You are on page 1of 32

Section 1 Etiology, pathophysiology, and imaging

Chapter
Neuropathology and pathophysiology

1 of stroke
Konstantin-A. Hossmann and Wolf-Dieter Heiss

and fibromuscular dysplasia, but these disorders are


The vascular origin of cerebrovascular characterized by typical locations of the vascular
disease changes. Some arteriopathies are hereditary, such as
All cerebrovascular diseases (CVDs) have their origin CADASIL (cerebral autosomal dominant arteriopa-
in the vessels supplying or draining the brain. There- thy with subcortical infarcts and leukoencephalopa-
fore, the knowledge of pathological changes occurring thy), in some such as cerebral amyloid angiopathy a
in the vessels and in the blood are essential for under- degenerative cause has been suggested. All these vas-
standing the pathophysiology of the various types of cular disorders can cause obstruction, and lead to
CVD and for planning of efficient therapeutic strat- thrombosis and embolizations. Small vessels of the
egies. Changes in the vessel wall lead to obstruction of brain are affected by hyalinosis and fibrosis; this
blood flow, by interacting with blood constituents “small-vessel disease” can cause lacunes and, if wide-
they may cause thrombosis and blockade of blood spread, is the substrate for vascular cognitive impair-
flow in this vessel. In addition to vascular stenosis or ment and vascular dementia.
occlusion at the site of vascular changes, disruption of Atherosclerosis is the most widespread disorder
blood supply and consecutive infarcts can also be leading to death and serious morbidity including
produced by emboli arising from vascular lesions stroke [1]. The basic pathological lesion is the ather-
situated proximally to otherwise healthy branches omatous plaque, and the most commonly affected
located more distal in the arterial tree or from a sites are the aorta, the coronary arteries, the carotid
source located in the heart. At the site of occlusion, artery at its bifurcation, and the basilar artery.
opportunity exists for thrombus to develop in ante- Arteriosclerosis, a more generic term describing
rograde fashion throughout the length of the vessel, hardening and thickening of the arteries, includes as
but this event seems to occur only rarely. additional types Mönkeberg’s sclerosis which is char-
Changes in large arteries supplying the brain, acterized by calcification in the tunica media and
including the aorta, are mainly caused by atheroscler- arteriolosclerosis with proliferative and hyaline
osis. Middle-sized and intracerebral arteries can also changes affecting the arterioles. Atherosclerosis starts
be affected by acute or chronic vascular diseases of at young age, lesions accumulate and grow through-
inflammatory origin due to subacute to chronic infec- out life and become symptomatic and clinically evi-
tions, e.g. tuberculosis and lues, or due to collagen dent when end organs are affected [2].
disorders, e.g. giant cell arteritis, granulomatous Atherosclerosis: atheromatous plaques, most commonly
angiitis of the central nervous system, panarteritis in the aorta, the coronary arteries, the bifurcation of the
nodosa, and even more rarely systemic lupus erythe- carotid artery and the basilar artery.
matosus, Takayasu’s arteritis, Wegener granulomato- The initial lesion of atherosclerosis has been attrib-
sis, rheumatoid arteritis, Sjögren’s syndrome, or uted to “fatty streaks” and the “intimal cell mass.”
Sneddon and Behçet’s disease. In some diseases Those changes already occur in childhood and ado-
affecting the vessels of the brain the etiology and lescence and do not necessarily correspond to the
pathogenesis are still unclear, e.g. moyamoya disease future sites of atherosclerotic plaques. Fatty streaks

Textbook of Stroke Medicine, Second Edition, ed. Michael Brainin and Wolf-Dieter Heiss. Published by Cambridge University 1
Press. © Michael Brainin and Wolf-Dieter Heiss 2014.

Downloaded from https://www.cambridge.org/core. University of Sussex Library, on 17 Apr 2018 at 16:27:57, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781107239340.002
Section 1: Etiology, pathophysiology, and imaging

Figure 1.1. The stages of development of an atherosclerotic plaque. (1) LDL moves into the subendothelium and (2) is oxidized by
macrophages and smooth muscle cells (SMC). (3) Release of growth factors and cytokines (4) attracts additional monocytes. (5) Macrophages
and (6) foam cell accumulation and additional (7) SMC proliferation result in (8) growth of the plaque. (9) Fibrous cap degradation and plaque
rupture (collagenases, elastases). (10) Thrombus formation. IL-1 ¼ interlenkin-1; MCP-1 ¼ Monocyte chemotactic protein-1. (Modified with
permission from Faxon et al. [5].)

are focal areas of intra cellular lipid collection in both larger lipid core and a thin fibrous cap – can lead to
macrophages and smooth muscle cells. Various con- plaque disruption with formation of a thrombus or
cepts have been proposed to explain the progression hematoma or even to total occlusion of the vessel.
of such precursor lesions to definite atherosclerosis During the development of atherosclerosis the entire
[2, 3], most remarkable of which is the response-to- vessel can enlarge or constrict in size [10]. However,
injury hypothesis postulating a cellular and molecular once the plaque covers >40% of the vessel wall, the
response to various atherogenic stimuli in the form of artery no longer enlarges, and the lumen narrows as
an inflammatory repair process [4]. This inflamma- the plaque grows. In vulnerable plaques thrombosis
tion develops concurrently with the accumulation of forming on the disrupted lesion further narrows
minimally oxidized low-density lipoproteins (LDLs) the vessel lumen and can lead to occlusion or be
[5, 6], and stimulates vascular smooth muscle cells the origin of emboli. Less commonly, plaques
(VSMCs), endothelial cells, and macrophages [7], and have reduced collagen and elastin with a thin and
as a result foam cells aggregate with an accumulation weakened arterial wall, resulting in aneurysm forma-
of oxidized LDL. In the further stages of arthero- tion which when ruptured may be the source of
sclerotic plaque development VSMCs migrate, prolif- intracerebral hemorrhage (Figure 1.1).
erate, and synthesize extracellular matrix components
on the luminal side of the vessel wall, forming the Injury hypothesis of progression to atherosclerosis: fatty
streaks (focal areas of intra cellular lipid collection) !
fibrous cap of the atherosclerotic lesion [8]. In this
inflammatory repair process with stimulation of vascular
complex process of growth, progression, and finally
smooth muscle cells ! atheromatous plaque.
rupture of an atherosclerotic plaque a large number of
matrix modulators, inflammatory mediators, growth Thromboembolism: immediately after plaque rupture
factors, and vasoactive substances are involved. The or erosion, subendothelial collagen, the lipid core, and
complex interactions of these many factors are dis- procoagulants such as tissue factor and von Wille-
cussed in the special literature [5–9]. brand factor are exposed to circulating blood. Platelets
The fibrous cap of the atherosclerotic lesion rapidly adhere to the vessel wall through the platelet
covers the deep lipid core with a massive accumula- glycoproteins (GP) Ia/IIa and GP Ib/IX [11] with
tion of extracellular lipids (atheromatous plaque), or subsequent aggregation to this initial monolayer
fibroblasts and extracellular calcifications may con- through linkage with fibrinogen and the exposed GP
tribute to a fibrocalcific lesion. Mediators from inflam- IIb/IIIa on activated platelets. As platelets are a source
2 matory cells at the thinnest portion of the cap surface of nitric oxide (NO), the resulting deficiency of bioac-
of a vulnerable plaque – which is characterized by a tive NO, which is an effective vasodilator, contributes

Downloaded from https://www.cambridge.org/core. University of Sussex Library, on 17 Apr 2018 at 16:27:57, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781107239340.002
Chapter 1: Neuropathology and pathophysiology of stroke

to the progression of thrombosis by augmenting


platelet activation, enhancing VSMC proliferation Types of acute cerebrovascular diseases
and migration, and participating in neovasculariza- Numbers relating to the frequency of the different
tion [12]. The activated platelets also release adeno- types of acute CVD are highly variable depending
sine diphosphate (ADP) and thromboxane A2 with on the source of data. The most reliable numbers
subsequent activation of the clotting cascade. The come from the in-hospital assessment of stroke in
growing thrombus obstructs or even blocks the the Framingham study determining the frequency
blood flow in the vessel. Atherosclerotic thrombi of completed stroke: 60% were caused by athero-
are also the source for embolisms, which are the thrombotic brain infarction, 25.1% by cerebral
primary pathophysiological mechanism of ischemic embolism, 5.4% by subarachnoid hemorrhage,
strokes, especially from carotid artery disease or of 8.3% by intracerebral hemorrhage, and 1.2% by
cardiac origin. undefined diseases. In addition, transient ischemic
attacks (TIAs) accounted for 14.8% of the total
Rupture or erosion of atheromatous plaques ! adhesion
of platelets ! thrombus ! obstruction of blood flow cerebrovascular events [21].
and source of emboli. Ischemic strokes result from a critical reduction of
regional cerebral blood flow (rCBF) lasting beyond a
Small-vessel disease usually affects the arterioles and is critical duration, and are caused by atherothrombotic
associated with hypertension. It is caused by suben- changes of the arteries supplying the brain or by
dothelial accumulation of a pathological protein, the emboli from sources in the heart, the aorta, or the
hyaline, formed from mucopolysaccharides and large arteries. The pathological substrate of ischemic
matrix proteins, which leads to narrowing of the stroke is ischemic infarction of brain tissue, the loca-
lumen or even occlusion of these small vessels. Often tion, extension, and shape of which depend on the
it is associated with fibrosis, which affects not only size of the occluded vessel, the mechanism of arterial
arterioles, but also other small vessels and capillaries obstruction, and the compensatory capacity of the
and venules. Lipohyalinosis also weakens the vessel vascular bed. Occlusion of arteries supplying defined
wall, predisposing for the formation of “miliary brain territories by atherothrombosis or embolization
aneurysms.” Small-vessel disease results in two patho- lead to territorial infarcts of variable size: they may be
logical conditions: status lacunaris (lacunar state) and large – e.g. the whole territory supplied by the middle
status cribrosus (état criblé). Status lacunaris is char- cerebral artery (MCA) – or small, if branches of large
acterized by small irregularly shaped infarcts due to arteries are occluded or if compensatory collateral
occlusion of small vessels; it is the pathological sub- perfusion – e.g. via the circle of Willis or leptomenin-
strate of lacunar strokes and vascular cognitive geal anastomoses – is efficient in reducing the area of
impairment and dementia. In status cribrosus small critically reduced flow (Figure 1.2) [14, 16]. In a
round cavities develop around affected arteries due smaller number of cases infarcts can also develop at
to disturbed supply of oxygen and metabolic sub- the borderzones between vascular territories, when
strate. These “criblures” together with miliary aneur- several large arteries are stenotic and the perfusion
ysms are the sites of vessel rupture causing typical in these “last meadows” cannot be constantly main-
hypertonic intracerebral hemorrhages [13–16]. tained above the critical threshold of morphological
A second type of small-vessel disease is characterized integrity [22]. Borderzone infarctions are a subtype of
by the progressive accumulation of congophilic, βA4 the low-flow or hemodynamically induced infarctions
immunoreactive, amyloid protein in the walls of which are the result of critically reduced cerebral
small- to medium-sized arteries and arterioles. Cere- perfusion pressure in far-downstream brain arteries.
bral amyloid angiopathy is a pathological hallmark The more common low-flow infarctions affect sub-
of Alzheimer’s disease and also occurs in rare genet- cortical structures within a vascular bed with pre-
ically transmitted diseases, e.g. CADASIL and Fabry’s served but marginal irrigation [23]. Lacunar infarcts
disease [17]. For a more detailed discussion of the reflect disease of the vessels penetrating the brain to
etiology and pathophysiology of the various specific supply the capsule, the basal ganglia, thalamus, and
vascular disorders see [18–20]. paramedian regions of the brainstem [24]. Most often
Small-vessel disease: subendothelial accumulation of they are caused by lipohyalinosis of deep arteries 3
hyaline in arterioles. (small-vessel disease); less frequent causes are stenosis

Downloaded from https://www.cambridge.org/core. University of Sussex Library, on 17 Apr 2018 at 16:27:57, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781107239340.002
Section 1: Etiology, pathophysiology, and imaging

Figure 1.2. Various types and sizes of infarcts due to different hemodynamic patterns
a. Total territorial infarct due to defective collateral supply
b. Core infarct, meningeal anastomosis supply peripheral zones
c. Territorial infarct in center of supply area, due to branch occlusion
d. Borderzone infarction in watershed areas due to stenotic lesions in arteries supplying neighboring areas
e. Lacunar infarctions due to small-vessel disease. (Modified with permission from Zülch [14].)

of the MCA stem and microembolization to penetrant infarcts in which varying numbers of blood cells are
arterial territories. Pathologically these lacunes are found within the necrotic tissue. The size can range
defined as small cystic trabeculated scars about from a few petechial bleeds in the gray matter of
5 mm in diameter, but they are more often observed cortex and basal ganglia to large hemorrhages involv-
on magnetic resonance images, where they are ing the cortical and deep hemispheric regions. Hem-
accepted as lacunes up to 1.5 cm diameter. The classic orrhagic transformation frequently appears during
lacunar syndromes include pure motor, pure sensory, the second and third phase of infarct evolution, when
and sensorimotor syndromes, sometimes ataxic hemi- macrophages appear and new blood vessels are
paresis, clumsy hand, dysarthria, and hemichorea/ formed in tissue consisting of neuronal ghosts and
hemiballism, but higher cerebral functions are not proliferating astrocytes. However, the only significant
involved. A new classification of stroke subtypes is difference between “pale” and “red infarcts” is the
mainly oriented on the most likely cause of stroke: intensity and extension of the hemorrhagic compon-
atherosclerosis, small-vessel disease, cardiac source, ent, since in at least two-thirds of all infarcts petechial
or other cause [25]. hemorrhages are microscopically detectable. Macro-
Territorial infarcts are caused by an occlusion of arteries scopically, red infarcts contain multifocal bleedings
supplying defined brain territories by atherothrombosis which are more or less confluent and predominate
or embolizations. in cerebral cortex and basal ganglia, which are richer
Borderzone infarcts develop at the borderzone between in capillaries than the white matter[26]. If the hemor-
vascular territories and are the result of a critically rhages become confluent intrainfarct hematomas
reduced cerebral perfusion pressure (low-flow might develop, and extensive edema may contribute
infarctions). to mass effects and lead to malignant infarction. The
Lacunar infarcts are mainly caused by small-vessel frequency of hemorrhagic infarctions in anatomical
disease. studies ranged from 18% to 42% [27], with a high
4 Hemorrhagic infarctions, i.e. “red infarcts” in contrast incidence (up to 85% of hemorrhagic infarcts) in
to the usual “pale infarcts,” are defined as ischemic cardioembolic stroke [28].

