mlynash M, Thijs VN, Purushotham A, Kemp S,
Lansberg MG, Wechsler L, Bammer R, Marks MP, Albers
GW (2009) Relationships between cerebral perfusion
and reversibility of acute diffusion lesions in DEFUSE:
insights from RADAR. Stroke 40:16927
Parsons MW, Barber PA, Chalk J, Darby DG, Rose S,
Desmond PM, Gerraty RP, Tress BM, Wright PM,
Donnan GA, Davis SM (2002) Diffusion- and perfusion-weighted MRI response to thrombolysis in
stroke. Ann Neurol 51:2837
Parsons MW, Christensen S, McElduff P, Levi CR,
Butcher KS, De Silva DA, Ebinger M, Barber PA,
Bladin C, Donnan GA, Davis SM, Echoplanar Imaging
Thrombolytic Evaluation Trial (EPITHET) investigators
(2010) Pretreatment diffusion- and perfusion-MR lesion
volumes have a crucial influence on clinical response
to stroke thrombolysis. J Cereb Blood Flow Metab
30:121425
Parsons MW, Yang Q, Barber PA, Darby DG, Desmond PM,
Gerraty RP, Tress BM, Davis SM (2001) Perfusion
magnetic resonance imaging maps in hyperacute stroke:
relative cerebral blood flow most accurately identifies
tissue destined to infarct. Stroke 32:15817
Paschen W (1996) Disturbances of calcium homeostasis
within the endoplasmic reticulum may contribute to the
development of ischemic-cell damage. Med Hypotheses
47:2838
Paschen W, Gissel C, Linden T, Althausen S, Doutheil J
(1998) Activation of gadd153 expression through transient cerebral ischemia: evidence that ischemia causes
endoplasmic reticulum dysfunction. Brain Res Mol
Brain Res 60:11522
Powers WJ, Grubb Jr RL, Darriet D, Raichle ME (1985)
Cerebral blood flow and cerebral metabolic rate of
oxygen requirements for cerebral function and viability
in humans. J Cereb Blood Flow Metab 5:6008
Quistorff B, Chance B, Hunding A (1977) An experimental
model of the Krogh tissue cylinder: two dimensional
quantitation of the oxygen gradient. Adv Exp Med Biol
94:12736
Rajdev S, Hara K, Kokubo Y, Mestril R, Dillmann W,
Weinstein PR, Sharp FR (2000) Mice overexpressing rat
heat shock protein 70 are protected against cerebral
infarction. Ann Neurol 47:78291

Heterogeneity in the penumbra


GJ del Zoppo et al

Ringer TM, Neumann-Haefelin T, Sobel RA, Moseley ME,


Yenari MA (2001) Reversal of early diffusion-weighted
magnetic resonance imaging abnormalities does not
necessarily reflect tissue salvage in experimental cerebral ischemia. Stroke 32:23629
Rosenberg RD, Aird WC (1999) Vascular-bedspecific
hemostasis and hypercoagulable states. N Engl J Med
340:155564
Rosner G, Graf R, Kataoka K, Heiss WD (1986) Selective
functional vulnerability of cortical neurons following
transient MCA-occlusion in the cat. Stroke 17:7682
Ruetzler CA, Furuya K, Takeda H, Hallenbeck JM (2001)
Brain vessels normally undergo cyclic activation and
inactivation: evidence from tumor necrosis factor-alpha,
heme oxygenase-1, and manganese superoxide dismutase immunostaining of vessels and perivascular brain
cells. J Cereb Blood Flow Metab 21:24452
Schroder M, Kaufman RJ (2005) The mammalian unfolded
protein response. Annu Rev Biochem 74:73989
Senger DR, Ledbetter SR, Claffey KP, Papdopoulos-Sergiou
A, Peruzzi CA, Detmar M (1996) Stimulation of
endothelial cell migration by vascular permeability
factor/vascular endothelial growth factor through cooperative mechanisms involving the aVb3 integrin,
osteopontin, and thrombin. Am J Pathol 149:293305
Sharp FR, Lu A, Tang Y, Millhorn DE (2000) Multiple
molecular penumbras after focal cerebral ischemia.
J Cereb Blood Flow Metab 20:101132
Sokoloff L (1999) Energetics of functional activation in
neural tissues. Neurochem Res 24:3219
Somjen GG (2001) Mechanisms of spreading depression
and hypoxic spreading depression-like depolarization.
Physiol Rev 81:106596
Spratt NJ, Howells DW, Donnan GA (2007) Imaging the
penumbra: positron emission tomography fluoromisonidazole. In: The Ischemic Penumbra: Pathophysiology,
Imaging and Therapy (Donnan GA, Baron J-C, Davis SM,
Sharp FR, eds), New York: Informa Healthcare, 14964
Strong AJ (2009) Spreading depolarisations: Tsunamis in
the injured brain. Adv Clin Neurosci Rehab 9:325
Symon L (1980) The relationship between CBF, evoked
potentials and the clinical features in cerebral ischaemia.
Acta Neurol Scand Suppl 78:17590
Symon L, Pasztor E, Branston NJM (1974) The distribution
and density of reduced cerebral blood flow following
acute middle cerebral artery occlusion: an experimental
study by the technique of hydrogen clearance in
baboons. Stroke 5:35564
Tagaya M, Haring H-P, Stuiver I, Wagner S, Abumiya T,
Lucero J, Lee P, Copeland BR, Seiffert D, del Zoppo GJ