Downloaded from https://www.cambridge.org/core. University of Sussex Library, on 17 Apr 2018 at 16:27:57, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781107239340.002
Chapter 1: Neuropathology and pathophysiology of stroke

Mechanisms for hemorrhagic transformation are thrombolytic treatment of ischemic diseases of the
manifold and vary with regard to the intensity of brain, heart, and other organs [31, 32].
bleeding. Petechial bleeding results from diapedesis Spontaneous ICH occurs predominantly in the
rather than vascular rupture. In severe ischemic tissue deep portions of the cerebral hemispheres (“typical
vascular permeability is increased and endothelial ICH”) [33]. Its most common location is the putamen
tight junctions are ruptured. When blood circulation (35–50% of cases). The subcortical white matter is
is spontaneously or therapeutically restored, blood the second most frequent location (approx. 30%).
can leak out of these damaged vessels. This can also Hemorrhages in the thalamus are found in 10–15%,
happen with fragmentation and distal migration of in the pons in 5–12%, and in the cerebellum in 7%
an embolus (usually of cardiac origin) in the damaged [34]. Most ICHs originate from the rupture of small,
vascular bed, explaining delayed clinical worsening deep arteries with diameters of 50 to 200 μm which
in some cases. For the hemorrhagic transformation are affected by lipohyalinosis due to chronic hyper-
the collateral circulation might also have an impact: tension. These small-vessel changes lead to weakening
in some instances reperfusion via pial networks of the vessel wall and miliary microaneurysm and
may develop with the diminution of peri-ischemic consecutive small local bleedings, which might be
edema at borderzones of cortical infarcts. Risk followed by secondary ruptures of the enlarging
of hemorrhage is significantly increased in large hematoma in a cascade or avalanche fashion [35].
infarcts, with mass effect supporting the importance After active bleeding has started it can continue
of edema for tissue damage and the deleterious for a number of hours with enlargement of hema-
effect of late reperfusion when edema resolves. In toma that is frequently associated with clinical
some instances also the rupture of the vascular wall deterioration [36].
secondary to ischemia-induced endothelial necrosis Putaminal hemorrhages originate from a lateral
might cause an intra-infarct hematoma. Vascular branch of the striate arteries at the posterior angle,
rupture can explain very early hemorrhagic infarcts resulting in an ovoid mass pushing the insular cortex
and early intrainfarct hematoma (between 6 and laterally and displacing or involving the internal
18 hours after stroke), whereas hemorrhagic trans- capsule. From this initial putaminal-claustral loca-
formation usually develops within 48 hours to 2 tion a large hematoma may extend to the internal
weeks. capsule and lateral ventricle, into the corona radiata,
Hemorrhagic infarctions are defined as ischemic infarcts and into the temporal white matter. Putaminal
in which varying amounts of blood cells are found ICHs are considered the typical hypertensive
within the necrotic tissue. They are caused by leakage hemorrhages.
from damaged vessels, due to increased vascular Caudate hemorrhage, a less common form of
permeability in ischemic tissue or vascular rupture bleeding from distal branches of lateral striate arter-
secondary to ischemia ies, occurs in the head of the caudate nucleus. This
Intracerebral hemorrhage (ICH) occurs as a result of bleeding soon connects to the ventricle and usually
bleeding from an arterial source directly into the involves the anterior limb of the internal capsule.
brain parenchyma and accounts for 5–15% of all Thalamic hemorrhages can involve most of this
strokes [29, 30]. Hypertension is the leading risk nucleus and extend into the third ventricle medially
factor, but in addition advanced age, race and also and the posterior limb of the internal capsule laterally.
cigarette smoking, alcohol consumption, and high The hematoma may press on or even extend into the
serum cholesterol levels have been identified. In a midbrain. Larger hematomas often reach the corona
number of instances ICH occurs in the absence of radiata and the parietal white matter.
hypertension, usually in atypical locations. The causes Lobar (white matter) hemorrhages originate at the
include small vascular malformations, vasculitis, cortico-subcortical junction between gray and white
brain tumors, and sympathomimetic drugs (e.g. matter and usually spread along the fiber bundles in
cocaine). ICH may also be caused by cerebral amyloid the parietal and occipital lobes. The hematomas are
angiopathy and rarely damage is elicited by acute close to the cortical surface and usually not in direct
changes in blood pressure, e.g. due to exposure to contact with deep hemisphere structures or the ven-
cold. The occurrence of ICH is also influenced tricular system. As atypical ICHs they are not neces- 5
by the increasing use of antithrombotic and sarily correlated with hypertension.

Downloaded from https://www.cambridge.org/core. University of Sussex Library, on 17 Apr 2018 at 16:27:57, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781107239340.002
Section 1: Etiology, pathophysiology, and imaging

Cerebellar hemorrhages usually originate in the and causes contribute to the development of this dis-
area of the dentate nucleus from rupture of distal order [38]. The incidence of septic CVT has been
branches of the superior cerebellar artery and extend reduced to less than 10% of cases, but septic cavernous
into the hemispheric white matter and into the fourth sinus thrombosis is still a severe, however, rare prob-
ventricle. The pontine tegmentum is often com- lem. Aseptic CVT occurs during puerperium and less
pressed. A variant, the midline hematoma, originates frequently during pregnancy, but may also be related
from the cerebellar vermis, always communicates to use of oral contraceptives. Among the non-
with the fourth ventricle, and frequently extends infectious causes of CVT congenital thrombophilia,
bilaterally into the pontine tegmentum. particularly prothrombin and factor V Leiden gene
Pontine hemorrhages from bleeding of small para- mutations, as well as antithrombin, protein C, and
median basilar perforating branches cause medially protein S deficiencies must be considered. Other con-
placed hematomas involving the basis of the pons. ditions with risk for CVT are malignancies, inflamma-
A unilateral variety results from rupture of distal tory diseases, and systemic lupus erythematosus.
long circumferential branches of the basilar artery. However, in 20–35% of CVT the etiology remains
These hematomas usually communicate with the unknown. The fresh venous thrombus is rich in red
fourth ventricle, and extend laterally and ventrally blood cells and fibrin and poor in platelets. Later on,
into the pons. it is replaced by fibrous tissue, occasionally with reca-
The frequency of recurrent ICHs in hypertensive nalization. The most common location of CVT is the
patients is rather low (6%) [37]. Recurrence rate is superior sagittal sinus and the tributary veins.
higher with poor control of hypertension and also in Whereas some thromboses, particularly of the
hemorrhages due to other causes. In some instances lateral sinus, may have no pathological consequences
multiple simultaneous ICHs may occur, but also in for the brain tissue, occlusion of large cerebral veins
these cases the cause is other than hypertension. usually leads to a venous infarct. These infarcts are
In ICHs, the local accumulation of blood des- located in the cortex and adjacent white matter and
troys the parenchyma, displaces nervous structures, often are hemorrhagic. Thrombosis of the superior
and dissects the tissue. At the bleeding sites fibrin sagittal sinus may lead only to brain edema, but
globes are formed around accumulated platelets. usually causes bilateral hemorrhagic infarcts in both
After hours or days extracellular edema develops at hemispheres. These venous infarcts are different from
the periphery of the hematoma. After 4 to 10 days arterial infarcts: cytotoxic edema is absent or mild,
the red blood cells begin to lyse, granulocytes and vasogenic edema is prominent, and hemorrhagic
thereafter microglial cells arrive, and foamy macro- transformation or bleeding is usual. Despite this
phages are formed, which ingest debris and hemosi- hemorrhagic component heparin is the treatment of
derin. Finally, the astrocytes at the periphery of the choice.
hematoma proliferate and turn into gemistocytes Cerebral venous thrombosis can lead to a venous infarct.
with eosinophylic cytoplasma. When the hematoma Venous infarcts are different from arterial infarcts:
is removed, the astrocytes are replaced by glial cytotoxic edema is absent or mild, vasogenic edema is
fibrils. After that period – extending to months – prominent, and hemorrhagic transformation or bleeding
the residue of the hematoma is a flat cavity with a is usual.
reddish lining resulting from hemosiderin-laden
macrophages [34].
Intracerebral hemorrhage (ICH) occurs as a result of
Cellular pathology of ischemic stroke
bleeding from an arterial source directly into the brain Acute occlusion of a major brain artery causes a
parenchyma, predominantly in the deep portions of stereotyped sequel of cellular alterations which evolve
the cerebral hemispheres (typical ICH). Hypertension over a protracted period of time and which depend on
is the leading risk factor, and the most common location the topography, severity, and duration of ischemia
is the putamen. [39]. The most sensitive brain cells are neurons,
Cerebral venous thrombosis (CVT) can develop from followed – in this order – by oligodendrocytes, astro-
many causes and due to predisposing conditions. cytes, and vascular cells. The most vulnerable brain
6 CVT is often multifactorial, when various risk factors regions are hippocampal subfield CA1, neocortical

Downloaded from https://www.cambridge.org/core. University of Sussex Library, on 17 Apr 2018 at 16:27:57, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781107239340.002
Chapter 1: Neuropathology and pathophysiology of stroke

layers 3, 5, and 6, the outer segment of striate nucleus, swelling or shrinkage, the cytoplasm exhibiting
and the Purkinje and basket cell layers of cerebellar microvacuolation (MV) which ultrastructurally has
cortex. If blood flow decreases below the threshold of been associated with mitochondrial swelling [40].
energy metabolism, the primary pathology is necrosis These changes are potentially reversible if blood flow
of all cell elements, resulting in ischemic brain infarct. is restored before mitochondrial membranes begin
If ischemia is not severe enough to cause primary to rupture. One to two hours after the onset of ische-
energy failure, or if it is of so short duration that mia, neurons undergo irreversible necrotic alterations
energy metabolism recovers after reperfusion, a (red neuron or ischemic cell change). In conven-
delayed type of cell death may evolve which exhibits tional hematoxylin-eosin-stained brain sections such
the morphological characteristics of necrosis, apop- neurons are characterized by intensively stained eosi-
tosis, or a combination of both. In the following, nophilic cytoplasma, formation of triangular nuclear
primary and delayed cell death will be described pyknosis, and direct contact with swollen astrocytes
separately. (Figure 1.3). Electron-microscopically mitochondria
exhibit flocculent densities which represent denatu-
rated mitochondrial proteins. Ischemic cell change
Primary ischemic cell death must be distinguished from artifactual dark neurons
In the core of the territory of an occluded brain which stain with all (acid or basic) dyes and are not
artery the earliest sign of cellular injury is neuronal surrounded by swollen astrocytes [41].

Light-microscopical characteristics of rat infarction Figure 1.3. Light-microscopical


Acute ischemic changes evolution of neuronal changes after
experimental middle cerebral occlusion.
Control swelling shrinkage (Modified with permission from Garcia
et al. [126].)

sham surgery 4 hours 2 hours


Necrotic changes
red neuron ghost neuron Dark neuron artifact

1 day 3 days sham surgery 7

Downloaded from https://www.cambridge.org/core. University of Sussex Library, on 17 Apr 2018 at 16:27:57, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781107239340.002
Section 1: Etiology, pathophysiology, and imaging

Figure 1.4. Transformation of acute


ischemic alterations into cystic infarct.
Note pronounced inflammatory reaction
prior to tissue cavitation. PMN ¼
polymorphonuclear leukocyte. (Modified
with permission from Petito [39].)

With ongoing ischemia, neurons gradually loose global ischemia [42]. In focal ischemia delayed neur-
their stainability with hematoxylin, they become onal death may occur in the periphery of cortical
mildly eosinophilic, and, after 2–4 days, transform infarcts or in regions which have been reperfused
to ghost cells with hardly detectable pale outline. before ischemic energy failure becomes irreversible.
Interestingly, neurons with ischemic cell change are Cell death is also observed in distant brain regions,
mainly located in the periphery and ghost cells in the notably in the substantia nigra and thalamus.
center of the ischemic territory, which suggests that The morphological appearance of neurons during
manifestation of ischemic cell change requires some the interval between ischemia and the manifestation
residual or restored blood flow whereas ghost cells of delayed cell death exhibits a continuum that
may evolve in the absence of flow [39]. ranges from necrosis to apoptosis with all possible
Primary ischemic cell death induced by focal combinations of cytoplasmic and nuclear morph-
ischemia is associated with reactive and secondary ology that are characteristic for the two types of cell
changes. The most prominent alteration during the death [43]. In its pure form, necrosis combines
initial 1–2 hours is perivascular and perineuronal karyorrhexis with massive swelling of endoplasmic
astrocytic swelling, after 4–6 hours the blood–brain reticulum and mitochondria, whereas in apoptosis
barrier breaks down, resulting in the formation of mitochondria remain intact and nuclear fragmenta-
vasogenic edema, after 1–2 days inflammatory cells tion with condensation of nuclear chromatin gives
accumulate throughout the ischemic infarct, and way to the development of apoptotic bodies.
within 1.5–3 months cystic transformation of the A frequently used histochemical method for the
necrotic tissue occurs together with the development visualization of apoptosis is terminal deoxyribonu-
of a peri-infarct astroglial scar (Figure 1.4) cleotidyl transferase (TdT)-mediated dUTP-biotin
nick-end labeling (TUNEL assay), which detects
DNA strand breaks. However, as this method may
Delayed neuronal death also stain necrotic neurons, a clear differentiation is
The prototype of delayed cell death is the slowly not possible [44].
8 progressing injury of pyramidal neurons in the CA1 A consistent ultrastructural finding in neurons
sector of the hippocampus after a brief episode of undergoing delayed cell death is disaggregation of

Downloaded from https://www.cambridge.org/core. University of Sussex Library, on 17 Apr 2018 at 16:27:57, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781107239340.002
Chapter 1: Neuropathology and pathophysiology of stroke

ribosomes, which reflects the inhibition of protein multiple regions. In experimental stroke research, this
synthesis at the initiation step of translation [45]. situation is reflected by the preferential use of MCA
Light-microscopically, this change is equivalent to occlusion models.
tigrolysis, visible in Nissl-stained material. Disturb- Transorbital middle cerebral artery occlusion: this
ances of protein synthesis and the associated endo- model was introduced in the 1970s for the production
plasmic reticulum (ER) stress are also responsible for of stroke in monkeys [48] and later modified for use
cytosolic protein aggregation and the formation of in cats, dogs, rabbits, and even rats. The procedure is
stress granules [46]. In the hippocampus, stacks technically demanding and requires microsurgical
of accumulated ER may become visible but in other skills. The advantage of this approach is the possibility
areas this is not a prominent finding. to expose the MCA at its origin from the internal
carotid artery without retracting parts of the brain.
Vascular occlusion can thus be performed without the
Pathology of the neurovascular unit risk of brain trauma. On the other hand, removal
The classical pathology of ischemic injury differenti- of the eyeball is invasive and may evoke functional
ates between the sensitivity of the various cell types disturbances which should not be ignored. Surgery
of brain parenchyma with the neurons as the most may also cause generalized vasospasm which may
vulnerable elements. The molecular analysis of injury interfere with the collateral circulation and, hence,
evolution, however, suggests that ischemia initiates a induce variations in infarct size. The procedure there-
coordinated multi-compartmental response of brain fore requires extensive training before reproducible
cells and vessels, also referred to as the neurovascular results can be expected.
unit [47]. This unit includes microvessels (endothelial The occlusion of the MCA at its origin interrupts
cells, basal lamina matrix, astrocytic endfeet, peri- blood flow to the total vascular territory, including
cytes, and circulating blood elements), the cell body the basal ganglia which are supplied by the lenticulo-
and main processes of astrocytes, the nearby neurons striate arteries. These MCA branches are end-arteries
together with their axons, and supporting cells, which in contrast to the cortical branches do not form
notably microglia and oligodendrocytes. It provides collaterals with the adjacent vascular territories. As a
the framework for the bi-directional communication consequence, the basal ganglia are consistently part of
between neuron and supplying microvessel. Under the infarct core whereas the cerebral cortex exhibits
physiological conditions, the most prominent func- a gradient of blood flow which decreases from the
tion is the neurovascular coupling for maintaining peripheral towards the central parts of the vascular
adequate supply of brain nutrients and clearance of territory. Depending on the steepness of this gradient,
waste products. Pathophysiological disturbances of a cortical core region with the lowest flow values
microcirculation, conversely, provoke coincidental in the lower temporal cortex is surrounded by a
microvessel–neuron responses, possibly mediated variably sized penumbra which may extend up to
by alterations in the matrix of the vascular and non- the parasagittal cortex.
vascular compartments of the ischemic territory. Transcranial occlusion of the middle cerebral
Severe ischemia induces primary cell death due to artery: post- or retro-orbital transcranial approaches
necrosis of all cell elements. Not so severe or short-term for MCA occlusion are mainly used in rats and mice
ischemia induces delayed cell death with necrosis, because in these species the main stem of the artery
apoptosis, or a combination of both. The neurovascular appears on the cortical surface rather close to its
unit provides the conceptual framework for the origin from the internal carotid artery [49]. In con-
propagation of injury from microvessels to neurons. trast to transorbital MCA occlusion, transcranial
models do not produce ischemic injury in the basal
ganglia because the lenticulostriate branches origin-
Animal models of stroke ate proximal to the occlusion site. Infarcts, therefore,
According to the Framingham study, 65% of strokes are mainly located in the temporo-parietal cortex with
that result from vascular occlusion present lesions in a gradient of declining flow values from the periph-
the territory of the MCA, 2% in the anterior and 9% eral to the central parts of the vascular territory.
in the posterior cerebral artery territories; the rest is Filament occlusion of the middle cerebral artery: 9
located in brainstem, cerebellum, in watershed, or the presently most widely used procedure for MCA

Downloaded from https://www.cambridge.org/core. University of Sussex Library, on 17 Apr 2018 at 16:27:57, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781107239340.002
Section 1: Etiology, pathophysiology, and imaging

occlusion in rats and mice is the intraluminal filament brain infarcts [53]. The most reliable procedure for
occlusion technique, first described by Koizumi et al. clot preparation is thrombin-induced clotting of auto-
[50]. A nylon suture with an acryl-thickened tip is logous blood within calibrated tubings, which results
inserted into the common carotid artery and ortho- in cylindrical clots that can be dissected in segments
gradely advanced, until the tip is located at the origin of equal length. Selection of either fibrin-rich (white)
of the MCA. Modifications of the original technique or fibrin-poor (red) segments influences the speed
include different thread types for isolated or com- of spontaneous reperfusion and results in different
bined vascular occlusion, adjustments of the tip size outcome. Clots can also be produced in situ by
to the weight of the animal, poly-L-lysine coating of microinjection of thrombin [54] or photochemically
the tip to prevent incomplete MCA occlusion, or the by ultraviolet illumination of the MCA following
use of guide-sheaths to allow remote manipulation of injection of rose Bengal [55].
the thread for occlusion during polygraphic record- The main application of clot embolism is for the
ings or magnetic resonance imaging. investigation of experimental thrombolysis. The drug
The placement of the suture at the origin of the most widely used is human recombinant tissue plas-
MCA obstructs blood supply to the total MCA sup- minogen activator (rtPA) but the dose required in
plied territory, including the basal ganglia. It may also animals is much higher than in humans, which must
reduce blood flow in the anterior and posterior cere- be remembered when possible side-effects such as
bral arteries, particularly when the common carotid rtPA toxicity are investigated. The hemodynamic
artery is ligated to facilitate the insertion of the thread. effect, in contrast, is similar despite the higher dose
As this minimizes collateral blood supply from these and adequately reproduces the slowly progressing
territories, infarcts are very large and produce massive recanalization observed under clinical conditions.
ischemic brain edema with a high mortality when A recent development of clinical stroke treatment
experiments last for more than a few hours. For this and possibly the central challenge for future animal
reason, threads are frequently withdrawn 1–2 hours research is interventional thrombectomy [56]. The
following insertion. The resulting reperfusion sal- animal most widely used for this research is the swine
vages the peripheral parts of the MCA territory, and but as in this species the carotid access to the anterior
infarcts become smaller [51]. However, the pathophy- cerebral vasculature is impeded by a rete mirabile, clot
siology of transient MCA occlusion differs basically embolism and retrieval is carried out via the internal
from that of the clinically more relevant permanent maxillary or lingual artery [57]. Angiographic studies
occlusion models, and neither the mechanisms of confirm that clot retrieval using either aspiration or
infarct evolution nor the pharmacological responsive- removable stent devices results in immediate recana-
ness of the resulting lesions replicate that of clinical lization but a detailed pathophysiological analysis of
stroke [52]. post-ischemic reperfusion is not yet available. It is,
Transient filament occlusion is also an inappro- therefore, premature to speculate whether this treat-
priate model for the investigation of spontaneous or ment and its effect on post-ischemic recovery can also
thrombolysis-induced reperfusion. Withdrawal of the be replicated in smaller animals by technically simpler
intraluminal thread induces instantaneous reperfu- mechanical occlusion models, such as transient fila-
sion whereas spontaneous or thrombolysis-induced ment occlusion.
recanalization results in slowly progressing recir- Various procedures for artery occlusion models, mostly
culation. As post-ischemic recovery is greatly influ- middle cerebral artery occlusion models, were developed
enced by the dynamics of reperfusion, outcome and to study focal ischemia in animals.
pharmacological responsiveness of transient filament
occlusion is distinct from most clinical situations of
reversible ischemia, where the onset of reperfusion is Hemodynamics of stroke
much less abrupt.
Clot embolism of middle cerebral artery: MCA Normal regulation of blood flow
embolism with autologous blood clots is a clinically In the intact brain, CBF is tightly coupled to the
highly relevant but also inherently variable stroke metabolic requirements of tissue (metabolic regula-
10 model which requires careful preparation and place- tion) but the flow rate remains essentially constant
ment of standardized clots to induce reproducible over a wide range of blood pressures (autoregulation).

Downloaded from https://www.cambridge.org/core. University of Sussex Library, on 17 Apr 2018 at 16:27:57, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781107239340.002
Chapter 1: Neuropathology and pathophysiology of stroke

An important requirement for metabolic regula- brain perfusion pressure cannot be compensated by
tion is the responsiveness of blood vessels to carbon further vasorelaxation whereas an increase may shift
dioxide (CO2 reactivity), which can be tested by the the local perfusion pressure into the autoregulatory
application of carbonic anhydrase inhibitors or CO2 range and cause vasoconstriction. An alternative
ventilation. Under physiological conditions, blood explanation is “false autoregulation” due to brain
flow doubles when CO2 rises by about 30 mmHg, edema which causes an increase in local tissue pres-
and is reduced by one-third when CO2 declines by sure that precludes a rise of the actual tissue perfusion
15 mmHg. The vascular response to CO2 depends pressure. Failure of cerebral autoregulation can be
mainly on the changes of extracellular pH but it is demonstrated in such instances by dehydrating the
also modulated by other factors such as prostanoids, brain in order to reduce brain edema.
NO, and neurogenic influences. After transient ischemia, vasorelaxation persists
Autoregulation of CBF is the remarkable capacity for some time, which explains the phenomenon of
of the vascular system to adjust its resistance in such a post-ischemic hyperemia or luxury perfusion. During
way that blood flow is kept constant over a wide range luxury perfusion, oxygen supply exceeds oxygen require-
of cerebral perfusion pressures (80–150 mmHg). The ments of the tissue, as reflected by the appearance of
range of autoregulation is shifted to the right, i.e. to red venous blood. With the cessation of tissue acido-
higher values, in patients with hypertension and to sis, vascular tone returns, and blood flow declines to
the left during hypercarbia. or below normal. At longer recirculation times auto-
The mechanism of autoregulation is complex [58]. regulation – but not CO2 reactivity – may recover,
The dominating factor is a pressure-sensitive direct resulting in persisting failure of metabolic regulation.
myogenic response initiated by the activation of This is one of the reasons why primary post-ischemic
stretch-sensitive cation channels of vascular smooth recovery may be followed by delayed post-ischemic
muscle. In addition, a flow-sensitive indirect smooth hypoxia and secondary metabolic failure [60].
muscle response is initiated by changes in the shear Disturbances of flow regulation through ischemia:
stress of endothelial cells, which result in activation of tissue acidosis leads to vasorelaxation, CO2 reactivity is
various signal transduction pathways. Other influ- abolished or even reversed, and autoregulation is
ences are mediated by metabolic and neurogenic impaired.
factors but these may be secondary effects and are of
lesser significance.
Metabolic regulation: cerebral blood flow is coupled Disturbances of microcirculation
to metabolic requirements of tissue by a vascular With the increasing understanding of the pathobiol-
response to changes in CO2. Autoregulation: cerebral ogy of the neurovascular unit, microcirculatory
blood flow is kept constant over a wide range disturbances are recognized to contribute to the evo-
of cerebral perfusion pressures. lution of ischemic brain injury [61]. Such disturb-
ances develop at the capillary level within the first
hour of focal ischemia and may persist even after full
Disturbances of flow regulation reversal of vascular occlusion (incomplete microcir-
Focal cerebral ischemia is associated with tissue acid- culatory reperfusion). The dominating pathology is
osis which leads to vasoparalysis and, in consequence, the narrowing of the capillary lumen, induced by
to a severe disturbance of the regulation of blood flow constriction of pericytes and swelling of pericapillary
[59]. In the center of the ischemic territory, CO2 astrocytic endfeet. The capillaries are filled with
reactivity is abolished or even reversed, i.e. blood flow aggregated red blood cells, leukocytes, and fibrin/
may decrease with increasing arterial pCO2. This platelet deposits, the high viscosity of which adds to
paradoxical “steal” effect has been attributed to the the increased vascular resistance of the reduced
rerouting of blood to adjacent non-ischemic brain capillary lumen.
regions in which CO2 reactivity remains intact. The mechanism of microcirculatory impairment
Stroke also impairs autoregulation but the dis- is multifactorial. Pericytes constrict in response to the
turbance is more severe with decreasing rather than generation of reactive oxygen species (ROS), swelling
with increasing blood pressure. This is explained by of astrocytic endfeet is due to cytotoxic brain edema, 11
the fact that in the ischemic tissue a decrease of local and leukocyte adhesion to the vessel wall is part of the