(2001) Rapid loss of microvascular integrin expression


during focal brain ischemia reflects neuron injury.
J Cereb Blood Flow Metab 21:83546
Tagaya M, Liu K-F, Copeland B, Seiffert D, Engler R,
Garcia JH, del Zoppo GJ (1997) DNA scission after focal
brain ischemia. Temporal differences in two species.
Stroke 28:124554
Takasawa M, Beech JS, Fryer TD, Hong YT, Hughes JL,
Igase K, Jones PS, Smith R, Aigbirhio FI, Menon DK,
Clark JC, Baron JC (2007) Imaging of brain hypoxia
in permanent and temporary middle cerebral artery
occlusion in the rat using 18F-fluoromisonidazole and
positron emission tomography: a pilot study. J Cereb
Blood Flow Metab 27:67989
Thijs VN, Adami A, Neumann-Haefelin T, Moseley ME,
Albers GW (2002) Clinical and radiological correlates of
reduced cerebral blood flow measured using magnetic
resonance imaging. Arch Neurol 59:2338
Toth G, Albers GW (2009) Use of MRI to estimate the
therapeutic window in acute stroke: is perfusionweighted imaging/diffusion-weighted imaging mismatch
an EPITHET for salvageable ischemic brain tissue? Stroke
40:3335
Wagner S, Tagaya M, Koziol JA, Quaranta V, del Zoppo GJ
(1997) Rapid disruption of an astrocyte interaction with
the extracellular matrix mediated by integrin alpha6beta4 during focal cerebral ischemia/reperfusion. Stroke
28:85865
Welch WJ (1993) Heat shock proteins functioning as molecular chaperones: their roles in normal and stressed
cells. Philos Trans R Soc Lond B Biol Sci 339:32733
Welch WJ, Brown CR (1996) Influence of molecular and
chemical chaperones on protein folding. Cell Stress
Chaperones 1:10915
Yenari MA (2002) Heat shock proteins and neuroprotection. Adv Exp Med Biol 513:28199
Zaro-Weber O, Moeller-Hartmann W, Heiss WD, Sobesky J
(2009) The performance of MRI-based cerebral blood
flow measurements in acute and subacute stroke compared with 15O-water positron emission tomography:
identification of penumbral flow. Stroke 40:241321
Zaro-Weber O, Moeller-Hartmann W, Heiss WD, Sobesky J
(2010) Maps of time to maximum and time to peak for
mismatch definition in clinical stroke studies validated with positron emission tomography. Stroke 41:
281721
Zhan X, Kim C, Sharp FR (2008) Very brief focal ischemia
simulating transient ischemic attacks (TIAs) can injure
brain and induce Hsp70 protein. Brain Res 1234:
18397

1851

Journal of Cerebral Blood Flow & Metabolism (2011) 31, 18361851

Você também pode gostar