Downloaded from https://www.cambridge.org/core. University of Sussex Library, on 17 Apr 2018 at 16:27:57, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781107239340.002
Section 1: Etiology, pathophysiology, and imaging

inflammatory response mediated by the generation of not recommend such manipulations for the treatment
chemoattractants, cytokines, and chemokines. Finally, of stroke.
the activation of proteolytic enzymes contributes “Steal”: decrease in focal blood flow when blood is
to the dismantlement of basal lamina and results in diverted from one brain region to another by
damage of the blood–brain barrier, an increase anastomotic channels; “inverse steal” if that results in an
in interstitial tissue pressure, and the risk of hemor- improvement in flow.
rhagic transformation.
The impairment of microcirculation is equivalent
to a reduction of nutritional blood flow. During per- The concept of ischemic penumbra
manent vascular occlusion it aggravates the effect of
primary ischemia, particularly in the borderzone of
Energy requirements of brain tissue
the infarct, and after transient vascular occlusion it The energy demand of the central nervous tissue is
prevents adequate reoxygenation despite recanaliza- very high and therefore sufficient blood supply to the
tion of the supplying artery. It is still unresolved to brain must be maintained constantly. A normal adult
what extent microcirculatory impairment contributes male’s brain containing about 130 billion neurons
to or originates from ischemic injury but there is (21.5 billion in the neocortex) [65] comprises only
general consent that microvascular protection is a 2% of total body mass, yet consumes at rest approxi-
requirement for successful stroke treatment [62]. mately 20% of the body’s total basal oxygen consump-
tion supplied by 16% of the cardiac blood output. The
Focal brain ischemia is aggravated by microcirculatory
disturbances which may persist despite recanalization.
brain’s oxygen consumption is almost entirely for the
oxidative metabolism of glucose, which in normal
physiological conditions is the almost exclusive sub-
Anastomotic steal phenomena strate for the brain’s energy metabolism (Table 1.1)
The brain is protected against focal disturbances of [66]. Glucose metabolized in neuronal cell bodies is
blood flow by the collateral circulation, which pro- mainly to support cellular vegetative and house-
vides a subsidiary network of vascular channels when keeping functions, e.g. axonal transport, biosynthesis
principal conduits fail [63]. However, the connection of nucleic acids, proteins, and lipids, as well as other
of ischemic and non-ischemic vascular territories by energy-consuming processes not related directly to
anastomotic channels may divert blood from one the generation of action potentials. Therefore, the rate
brain region to another, depending on the magnitude of glucose consumption of neuronal cell bodies is
and direction of the blood pressure gradients across essentially unaffected by neuronal functional acti-
the anastomotic connections (for review see Tode and vation. Increases in glucose consumption (and
McGraw [64]). The associated change of regional regional blood flow) evoked by functional activation
blood flow is called “steal” if it results in a decrease in are confined to synapse-rich regions, i.e. the neuropil,
flow, or “inverse steal” if it results in an improvement which contains axonal terminals, dendritic processes,
in flow. Inverse steal has also been referred to as the and also the astrocytic processes that envelop the
Robin Hood syndrome in analogy to the legendary synapses. The magnitudes of these increases are lin-
hero who took from the rich and gave to the poor. early related to the frequency of action potentials in
Steals are not limited to a particular vascular ter-
ritory and may affect both the extra- and intracerebral Table 1.1. Cerebral blood flow (CBF), oxygen utilization
(CMRO2), and metabolic rates of glucose (CMRGIc) in man
circulation. Examples of extracerebral steals are the (approximated values)
subclavian, the occipital-vertebral, and the ophthal-
mic steal syndrome. Intracerebral steal occurs across Cortex White Global
matter
collateral pathways of brain, notably the circle of
Willis and Heubner’s network of pial anastomoses. CBF (ml/100 g/min) 65 21 47
The pathophysiological importance of steal has been CMRO2 230 80 160
disputed but as it depends on the individual hemody- (μmol/100 g/min)
namic situation it may explain unintended effects
CMRGlc 40 20 32
12 when flow is manipulated by alterations of arterial
(μmol/100 g/min)
pCO2 or vasoactive drugs. Most authors, therefore, do

Downloaded from https://www.cambridge.org/core. University of Sussex Library, on 17 Apr 2018 at 16:27:57, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781107239340.002
Chapter 1: Neuropathology and pathophysiology of stroke

(a) (b)
Glutamate-releasing
Astrocyte Capillary
presynaptic terminal

Glucose
34% postsynaptic ion fluxes

Glycolysis
Lactate

Oxidative Glucose 10% neuronal resting


phosphorylation potential
Gln
Glu Gln
3% glial resting potential
+
Glu K 3% glutamate recycling
EAAT Na
+ Na+/K+-ATPase 3% presynaptic Ca2+
+
Na

47% action potential propagation


Postsynaptic site lonotropic glutamate receptor
NMDAR

Figure 1.5. (a) Schematic representation of the mechanism for glutamate-induced glycolysis in astrocytes during physiological activation
[127]. (b). Distribution of energy expenditure in rat cortex at a mean spike rate of 4 Hz: most energy is required for activity, only 13% is
used for maintenance of resting potentials of neurons and glial cells [69,128]. EAAT ¼ excitatory amino-acid transporter; NMDAR ¼ N-methyl-
D-aspartate receptor.

the afferent pathways, and increases in the projection


zones occur regardless of whether the pathway is
Viability thresholds of brain ischemia
excitatory or inhibitory. Energy requirements of func- The different amounts of energy required for the
tional activation are due mostly to stimulation of the generation of membrane potential and the propaga-
Na+/K+-ATPase activity to restore the ionic gradients tion of electrical activity are reflected by different
across the cell membrane and the membrane poten- thresholds of oxygen consumption and blood
tials following spike activity, and are rather high com- flow that must be maintained to preserve neuronal
pared to the basal energy demands of neuronal cell function and morphological integrity. Flow values
bodies (Figure 1.5) [67]. below normal but above the threshold of neuronal
In excitatory glutamatergic neurons, which function are referred to as “benign oligemia.”
account for 80% of the neurons in the mammalian The flow range between the thresholds of neuronal
cortex, glucose utilization during activation is medi- function and morphological integrity is called the
ated by astrocytes which by anaerobic glycolysis pro- “ischemic penumbra” [71]. It is characterized by
vide lactate to the neurons where it is used for the preservation of membrane polarization and the
oxidative metabolism [68]. Overall, 87% of the total potential of functional recovery without morpho-
energy consumed is required for signaling, mainly logical damage, provided that local blood flow can
action potential propagation and postsynaptic ion be re-established [72, 73]. The “infarct core” is the
fluxes, and only 13% is expended in maintaining area in which blood flow declines below the thresh-
membrane resting potential (Figure 1.5) [69]. old of morphological integrity and in which tissue
The mechanisms by which neurotransmitters necrosis evolves.
other than glutamate influence blood flow and energy According to the classical concept of viability
metabolism in the brain are still not understood [70]. thresholds, functional activity – reflected by the
amplitudes of spontaneous and evoked electrical
A normal adult male’s brain comprises only 2% of total
activity – begins to decline at flow values below 50%
body mass, yet consumes at rest approximately 20% of
the body’s total basal oxygen consumption. Glucose is of control and is completely suppressed at about 30%
the almost exclusive substrate for the brain’s energy of control [71]. In awake monkeys these values cor-
metabolism; 87% of the total energy consumed is respond to the progression of neurological injury
required for signaling, mainly action potential from mild paresis at 22 ml/100 g/min to complete 13
propagation and postsynaptic ion fluxes. paralysis at 8 ml/100 g/min. Morphological damage

Downloaded from https://www.cambridge.org/core. University of Sussex Library, on 17 Apr 2018 at 16:27:57, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781107239340.002
Section 1: Etiology, pathophysiology, and imaging

Figure 1.6. Diagrammatic


representation of viability thresholds
of focal brain ischemia.
EEG ¼ electroencephalogram; OEF ¼
oxygen extraction fraction; SEP ¼
somatosensory evoked potential.

evolves as soon as cell membranes depolarize (“ter- varying degree and duration it was possible to con-
minal” depolarization) and occurs at flow values struct a discriminant curve representing the worst
below 15–20% of control. Biochemically, functional possible constellation of residual blood flow and dur-
suppression is associated with the inhibition of pro- ation of ischemia still permitting neuronal recovery
tein synthesis at about 50% and the development of (Figure 1.7). These results broaden the concept of the
lactacidosis at 30–40% of control, whereas membrane ischemic penumbra: the tissue fate – potential of
depolarization and morphological injury correspond recovery or irreversible damage – is determined not
to the breakdown of energy metabolism and the loss only by the level of residual flow but also by the
of adenosine triphosphate (ATP) at about 18% of duration of the flow disturbance. Each level of
control (Figure 1.6). decreased flow can, on average, be tolerated for a
A more detailed picture of the dynamics of injury defined period of time; flow between 17 and 20 ml/
evolution is obtained by the simultaneous recording 100 g/min can be tolerated for prolonged but yet
of local blood flow and spontaneous unit activity of undefined periods of time. As a rule used in many
cortical neurons [74]. According to these measure- experimental models, flow rates of 12 ml/100 g/min
ments, unit activity disappeared at a mean value of lasting for 2–3 hours lead to large infarcts, but indi-
18 ml/100 g/min but the large variability of the func- vidual cells may become necrotic after shorter periods
tional thresholds of individual neurons (6–22 ml/ of time and at higher levels of residual flow.
100 g/min) indicates differential vulnerability even The ischemic penumbra is the range of perfusion
within small cortical sectors. This explains the gradual between the flow threshold for preservation of function
development of neurological deficits, which may be and the flow threshold for preservation of morphological
related to differences in single cell activity with regu- integrity. It is characterized by the potential for
lar or irregular discharges at flow levels above the functional recovery without morphological damage.
threshold of membrane failure.
Whereas neuronal function is impaired immedi-
ately when flow drops below the threshold, the devel- Imaging of penumbra
opment of irreversible morphological damage is time Based on the threshold concept of brain ischemia, the
14 dependent. Based on recordings from a considerable penumbra can be imaged on quantitative flow maps
number of neurons during and after ischemia of using empirically established flow thresholds. A more

Downloaded from https://www.cambridge.org/core. University of Sussex Library, on 17 Apr 2018 at 16:27:57, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781107239340.002
Chapter 1: Neuropathology and pathophysiology of stroke

Figure 1.7. (A) Activity of a single neuron during graded ischemia before, during, and after reversible MCA occlusion. (B) Recovery of neuronal
function after a limited period of ischemia. (C) 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 (upper dashed line) and returns
when flow is raised again above this threshold. The fate of a single cell depends on the duration for which CBF is impaired below a certain level.
The solid line separates structurally damaged from functionally impaired, but morphologically intact tissue, the “penumbra.” The upper dashed 15
line distinguishes viable from functionally impaired tissue. EP ¼ evoked potentials. (Modified with permission from Heiss and Rosner [129]).

Downloaded from https://www.cambridge.org/core. University of Sussex Library, on 17 Apr 2018 at 16:27:57, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781107239340.002
Section 1: Etiology, pathophysiology, and imaging

Figure 1.8. Biochemical imaging of


infarct core, penumbra, and benign
oligemia after experimental middle
cerebral artery occlusion. The core is
identified by ATP depletion, the
penumbra by the mismatch between the
suppression of protein synthesis and ATP
depletion (top) or by the mismatch
between tissue acidosis and ATP
(bottom), and benign oligemia by the
reduction of blood flow in the absence of
biochemical alterations. (Modified
with permission from Hossman and
Mies [130]).

precise approach is the imaging of threshold- with the biochemically characterized penumbra for
dependent biochemical disturbances to demarcate ADC values between 90% and 77% of control [80].
the mismatch between disturbances which occur only Recently MR stroke imaging has been performed by
in the infarct core and others which also affect the combining PWI, DWI, and pH-weighted imaging
penumbra (Figure 1.8) [75]. Under experimental con- (pHWI) [81]. The mismatch between DWI and
ditions the most reliable way to localize the infarct core pHWI detects the penumbra, and that between PWI
is the loss of ATP on bioluminescent images of tissue and pHWI the area of benign oligemia, i.e. a region in
ATP content. A biochemical marker of core plus pen- which flow reduction is not severe enough to cause
umbra is tissue acidosis or the inhibition of protein metabolic disturbances. Diffusion kurtosis imaging
synthesis. The penumbra is the difference between the (DKI), an extension of diffusion imaging, demarcates
respective lesion areas. The reliability of this approach the regions with structural damage that cannot be
is supported by the precise colocalization of gene tran- salvaged upon reperfusion [82].
scripts that are selectively expressed in the penumbra, Finally, new developments in MR molecular
such as the stress protein hsp70, or the documentation imaging are of increasing interest for stroke research
of the gradual disappearance of the penumbra with [83]. These methods make use of contrast-enhanced
increasing ischemia time [76]. probes that trace gene transcription or of intracellular
Non-invasive imaging of the penumbra is possible conjugates that reflect the metabolic status and/or
using positron emission tomography (PET) or mag- bind to stroke markers. The number of molecules that
netic resonance imaging (MRI). Widely used PET can be identified by these methods rapidly expands
parameters are the increase in oxygen extraction or and greatly facilitates the regional analysis of stroke
the mismatch between reduced blood flow and the injury.
preservation of vitality markers, such as flumazenil Non-invasive imaging of the penumbra is possible using
binding to central benzodiazepine receptors [77]. An positron emission tomography (PET) or magnetic
alternative PET approach is the use of hypoxia resonance imaging (MRI).
markers such as 18F-misonidazole (F-MISO), which
is trapped in viable hypoxic but not in normoxic or
necrotic tissue [78]. Mechanisms of infarct expansion
The best-established MRI approach for penumbra With the advent of non-invasive imaging evidence has
imaging is the calculation of mismatch maps between been provided that brain infarcts grow (Figure 1.9).
the signal intensities of perfusion (PWI) and diffusion- This growth is not due to the progression of ischemia
weighted images (DWI), but its reliability has been because the activation of collateral blood supply and
questioned [79]. An alternative method is quanti- spontaneous thrombolysis tend to improve blood
16 tative mapping of the apparent diffusion coefficient flow over time. Infarct progression can be differenti-
(ADC) of water, which reveals a robust correlation ated into three phases. During the acute phase tissue

Downloaded from https://www.cambridge.org/core. University of Sussex Library, on 17 Apr 2018 at 16:27:57, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781107239340.002
Chapter 1: Neuropathology and pathophysiology of stroke

Figure 1.9. Relationship between peri-


infarct spreading depressions (above) and
infarct growth (below) during permanent
focal brain ischemia induced by occlusion
of the middle cerebral artery in rat. The
effect of spreading depressions on
electrical brain activity (EEG) and blood
flow (LDF) are monitored by DC recording
of the cortical steady potential, and infarct
growth by MR imaging of the apparent
diffusion coefficient (ADC) of brain water.
(Modified with permission from
Hossmann [131,132]).

injury is the direct consequence of the ischemia- animals the infarct core may be heterogeneous with
induced energy failure and the resulting terminal multiple mini-cores surrounded by multiple mini-
depolarization of cell membranes. At flow values penumbras but these lesions also expand and eventu-
below the threshold of energy metabolism this injury ally progress to a homogeneous defect with a similar
is established within a few minutes after the onset of time course [84].
ischemia. During the subsequent subacute phase, the Brain infarcts evolve in three phases:
infarct core expands into the peri-infarct penumbra
 acute phase, within a few minutes after the onset of
until, after 4–6 hours, core and penumbra merge. The ischemia; terminal depolarization of cell
reasons for this expansion are peri-infarct spreading membranes;
depressions and a multitude of cell biological disturb-  subacute phase, within 4–6 hours; spreading
ances, collectively referred to as molecular cell injury. depression and molecular cell injury, the infarct core
Moreover, a delayed phase of injury evolves which expands into the peri-infarct penumbra;
may last for several days or even weeks. During this  delayed phase, several days to weeks; vasogenic
phase secondary phenomena such as vasogenic edema, inflammation, and possibly programmed
edema, inflammation, and possibly programmed cell cell death.
death may contribute to a further progression of In the following, the most important mediators of
injury. infarct progression will be discussed.
The largest increment of infarct volume occurs
during the subacute phase in which the infarct core
expands into the penumbra. Using multiparametric Peri-infarct spreading depression
imaging techniques for the differentiation between A functional disturbance contributing to the growth
core and penumbra, evidence could be provided that of the infarct core into the penumbra zone is the
in small rodents submitted to permanent occlusion of generation of peri-infarct spreading depression-like
the MCA at its origin, the penumbra equals the depolarizations (Figure 1.9) [85]. These depolariza-
volume of the infarct core at 1 hour, but after 3 hours tions are initiated at the border of the infarct core
more than 50% and between 6 and 8 hours almost all and spread over the entire ipsilateral hemisphere.
of the penumbra has disappeared and is now part of During spreading depression the metabolic rate of 17
the irreversibly damaged infarct core [76]. In larger the tissue markedly increases in response to the

Downloaded from https://www.cambridge.org/core. University of Sussex Library, on 17 Apr 2018 at 16:27:57, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781107239340.002
Section 1: Etiology, pathophysiology, and imaging

Figure 1.10. Schematic representation


of molecular injury pathways leading to
necrotic or apoptotic brain injury after
focal brain ischemia. Injury pathways can
be blocked at numerous sites, providing
multiple approaches for the amelioration
of both necrotic and apoptotic cell death.
DAG ¼ diacylglycerol; IP3 ¼ inositol 1, 4,
5-trisphosphate; PARP ¼ poly (ADP-
ribose) polymerase; TPA ¼ tissue
plasminogen activator.

greatly enhanced energy demands of the activated ion referred to as molecular injury, where the term
exchange pumps. In the healthy brain the associated “molecular” does not anticipate any particular injury
increase of glucose and oxygen demands are coupled pathway (for reviews see [88], [89]). The molecular
to a parallel increase of blood flow but in the peri- injury cascades (Figure 1.10) are interconnected in
infarct penumbra this flow response is suppressed or complex ways, which makes it difficult to predict their
even reversed [86]. As a result, a misrelationship relative pathogenic importance in different ischemia
arises between the increased metabolic workload models. In particular, molecular injury induced by
and the low oxygen supply, leading to transient epi- transient focal ischemia is not equivalent to the alter-
sodes of hypoxia and the stepwise increase in lactate ations that occur in the penumbra of permanent
during the passage of each depolarization. ischemia. Therefore, the relative contribution of the
The pathogenic importance of peri-infarct depo- following injury mechanisms differs in different types
larizations for the progression of ischemic injury is of ischemia.
supported by the linear relationship between the Acidotoxicity: during ischemia oxygen depletion
number of depolarizations and infarct volume. Cor- and the associated activation of anaerobic glycolysis
relation analysis of this relationship suggests that cause an accumulation of lactic acid which, depending
during the initial 3 hours of vascular occlusion each on the severity of ischemia, blood glucose levels, and
depolarization increases the infarct volume by more the degree of ATP hydrolysis, results in a decline
than 20%. This is probably one of the reasons that of intracellular pH to between 6.5 and below 6.0.
glutamate antagonists, which are potent inhibitors of As the severity of acidosis correlates with the
spreading depression, reduce the volume of brain severity of ischemic injury, it has been postulated
infarcts [87]. that acidosis is neurotoxic. Recently, evidence has
Peri-infarct spreading depressions are depolarizations been provided that ASICs (acid-sensing ion channels)
initiated at the border of the infarct core and may are glutamate-independent vehicles of calcium
contribute to progression of ischemic injury. flux, and that blockade of ASICs attenuates stroke
injury. This suggests that acidosis may induce calcium
toxicity, and that this effect is the actual mechanism
Molecular mechanisms of injury progression of acidotoxicity [90].
In the borderzone of permanent focal ischemia or in Excitotoxicity: shortly after the onset of ischemia,
the core of the ischemic territory after transient vas- excitatory and inhibitory neurotransmitters are
cular occlusion, cellular disturbances may evolve that released, resulting in the activation of their specific
18 cannot be explained by a lasting impairment of blood receptors. Among these neurotransmitters, particular
flow or energy metabolism. These disturbances are attention has been attributed to glutamate, which

Downloaded from https://www.cambridge.org/core. University of Sussex Library, on 17 Apr 2018 at 16:27:57, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781107239340.002
Chapter 1: Neuropathology and pathophysiology of stroke

under certain experimental conditions may produce of ER-dependent secondary disturbances, notably
excitotoxic cell death [91]. The activation of ionotro- inhibition of protein synthesis. Calcium-dependent
pic glutamate receptors results in the inflow of cal- pathological events are therefore complex and con-
cium from the extracellular into the intracellular tribute to a multitude of secondary molecular injury
compartment, leading to mitochondrial calcium pathways.
overload and the activation of calcium-dependent Free radicals: in brain regions with low or intermit-
catabolic enzymes. The activation of metabotropic tent blood perfusion, reactive oxygen species (ROS)
glutamate receptors induces the inositol 1,4,5- are formed which produce peroxidative injury of
trisphosphate (IP3)-dependent signal transduction plasma membranes and intracellular organelles [94].
pathway, leading among others to the stress response The reaction with NO leads to the formation of per-
of ER, and by induction of immediate-early genes oxynitrite, which also causes violent biochemical reac-
(IEGs) to adaptive genomic expressions. At very tions. Secondary consequences of free radical
high concentration, glutamate results in primary reactions are the release of biologically active free fatty
neuronal necrosis. However, following pharmaco- acids such as arachidonic acid, the induction of ER
logical inhibition of ionotropic glutamate receptors, stress and mitochondrial disturbances, the initiation
an apoptotic injury mechanism evolves that may of an inflammatory response, breakdown of the
prevail under certain pathophysiological conditions. blood–brain barrier, and fragmentation of DNA.
The importance of excitotoxicity for ischemic cell The last may induce apoptosis and thus enhance
injury has been debated, but this does not invalidate molecular injury pathways related to mitochondrial
the beneficial effect of glutamate antagonists for the dysfunction. The therapeutic benefit of free radial
treatment of focal ischemia. An explanation for this scavengers, however, is limited, as documented by
incongruity is the above-described pathogenic role the therapeutic failure of the free-radical-trapping
of peri-infarct depolarizations in infarct expansion. agent NXY-059 [95].
As glutamate antagonists inhibit the spread of these Nitric oxide toxicity: NO is a product of NO
depolarizations, the resulting injury is also reduced. synthase (NOS) acting on arginine. There are at least
Calcium toxicity: in the intact cell, highly efficient three isoforms of NOS: eNOS is constitutively
calcium transport systems assure the maintenance expressed in endothelial cells, nNOS in neurons, and
of a steep calcium concentration gradient of approxi- the inducible isoform iNOS mainly in macrophages.
mately 1:10 000 between the extra- and the intracel- Pathophysiologically, NO has two opposing effects
lular compartment on the one hand, and between the [96]. In endothelial cells the generation of NO leads
cytosol and the ER on the other. During ischemia to vascular dilatation, an improvement of blood flow
anoxic depolarization in combination with the acti- and the alleviation of hypoxic injury, whereas in
vation of ionotropic glutamate and acid-sensing ion neurons it contributes to glutamate excitotoxicity
channels causes a sharp rise of cytosolic calcium [92]. and – by formation of peroxynitrite – to free-
At the onset of ischemia this rise is further enhanced radical-induced injury. The net effect of NO thus
by activation of metabotropic glutamate receptors depends on the individual pathophysiological situ-
which mediate the release of calcium from ER, ation and is difficult to predict.
and after recovery from ischemia by activation of Zinc toxicity: zinc is an essential catalytic and
transient receptor potential (TRP) channels which structural element of numerous proteins and a sec-
perpetuate intracellular calcium overload despite ondary messenger which is released from excitatory
the restoration of ion gradients (Ca2+ paradox) synapses during neuronal activation. Cytosolic zinc
[93]. The changes in intracellular calcium activity overload may promote mitochondrial dysfunction
are highly pathogenic. Prolonged elevation of cyto- and generation of ROS, activate signal transduction
solic calcium causes mitochondrial dysfunction and pathways such as mitogen-activated protein kinase
induces catabolic changes, notably by activation of (MAPK), enhance calcium toxicity, and promote
Ca2+-dependent effector proteins and enzymes such apoptosis [97]. However, at low concentration zinc
as endonucleases, phospholipases, protein kinases, may also exhibit neuroprotective properties, indicat-
and proteases that damage DNA, lipids, and pro- ing that cells may possess a specific zinc set-point by
teins. The release of calcium from the ER evokes an which too little or too much zinc can promote ische- 19
ER stress response, which mediates a great number mic injury [98].

Downloaded from https://www.cambridge.org/core. University of Sussex Library, on 17 Apr 2018 at 16:27:57, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781107239340.002
Section 1: Etiology, pathophysiology, and imaging

ER stress and inhibition of protein synthesis: a mitochondrial proteins (see below). Ischemia-induced
robust molecular marker for the evolution of ische- mitochondrial disturbances thus contribute to delayed
mic injury is inhibition of protein synthesis, which cell death both by impairment of the energy state and
persists throughout the interval from the onset of the activation of apoptotic injury pathways [102].
ischemia until the manifestation of cell death [45]. It A large number of molecular disturbances are involved
is initiated by a disturbance of the calcium homoeo- in the progression of ischemic damage.
stasis of the ER, which results in ER stress and various
cell biological abnormalities such as un- or misfolding
of proteins, expression of stress proteins, and a global Inflammation
inhibition of the protein synthesizing machinery. Brain infarcts evoke a strong inflammatory response
The last is due to the activation of protein kinase which is thought to contribute to the progression of
R (PKR), which causes phosphorylation and inactiva- ischemic brain injury. Gene expressions related to this
tion of the alpha subunit of eukaryotic initiation response have, therefore, been extensively investigated
factor eIF2. This again leads to selective inhibition of to search for possible pharmacological targets (for
polypeptide chain initiation, disaggregation of ribo- review see Rothwell and Luheshi [103]). The inflamma-
somes, and inhibition of protein synthesis at the level tory response of the ischemic tissue has been associated,
of translation. among others, with the generation of free radicals in
To restore ER function un- or misfolded proteins reperfused or critically hypoperfused brain tissue. The
must be refolded (by activation of the unfolded pro- prostaglandin synthesizing enzyme cyclo-oxygenase-2
tein response, UPR) or degraded (by ER-associated (COX-2) and nuclear factor-kappa B (NF-kappa B), a
degradation, ERAD). Cells in which UPR and ERAD transcription factor that responds to oxidative stress, are
fail to restore ER function die by apoptosis [99]. strongly upregulated and may be neurotoxic, as sug-
Obviously, persistent inhibition of protein synthe- gested by the beneficial effect of COX-2 inhibitors.
sis is incompatible with cell survival but as the mani- Infarct reduction was also observed after genetic or
festation of cell death greatly varies in different cell pharmacological inhibition of matrix metalloproteinase
populations, other factors must also be involved. (MMP)-9 but this effect has been disputed.
Mitochondrial disturbances: to mitigate metabolic A key player in the intracellular response to cyto-
or environmental stress, functional mitochondria kines is the JAK (janus kinase)/STAT (signal trans-
are maintained by fission and fusion [100]. However, ducer and activator of transcription) pathway, which
the concurrence of an increased cytosolic calcium induces alterations in the pattern of gene transcription.
activity with the generation of ROS may lead to an These changes are associated with either cell death or
increase in permeability of the inner mitochondrial survival and suggest that inflammation may be both
membrane (mitochondrial permeability transition, neurotoxic and neuroprotective [104]. Inflammatory
MPT), which has been associated with the formation reactions and the associated free-radical-mediated pro-
of a permeability transition pore (PTP). The PTP is cesses are, therefore, important modulators of ischemic
a Ca2+-, ROS- and voltage-dependent, cyclosporine injury but the influence on the final outcome is difficult
A-sensitive high-conductance channel, located in to predict.
the inner mitochondrial membrane. It is also a Inflammatory reactions are important modulators of
reversible fast Ca2+ release channel, facilitated by the ischemic injury.
mitochondrial matrix protein cyclophilin D [101].
The increase in permeability of the inner mito-
chondrial membrane has two pathophysiologically Brain edema
important consequences. The breakdown of the Ischemic brain edema can be differentiated into two
electrochemical gradient interferes with mitochon- pathophysiologically different types: an early cytotoxic
drial oxidative phosphorylation and, in consequence, type, followed after some delay by a late vasogenic type
with aerobic energy production. Furthermore, the of edema. The cytotoxic type of edema is threshold
equilibration of mitochondrial ion gradients causes dependent. It is initiated at flow values of similar to
swelling of the mitochondrial matrix, which eventu- 30% of control when stimulation of anaerobic metab-
20 ally will cause disruption of the outer mitochon- olism causes an increase of brain tissue osmolality and,
drial membrane and the release of pro-apoptotic hence, an osmotically obliged cell swelling. At flow

Downloaded from https://www.cambridge.org/core. University of Sussex Library, on 17 Apr 2018 at 16:27:57, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781107239340.002
Chapter 1: Neuropathology and pathophysiology of stroke

values below 20% of control, anoxic depolarization ischemic brain edema. Similarly, the inhibition of
and equilibration of ion gradients across the cell mem- sodium transport across sodium channels has been
branes further enhance intracellular osmolality and the suggested to reduce edema formation. However, as
associated cell swelling. The intracellular uptake of the driving force for the generation of edema is the
sodium is also associated with a coupled movement gradient of osmotic and ionic concentration differ-
of water that is independent of an osmotic gradient ences built up during ischemia, aquaporin and
and which is referred to as “anomalous osmosis.” sodium channels may modulate the speed of edema
In the absence of blood flow, cell swelling occurs at generation but not the final extent of tissue water
the expense of the extracellular fluid volume, leading accumulation. Their pathophysiological importance
to the shrinkage of the extracellular compartment, but is, therefore, limited.
not to a change in the net water content. The shift of Early cytotoxic edema is caused by osmotically induced
fluid is reflected by a decrease of the ADC of water, cell swelling; the later vasogenic edema is isoosmotic,
which is the reason for the increase of signal intensity caused by breakdown of the blood–brain barrier, and
in diffusion-weighted MR imaging [80]. However, if accumulates in the extracellular compartment.
some residual blood flow persists, water is taken up
from the blood, and the net tissue water content
increases. After vascular occlusion this increase starts Apoptosis
within a few minutes after the onset of ischemia and Apoptosis is an evolutionary conserved form of pro-
causes a gradual increase in brain volume. grammed cell death that in multicellular organisms
With the evolution of tissue necrosis and the deg- matches cell proliferation to preserve tissue homoeo-
radation of basal lamina, the blood–brain barrier stasis [109]. It is an active process that requires intact
breaks down [105], and after 4–6 hours serum pro- energy metabolism and protein synthesis, and it is
teins begin to leak from the blood into the brain. This initiated essentially by two pathways: an extrinsic
disturbance initiates a vasogenic type of edema, which death receptor-dependent route, and an intrinsic
further enhances the water content of the tissue. pathway which depends on the mitochondrial release
Vasogenic edema reaches its peak at 1–2 days after of pro-apoptotic molecules such as apoptosis-
the onset of ischemia and may cause an increase of inducing factor (AIF) and cytochrome C. In focal
tissue water by more than 100%. If brain infarcts are ischemia pro-apoptotic pathways are also initiated
large, the volume increase of the edematous brain by activation of toll-like receptors 2 and 4, the
tissue may be so pronounced that transtentorial her- NOTCH-1 receptor, and the adiponectin receptor 1.
niation results in compression of the midbrain. Under Apoptotic pathways involve a series of enzymatic
clinical conditions, this “malignant” form of brain reactions and converge in the activation of caspase-
infarction is by far the most dangerous complica- 3, a cystine protease, which contributes to the execu-
tion of stroke and an indication for decompressive tion of cell death. An end stage of this process is the
craniectomy [106]. ordered disassembly of the genome, resulting in a
Vasogenic edema, in contrast to the early cyto- laddered pattern of oligonucleosomal fragments as
toxic type of edema, is isoosmotic and accumulates detected by electrophoresis or TUNEL.
mainly in the extracellular compartment. This Although apoptosis is mainly involved in physio-
reverses the cytotoxic narrowing of the extracellular logical cell death, it is widely assumed to contribute to
space and explains the “pseudonormalization” of the the pathogenesis of diseases, including cerebral ische-
signal intensity observed in diffusion-weighted MR mia [110]. In the context of stroke this is difficult to
imaging [107]. However, as the total tissue water understand because in areas with primary cell death
content is increased at this time, the high signal the obvious cause is energy failure, and in regions
intensity in T2-weighted images clearly differentiates with delayed injury the dominating biochemical dis-
this situation from a “real” recovery to normal. turbance is the irreversible suppression of protein
The formation of cytotoxic and, to a lesser extent, synthesis. However, ischemia induces a multitude of
also vasogenic edema requires the passage of water biochemical reactions that are reminiscent of apop-
through aquaporin channels located in the plasma tosis, such as the expression of p53, JNK, c-jun, p38,
membrane [108]. Inhibition of aquaporin water cyclin-dependent kinase 5, or caspase 3, all of which 21
conductance may, therefore, reduce the severity of correlate to some degree with the severity of injury.

Downloaded from https://www.cambridge.org/core. University of Sussex Library, on 17 Apr 2018 at 16:27:57, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781107239340.002
Section 1: Etiology, pathophysiology, and imaging

Moreover, inhibition of these reactions by gene protein kinases, such as extracellular signal-regulated
manipulation or pharmacological interventions kinase (ERK), p38 MAPK, and Akt. The possibility to
reduces the volume of brain infarcts. It has, therefore, influence ischemic injury after the primary impact
been suggested that ischemic cell death is a hybrid of is challenging but it remains to be shown for which
necrosis and apoptosis, appearing on a continuum kind of clinical situation this finding is of practical
with the two forms of cell death at its poles [111]. relevance.
Apoptosis, an active form of programmed cell death, Short episodes of ischemia can improve the
may contribute to a certain extent to ischemic cell death. tolerance of brain tissue for subsequent blood flow
disturbance.

Pre- and postconditioning of ischemic injury


The molecular signaling cascades initiated by brain Regeneration and cell therapy
ischemia are not solely destructive but may also exert Brain infarcts produced by focal ischemia are seem-
a neuroprotective effect. In fact, most of the above- ingly irresolvable in agreement with Cajal’s classical
described injury pathways including ischemia itself statement that in the adult brain “everything may die,
induce a transient state of increased ischemic toler- nothing may be regenerated.” This dogma was
ance, provided the initial injury remains subliminal reversed by the discovery of three permanently neuro-
for tissue destruction. This effect is called “ischemic genic regions, i.e. the subventricular zone (SVZ), the
preconditioning” and can be differentiated into three subgranular zone (SGZ), and the posterior perireticu-
phases: during the induction phase molecular sensors lar (PPr) area, which provide lifelong supply of
which respond to the preconditioning stimulus are newly generated neurons to the hippocampus and
activated by transcription factors; the transduction olfactory bulb. After stroke, neurogenesis increases
phase results in the amplification of the signal; and in these areas, and some of the newly formed cells
during the effector phase proteins with a protective migrate into the infarct penumbra, differentiate into
impact are switched on [112]. The increase in ische- glia and mature neurons, and survive for at least
mia tolerance appears 2 to 3 days after the precondi- several weeks [114]. Neurogenesis may also occur
tioning stimulus, and it slowly disappears after through the neurovascular unit. After ischemia peri-
1 week. cytes strongly migrate into the peri-infarct surround-
An important preconditioning pathway is the upre- ing and contribute to tissue repair by controlling
gulation of the hypoxia-inducible factor 1 (HIF-1) neurogenesis, angiogenesis, and blood–brain barrier
in astrocytes. HIF-1 is a transcription factor that among function [115].
others induces the expression of erythropoietin (EPO) Ischemia-induced neurogenesis is enhanced by
which binds to the neuronal EPO receptor and growth factors, NO, inflammation, and various hor-
which exhibits potent neuroprotective effects. Another mones and neurotransmitters, notably estradiol and
putative mechanism is the ER stress response. Depletion dopamine, but it is repressed by activation of the N-
of ER calcium stores causes accumulation of unfolded methyl-D-aspartate (NMDA) subtype of glutamate
proteins in the ER lumen and induces the activation receptors. The functional consequences of spontan-
of two highly conserved stress responses, the ER over- eous or drug-enhanced neurogenesis are modest but
load response (EOR) and the unfolded protein response optimism is building up for targeted interventions.
(UPR). EOR triggers activation of the transcription Similarly, considerable expectations are placed on the
factor NF-kappa B, and UPR causes a suppression of transplantation of neural progenitor cells, particularly
the initiation of protein synthesis. As the latter contri- in combination with growth factors and/or strategies
butes to delayed ischemic injury (see above) its reduc- that permit recruitment of transplanted cells to the
tion may have a neuroprotective effect. site of injury [116]. However, cell therapy carries the
Evidence has also been provided that ischemic risk of tumorigenesis, and as major breakthroughs
injury can be alleviated by repeated mechanical inter- have not yet been achieved, further research is neces-
ruptions of blood reperfusion after a period of transi- sary to explore the actual potentials of stroke regen-
ent focal ischemia [113]. This phenomenon, termed erative medicine.
22 “ischemic postconditioning,” has been associated Several brain regions may provide lifelong supply of
with the phosphorylation of several prosurvival newly generated neurons.

Downloaded from https://www.cambridge.org/core. University of Sussex Library, on 17 Apr 2018 at 16:27:57, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781107239340.002
Chapter 1: Neuropathology and pathophysiology of stroke

Translation of experimental concepts was observed in regions with decreased flow in the
first hours after the ictus. Since the 1980s, PET with
to clinical stroke oxygen-15 tracers became the gold standard for the
Experimental research has advanced our knowledge evaluation of pathophysiological changes in early
about the pathophysiology of brain disorders, but ischemic stroke [118]. The quantitative measurement
the transfer of this knowledge into clinical appli- of CBF, oxygen utilization (CMRO2), oxygen extrac-
cation is difficult and often lacks behind. One tion fraction (OEF), and cerebral blood volume
of the reasons are differences between the brain (CBV) permitted the independent assessment of per-
of experimental animals and humans with respect fusion and energy metabolism, and demonstrated the
to evolutionary state (non-gyrencephalic vs. gyrence- uncoupling of these usually closely related variables.
phalic anatomy (amount of gray vs. white matter), These studies provided data on flow and metabolic
relative size, cellular density, blood supply, and variables predicting final infarction on late CTs (rCBF
metabolism). Additionally, experimental models in less than 12 ml/100 g/min, CMRO2 less than 65 μmol/
animals cannot be easily compared to complex 100 g/min). Relatively preserved CMRO2 indicated
human diseases often based on a different patho- maintained neuronal function in regions with
physiology and affecting multimorbid patients. The severely reduced CBF; this pattern was coined “misery
other problem arises from the investigative pro- perfusion” and served as a definition for the penum-
cedures, which cannot be equally applied in animals bra, which is characterized by increased oxygen
and patients. This is especially true when pathophy- extraction fraction (up to more than 80% from the
siological changes obtained by invasive procedures in normal 40–50%). Late CT or MRI often showed these
animals, e.g. by analysis of tissue samples, by auto- regions as morphologically intact.
radiography, or by histology, should be related to the Sequential PET studies of CBF, CMRO2, and
course of a disease, but cannot be assessed repeatedly metabolic rate of glucose (CMRGlc) before and
and regionally. To facilitate the transfer of knowledge repeatedly up to 24 hours after MCA occlusion in
from experimental neuroscience to clinical neurology, cats could demonstrate the development and growth
it is necessary to develop methods which can be of irreversible ischemic damage. Immediately after
equally applied in patients and animal models, and MCA occlusion CBF within the supplied territory
which are not invasive and can be performed repeat- dropped, but CMRO2 was less diminished and was
edly without affecting or harming the object. To this preserved at an intermediate level. As a consequence,
task of transferring experimental results into clinical OEF was increased, indicating misery perfusion, i.e.
application functional imaging modalities are success- penumbra tissue. However, as OEF is also increased
fully applied. in benign oligemia, demarcation of the penumbra
from normal tissue is not possible. With time, OEF
was decreased, a process which started in the center
Detection of the penumbra by PET and developed centrifugally to the borderline of the
PET is still the only method allowing quantitative ischemic territory, indicating the conversion into irre-
determination of various physiological variables in versible damage and the growth of the MCA infarct.
the brain and was applied extensively for studies in In experiments with transient MCA occlusion it could
patients with acute, subacute, or chronic stages of be demonstrated that an infarct did not develop when
ischemic stroke (review in Heiss [77]). The introduc- reperfusion was initiated to tissue with increased
tion of scanners with high resolution (2.5–5 mm for OEF. Comparable to patients with early thrombolysis,
human, 1 mm for animal application) made PET a reperfusion could salvage ischemic tissue in the con-
tool for studying animal models and to compare dition of “penumbra” (Figure 1.11). Similar results
repeat examinations of various variables from experi- were obtained in ischemia models of baboons. PET
ments to the course of disease in humans. The thus permits the differentiation of various tissue com-
regional decrease of CBF can be directly observed in partments within an ischemic territory: irreversible
PET as in other studies (single-photon emission com- damage by decreased flow and oxygen consumption
puted tomography [SPECT], PWI-MRI, perfusion below critical thresholds; misery perfusion, i.e. pen-
computed tomography [PCT]). However, already in umbra, by decreased flow, but preserved oxygen util- 23
early PET studies [117] preserved glucose consumption ization above a critical threshold, expressed by

Downloaded from https://www.cambridge.org/core. University of Sussex Library, on 17 Apr 2018 at 16:27:57, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781107239340.002
Section 1: Etiology, pathophysiology, and imaging

CAT HUMAN

CBF

CMRO2

OEF

Reperfusion spontaneous course rtPA treatment

OEF CMRGlc CBF

CMRGlc

Figure 1.11 Sequential PET images of CBF, CMRO2, and OEF of MCA occlusion in cats compared to images of patients after stroke: Left
columns: In the right cat, the progressive decrease of CMRO2 and the reduction of OEF predict infarction that cannot benefit from reperfusion.
Only if OEF is increased until the start of reperfusion can the infarct be salvaged (left cat). Middle columns: In the patient the areas with
preserved OEF are not infarcted and can survive in spontaneous course (posterior part of ischemic cortex in left, anterior part in right patient,
as indicated on late MRI and CT). Right columns: In patients receiving rtPA treatment measurements of CMRO2 and OEF are not feasible, but
flow determinations show the effect. If reperfusion occurs early enough and before tissue damage, tissue can be salvaged (left patient). If
reperfusion is achieved too late, tissue cannot be salvaged despite hyperperfusion in some parts (right patient).

increased OEF; luxury perfusion by flow increased is of utmost importance for the efficiency of treat-
above the metabolic demand; anaerobic glycolysis by ment. Meticulous analyses of CBF and CMRO2 data
a change in the ratio between glucose metabolism and indicated that CMRO2 below 65 μmol/100 g/min
oxygen utilization. However, PET has severe disad- predicted finally infarcted tissue, but also large
vantages limiting its routine application in patients portions with flow and oxygen utilization in the pen-
with stroke: it is a complex methodology, requires umbra range were included in the final cortical–
multitracer application, and quantitative analysis subcortical infarcts. Determination of oxygen
necessitates arterial blood sampling. utilization additionally requires arterial blood sam-
Positron emission tomography (PET) is the most pling, which limits clinical applicability. These facts
reliable non-invasive method to identify irreversible stress the need for a marker of neuronal integrity
tissue damage and penumbra. that can identify irreversibly damaged tissue irre-
spective of the time elapsed since the vascular attack,
and irrespective of the variations in blood flow
over time.
Prediction of irreversible tissue damage Central benzodiazepine receptor (BZR) ligands
24 The prediction of the portion of irreversibly damaged can be used as markers of neuronal integrity as
tissue within the ischemic area early after the stroke they bind to the gamma-aminobutyric acid (GABA)

Downloaded from https://www.cambridge.org/core. University of Sussex Library, on 17 Apr 2018 at 16:27:57, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781107239340.002
Chapter 1: Neuropathology and pathophysiology of stroke

receptors abundant in cerebral cortex that are sensi- H2O and therefore can be used as a relative flow
tive to ischemic damage. After successful testing in tracer yielding reliable perfusion images. PET with
the cat MCA occlusion model, cortical binding of FMZ therefore can be used as a non-invasive proce-
flumazenil (FMZ) was investigated in patients with dure to image irreversible damage and critically
acute ischemic stroke [119]. In all patients, defects reduced perfusion (i.e. penumbra) in early ischemic
in FMZ binding were closely related to areas with stroke.
severely depressed oxygen consumption and pre- Special tracers, e.g. ligands to benzodiazepine
dicted the size of the final infarcts, whereas preserved receptors, can be used as early markers of irreversible
FMZ binding indicated intact cortex. Additionally, neuronal damage, since they bind to GABA receptors
FMZ distribution within 2 minutes after tracer injec- of cortical neurons, which are very sensitive to
tion was highly correlated with CBF measured by ischemic damage.

Figure 1.12. (a, b) Volumetric comparison of time to peak (TTP) (MRI) and OEF (PET) images in two patients measured in the chronic
phase of stroke. In both patients a TTP delay of >4 seconds indicates a considerable mismatch volume (red contour on TTP images).
The mismatch volumes were 473 cm3 for patient a and 199.7 cm3 for patient b. However, only patient b had a corresponding volume
of penumbra (260 cm3). (c) Volumes of penumbra defined by increased OEF (black) and mismatch defined by TTP > 4 (gray) in 13 patients:
all 13 patients showed mismatch, only 8 patients showed penumbra, which comprised 1–75% of the mismatch volume. (Modified with 25
permission from Sobesky et al. [124].)

Downloaded from https://www.cambridge.org/core. University of Sussex Library, on 17 Apr 2018 at 16:27:57, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781107239340.002
Section 1: Etiology, pathophysiology, and imaging

Figure 1.12. (cont.)

Surrogate markers for penumbra and adding the portion of reversible diffusion abnormality
[121] the definition of the penumbra remains contro-
irreversible damage versial and suffers from lack of standardization of
Although PET remains the imaging gold standard for methodological approaches to imaging, post process-
identification of the penumbra in stroke patients, MR ing, and analysis, which restricts pooling of data and
studies using DWI and perfusion-weighted imaging cross-comparison of results across studies.
(PWI) might provide a differentiation between the A validation of PWI/DWI results with quantitative
core and the penumbra: the early DWI lesion might measurements of flow values and oxygen consumption
define the ischemic core and adjacent critically hypo- or FMZ uptake in the same patients early after stroke
perfused tissue might be identified with PWI [120]. is necessary for the assessment of the accuracy of
Therefore, brain regions with hypoperfusion assessed the applied signatures for predicting tissue outcome.
by PWI but without restricted diffusion (PWI/DWI Several studies were performed in order to validate
mismatch) were assumed to represent the penumbra. mismatch as a surrogate of penumbra on PET-derived
This surrogate definition of the penumbra has several discrimination of irreversibly damaged, critically per-
uncertainties [121]: the initial diffusion lesion does fused “at risk,” and oligemic “not at risk” tissue. The
not only consist of irreversibly infarcted tissue; diffu- studies demonstrated that the DWI lesion predicts
sion lesions may be reversed if blood flow is restored more or less the finally infarcted tissue [122], but
at an early time point; critically perfused tissue (i.e. contains up to 25% false positive, i.e. surviving tissue.
penumbra) cannot be clearly differentiated from The inaccuracy in defining the penumbra with
tissue experiencing benign oligemia; the PWI abnor- PWI/DWI mismatch is thought to be mainly related
mality often overestimates the amount of tissue at to PW data acquisition, since the parameters used to
risk. These facts are further accentuated by methodo- estimate perfusion are variable and somewhat arbi-
logical limitations, because perfusion techniques and trary. As a consequence, perfusion lesion size differs
data evaluation are not quantitative and vary among markedly depending on the parameters calculated
26 centers. Despite the attempts to correct the mismatch [123] and usually is overestimated and extends into
area by subtracting the region of benign oligemia and considerable areas with non-critical oligemia especially

Downloaded from https://www.cambridge.org/core. University of Sussex Library, on 17 Apr 2018 at 16:27:57, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781107239340.002
Chapter 1: Neuropathology and pathophysiology of stroke

when short delays are used. Overall, PWI is unable to sinuses or veins and are often accompanied by
provide a reliable quantitative estimation of cerebal edema, hemorrhagic transformation, and bleeding.
perfusion when compared to gold-standards such as Primary ischemic cell death is the result of severe
PET, SPECT, or Xe-CT and overestimated the size of ischemia; early signs are potentially reversible swell-
the critically perfused tissue and therefore also the ing or shrinkage; irreversible necrotic neurons have
volume of critically perfused but salvageable tissue, condensed acidophilic cytoplasm and pyknotic
i.e. the penumbra (Figure 1.12) [124]. Of 13 patients nuclei. Delayed neuronal death can occur after
showing considerable PWI/DWI mismatch only moderate or short-term ischemia; it goes along
8 had areas with elevated OEF typical for penumbra with nuclear fragmentation and development of
apoptotic bodies. The pathophysiology of ischemic
tissue, and these areas were always smaller than MR
cell damage in animal models reflects only certain
mismatch. Overall, the mismatch volume in PWI/
aspects of ischemia and cannot give a complete
DWI as conventionally calculated does not reliably picture of ischemic stroke in humans. However, from
reflect misery perfusion, i.e. the penumbra as defined these experimental models principles of regulation of
by PET. Recently, several methods have been pro- cerebral blood flow and flow thresholds for mainten-
posed to improve the reliability of assessment of ance of function and morphology can be deduced.
perfusion using MR methods [125], but they all need As the energy requirement of the brain is very high,
to be validated by quantitative measures. already mild decreases of blood supply lead to
The mismatch between perfusion-weighted and potentially reversible disturbance of function and,
diffusion-weighted MRI can be used as a surrogate if the shortage is more severe and persists for cer-
marker of the penumbra, but the applied parameters tain periods, to irreversible morphological damage.
need to be validated. Tissue perfused in the range between the thresholds
of functional and morphological injury has been
called the penumbra, a concept which has great
Chapter summary importance for treatment. The ischemia-induced
energy failure triggers a complex cascade of electro-
physiological disturbances, biochemical changes,
Atherosclerosis is the most widespread disorder and molecular dysfunction, which lead to progres-
leading to death and serious morbidity including sive cell death and growth of infarction. The pro-
stroke. It develops over years from initial fatty streaks gression of ischemic injury is further boosted by
to atheromatous plaques with the risk of plaque inflammatory reactions and the development of
disruption and formation of thrombus, from which early cytotoxic and later vasogenic brain edema.
emboli might originate. Lipohyalinosis affects small The translation of these experimental concepts into
vessels, leading to lacunar stroke. The vascular clinical application and management of stroke
lesions and emboli from the heart cause territorial patients, however, is difficult. It can be achieved
infarcts, whereas borderzone infarcts are due to low in some instances by special functional imaging
perfusion in the peripheral parts of the vascular ter- techniques, such as positron emission tomography,
ritories (last meadows). Ischemic infarcts may be but requires further refinements to predict the
converted into hemorrhagic infarctions by leakage outcome of neuroprotective interventions. Solid
of vessels, whereas intracerebral hemorrhages understanding of experimental and clinical stroke
(5–15% of all strokes) result from rupture of arteries pathophysiology is, therefore, needed to improve
typically in deep portions of the hemispheres. the reliability of translational research.
Venous infarcts usually result from thrombosis of

References Ginsberg MD, Bogousslavsky J, 4. Ross R. Atherosclerosis–an


eds. Cerebrovascular Disease: inflammatory disease. N Engl
1. Sanz J, Moreno PR, Fuster V. Pathophysiology, Diagnosis and J Med 1999; 340:115–26.
The year in atherothrombosis. Management. London: Blackwell
J Am Coll Cardiol 2012; 5. Faxon DP, Fuster V, Libby P, et al.
Science; 1998: 308–18. Atherosclerotic Vascular Disease
60:932–42.
3. Willeit J, Kiechl S. Biology of Conference: Writing Group III:
2. Rajamani K, Fisher M, Fisher M. arterial atheroma. Cerebrovas Dis pathophysiology. Circulation
Atherosclerosis – pathogenesis 2000; 10(Suppl 5):1–8. 2004; 109:2617–25. 27
and pathophysiology. In:

Downloaded from https://www.cambridge.org/core. University of Sussex Library, on 17 Apr 2018 at 16:27:57, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781107239340.002
Section 1: Etiology, pathophysiology, and imaging

6. Aikawa M, Libby P. The 17. Dichgans M. Genetics of 27. Lodder J, Krijne-Kubat B,


vulnerable atherosclerotic plaque: ischaemic stroke. Lancet Neurol Broekman J. Cerebral
pathogenesis and therapeutic 2007; 6:149–61. hemorrhagic infarction at
approach. Cardiovasc Pathol 2004; 18. Mohr JP, Choi DW, Grotta JC, autopsy: cardiac embolic cause
13:125–38. Weir B, Wolf PA (eds.). Stroke – and the relationship to the cause
Pathophysiology, Diagnosis, and of death. Stroke 1986; 17:626–9.
7. Koga J, Aikawa M. Crosstalk
between macrophages and Management, 4th edn. 28. Fisher M, Adams RD.
smooth muscle cells in Philadelphia: Churchill Observations on brain embolism
atherosclerotic vascular Livingstone; 2004. with special reference to the
diseases. Vascul Pharmacol 19. Pantoni L. Cerebral small vessel mechanism of hemorrhagic
2012; 57:24–8. disease: from pathogenesis and infarction. J Neuropathol Exp
clinical characteristics to Neurol 1951; 10:92–4.
8. Dzau VJ, Braun-Dullaeus RC,
Sedding DG. Vascular therapeutic challenges. Lancet 29. Mohr JP, Caplan LR, Melski JW,
proliferation and atherosclerosis: Neurol 2010; 9:689–701. et al. The Harvard Cooperative
new perspectives and therapeutic 20. Ringelstein EB, Nabavi DG. Stroke Registry: a prospective
strategies. Nat Med 2002; Cerebral small vessel diseases: registry. Neurology 1978; 28:754–62.
8:1249–56. cerebral microangiopathies. 30. Sacco RL, Wolf PA, Bharucha NE,
9. Madden JA. Role of the vascular Curr Opin Neurol 2005; et al. Subarachnoid and
endothelium and plaque in acute 18:179–88. intracerebral hemorrhage: natural
ischemic stroke. Neurology 2012; 21. Wolf PA. Epidemiology of history, prognosis, and precursive
79:S58–62. stroke. In: Mohr JP, Choi DW, factors in the Framingham Study.
Grotta JC, Weir B, Wolf PA, Neurology 1984; 34:847–54.
10. Glagov S, Weisenberg E, Zarins
eds. Stroke – Pathophysiology, 31. Feldman E. Intracerebral
CK, Stankunavicius R, Kolettis GJ.
Diagnosis, and Management, Hemorrhage. Armonk, New York:
Compensatory enlargement of
4th edn. Philadelphia: Futura; 1994.
human atherosclerotic coronary
Churchill Livingstone; 2004:
arteries. N Engl J Med 1987; 32. Schütz H. Spontane intrazerebrale
13–34.
316:1371–5. Hämatome. Pathophysiologie,
22. Stochdorph O. Der Mythos Klinik und Therapie. Berlin,
11. Rauch U, Osende JI, Fuster V,
der letzten Wiese. Zentralbl Heidelberg, New York: Springer-
et al. Thrombus formation on
Allg Pathol Path Anat 1977; Verlag; 1988.
atherosclerotic plaques:
121:554.
pathogenesis and clinical 33. Qureshi AI, Tuhrim S, Broderick
consequences. Ann Intern Med 23. Ringelstein EB, Zunker P. Low- JP, et al. Spontaneous
2001; 134:224–38. flow infarction. In: Ginsberg MD, intracerebral hemorrhage. N Engl
Bogousslavsky J, eds. J Med 2001; 344:1450–60.
12. Lubos E, Handy DE, Loscalzo J. Cerebrovascular Disease:
Role of oxidative stress and nitric Pathophysiology, Diagnosis, and 34. Kase CS, Mohr JP, Caplan LR.
oxide in atherothrombosis. Front Management. London: Blackwell Intracerebral hemorrhage. In:
Biosci 2008; 13:5323–44. Science; 1998: 1075–89. Mohr JP, Choi DW, Grotta JC,
13. Fisher CM. Cerebral miliary Weir B, Wolf PA, eds. Stroke –
24. Fisher CM. Lacunes: small, deep
aneurysms in hypertension. Pathophysiology, Diagnosis, and
cerebral infarcts. Neurology 1965;
Am J Pathol 1972; 66: Management, 4th edn.
15:774–84.
313–30. Philadelphia: Churchill
25. Amarenco P, Bogousslavsky J, Livingstone; 2004: 327–76.
14. Zülch K-J. The Cerebral Infarct. Caplan LR, Donnan GA,
Pathology, Pathogenesis, and 35. Fisher CM. Pathological
Hennerici MG. New approach to
Computed Tomography. Berlin, observations in hypertensive
stroke subtyping: the A-S-C-O
Heidelberg, New York, Tokyo: cerebral hemorrhage.
(phenotypic) classification of
Springer-Verlag; 1985. J Neuropathol Exp Neurol 1971;
stroke. Cerebrovasc Dis 2009;
30:536–50.
15. Russell RWR. Observations on 27:502–8.
intracerebral aneurysms. Brain 26. Beghi E, Bogliun G, Cavaletti G, 36. Brott T, Broderick J, Kothari R,
1963; 86:425–42. et al. Hemorrhagic infarction: risk et al. Early hemorrhage growth in
16. Zülch K-J. Über die Entstehung factors, clinical and tomographic patients with intracerebral
und Lokalisation der Hirninfarkte. features, and outcome. A case- hemorrhage. Stroke 1997; 28:1–5.
28 Zentralbl Neurochir 1961; control study. Acta Neurol Scand 37. Gonzalez-Duarte A, Cantu C,
21:158–78. 1989; 80:226–31. Ruiz-Sandoval JL,

Downloaded from https://www.cambridge.org/core. University of Sussex Library, on 17 Apr 2018 at 16:27:57, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781107239340.002
Chapter 1: Neuropathology and pathophysiology of stroke

Barinagarrementeria F. Recurrent or consequence? J Cereb Blood imaging and histopathology. Eur


primary cerebral hemorrhage: Flow Metab 2012; 32:1207–21. J Radiol 2007; 63:68–75.
frequency, mechanisms, and 48. Hudgins WR, Garcia JH. 56. Mehra M, Henninger N, Hirsch
prognosis. Stroke 1998; 29: Transorbital approach to the JA, et al. Preclinical acute
1802–5. middle cerebral artery of the ischemic stroke modeling.
38. Bousser MG, Barnett HJM. squirrel monkey: a technique for J Neurointerv Surg 2012; 4:307–13.
Cerebral venous thrombosis. In: experimental cerebral infarction 57. Mordasini P, Frabetti N, Gralla J,
Mohr JP, Choi DW, Grotta JC, applicable to ultrastructural et al. In vivo evaluation of the first
Weir B, Wolf PA, eds. Stroke – studies. Stroke 1970; 1:107–11. dedicated combined flow-
Pathophysiology, Diagnosis, and 49. Tamura A, Graham DI, restoration and mechanical
Management, 4th edn. McCulloch J, Teasdale GM. Focal thrombectomy device in a swine
Philadelphia: Churchill cerebral ischaemia in the rat: 1. model of acute vessel occlusion.
Livingstone; 2004: 301–25. Description of technique and early AJNR Am J Neuroradiol 2011;
39. Petito CK. The Neuropathology of neuropathological consequences 32:294–300.
Focal Brain Ischemia. Basel: ISN following middle cerebral artery 58. Koller A, Toth P. Contribution of
Neuropath; 2005. occlusion. J Cereb Blood Flow flow-dependent vasomotor
40. Brown AW, Brierley JB. Anoxic- Metab 1981; 1:53–60. mechanisms to the autoregulation
ischaemic cell change in rat brain 50. Koizumi J, Yoshida Y, Nakazawa of cerebral blood flow. J Vasc Res
light microscopic and fine- T, Ooneda G. Experimental 2012; 49:375–89.
structural observations. J Neurol studies of ischemic brain 59. Symon L. Regional vascular
Sci 1972; 16:59–84. edema. 1. A new experimental reactivity in the middle cerebral
41. Jortner BS. The return of the dark model of cerebral embolism arterial distribution. An
neuron. A histological artifact in rats in which recirculation experimental study in baboons.
complicating contemporary can be introduced in the J Neurosurg 1970; 33:532–41.
neurotoxicologic evaluation. ischemic area. Jpn J Stroke 1986;
8:1–8. 60. Hata R, Maeda K, Hermann D,
Neurotoxicology 2006; 27:628–34. Mies G, Hossmann KA. Evolution
51. Rogers DC, Campbell CA, of brain infarction after transient
42. Kirino T, Sano K. Selective
Stretton JL, Mackay KB. focal cerebral ischemia in mice.
vulnerability in the gerbil
Correlation between motor J Cereb Blood Flow Metab 2000;
hippocampus following transient
impairment and infarct volume 20:937–46.
ischemia. Acta Neuropathol 1984;
after permanent and transient
62:201–8. 61. del Zoppo GJ. The neurovascular
middle cerebral artery occlusion
43. Martin LJ. Neuronal cell death in in the rat. Stroke 1997; 28:2060–5; unit in the setting of stroke.
nervous system development, discussion 2066. J Intern Med 2010; 267:156–71.
disease, and injury (Review). Int 62. Gursoy-Ozdemir Y, Yemisci M,
52. Hossmann KA. The two
J Mol Med 2001; 7:455–78. Dalkara T. Microvascular
pathophysiologies of focal brain
44. Charriaut-Marlangue C, Ben-Ari ischemia: implications for protection is essential for
Y. A cautionary note on the use of translational stroke research. successful neuroprotection in
the TUNEL stain to determine J Cereb Blood Flow Metab 2012; stroke. J Neurochem 2012; 123
apoptosis. Neuroreport 1995; 32:1310–16. (Suppl 2):2–11.
7:61–4.
53. DiNapoli VA, Rosen CL, 63. Liebeskind DS. Collateral
45. Hossmann KA. Disturbances of Nagamine T, Crocco T. Selective circulation. Stroke 2003;
cerebral protein synthesis and MCA occlusion: a precise embolic 34:2279–84.
ischemic cell death. Prog Brain Res stroke model. J Neurosci Methods 64. Toole JF, McGraw CP. The steal
1993; 96:161–77. 2006; 154:233–8. syndromes. Annu Rev Med 1975;
46. DeGracia DJ, Jamison JT, 54. Orset C, Macrez R, Young AR, et al. 26:321–9.
Szymanski JJ, Lewis MK. Mouse model of in situ 65. Pakkenberg B, Gundersen HJ.
Translation arrest and ribonomics thromboembolic stroke and Neocortical neuron number in
in post-ischemic brain: layers and reperfusion. Stroke 2007; 38:2771–8. humans: effect of sex and age.
layers of players. J Neurochem 55. Chen F, Suzuki Y, Nagai N, et al. J Comp Neurol 1997; 384:312–20.
2008; 106:2288–301. Rodent stroke induced by 66. Clarke DD, Sokoloff L.
47. Stanimirovic DB, Friedman A. photochemical occlusion of Circulation and energy
Pathophysiology of the proximal middle cerebral artery: metabolism of the brain. In: Siegel 29
neurovascular unit: disease cause evolution monitored with MR G, Agranoff B, Albers RW, Fisher

Downloaded from https://www.cambridge.org/core. University of Sussex Library, on 17 Apr 2018 at 16:27:57, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781107239340.002
Section 1: Etiology, pathophysiology, and imaging

S, eds. Basic Neurochemistry: cerebral artery occlusion in mice. 85. Dreier JP. The role of spreading
Molecular, Cellular, and Medical J Cereb Blood Flow Metab 2000; depression, spreading
Aspects, 6th edn. Philadelphia: 20:306–15. depolarization and spreading
Lippincott-Raven; 1999: 637–69. 77. Heiss WD. Ischemic penumbra: ischemia in neurological
67. Sokoloff L. Energetics of functional evidence from functional imaging disease. Nat Med 2011; 17:
activation in neural tissues. in man. J Cereb Blood Flow Metab 439–47.
Neurochem Res 1999; 24:321–9. 2000; 20:1276–93. 86. Strong AJ, Anderson PJ, Watts
68. Magistretti PJ, Pellerin L. 78. Takasawa M, Beech JS, Fryer TD, HR, et al. Peri-infarct
Astrocytes couple synaptic activity et al. Imaging of brain hypoxia in depolarizations lead to loss of
to glucose utilization in the brain. permanent and temporary middle perfusion in ischaemic
News Physiol Sci 1999; 14:177–82. cerebral artery occlusion in the rat gyrencephalic cerebral cortex.
using 18F-fluoromisonidazole and Brain 2007; 130:995–1008.
69. Laughlin SB, Attwell D. The
metabolic cost of neural positron emission tomography: a 87. Mies G, Iijima T, Hossmann K-A.
information: from fly eye to pilot study. J Cereb Blood Flow Correlation between periinfarct
mammalian cortex. In: Metab 2007; 27:679–89. DC shifts and ischemic neuronal
Frackowiak RSJ, Magistretti PJ, 79. Kane I, Sandercock P, Wardlaw J. damage in rat. Neuroreport 1993;
Shulman RG, Altman JS, Adams Magnetic resonance perfusion 4:709–11.
M, eds. Neuroenergetics: Relevance diffusion mismatch and 88. Moskowitz MA, Lo EH, Iadecola
for Functional Brain Imaging. thrombolysis in acute ischaemic C. The science of stroke:
Strasbourg: HFSP Workshop XI; stroke: a systematic review of the mechanisms in search of
2001: 54–64. evidence to date. J Neurol Neurosurg treatments. Neuron 2010;
70. Frackowiak RSJ, Magistretti PJ, Psychiatry 2007; 78:485–91. 67:181–98.
Shulman RG, Altman JS, Adams 80. Hoehn-Berlage M, Norris DG, 89. Tymianski M. Emerging
M (eds.). Neuroenergetics: Kohno K, et al. Evolution of mechanisms of disrupted cellular
Relevance for Functional Brain regional changes in apparent signaling in brain ischemia. Nat
Imaging. Strasbourg: HFSP diffusion-coefficient during focal Neurosci 2011; 14:1369–73.
Workshop XI; 2001. ischemia of rat brain: the 90. Simon R, Xiong Z. Acidotoxicity
relationship of quantitative in brain ischaemia. Biochem Soc
71. Astrup J, Siesjö BK, Symon L.
diffusion NMR imaging to Trans 2006; 34:1356–61.
Thresholds in cerebral ischemia –
reduction in cerebral blood flow
the ischemic penumbra. Stroke 91. Choi DW. Excitotoxicity,
and metabolic disturbances.
1981; 12:723–5. apoptosis and ischemic stroke.
J Cereb Blood Flow Metab 1995;
72. Heiss WD. Experimental evidence 15:1002–11. J Biochem Mol Biol 2001; 34:8–14.
of ischemic thresholds and 92. Szydlowska K, Tymianski M.
81. Sun PZ, Zhou J, Sun W, Huang J,
functional recovery. Stroke 1992; Calcium, ischemia and
van Zijl PC. Detection of the
23:1668–72. excitotoxicity. Cell Calcium 2010;
ischemic penumbra using pH-
73. Hossmann KA. Viability weighted MRI. J Cereb Blood Flow 47:122–9.
thresholds and the penumbra of Metab 2007; 27:1129–36. 93. MacDonald JF, Xiong ZG, Jackson
focal ischemia. Ann Neurol 1994;
82. Cheung JS, Wang E, Lo EH, Sun MF. Paradox of Ca2+ signaling,
36:557–65.
PZ. Stratification of cell death and stroke. Trends
74. Heiss W-D, Hayakawa T, Waltz heterogeneous diffusion MRI Neurosci 2006; 29:75–81.
AG. Cortical neuronal function ischemic lesion with kurtosis 94. Chan PH. Reactive oxygen
during ischemia. Effects of imaging: evaluation of mean radicals in signaling and damage
occlusion of one middle cerebral diffusion and kurtosis MRI in the ischemic brain. J Cereb
artery on single-unit activity in mismatch in an animal model of Blood Flow Metab 2001; 21:2–14.
cats. Arch Neurol 1976; 33:813–20. transient focal ischemia. Stroke 95. Shuaib A, Lees KR, Lyden P, et al.
75. Hossmann KA. Cerebral 2012; 43:2252–4. NXY-059 for the treatment of
ischemia: models, methods and 83. Kidwell CS, Heiss WD. Advances acute ischemic stroke. N Engl
outcomes. Neuropharmacology in stroke: imaging. Stroke 2012; J Med 2007; 357:562–71.
2008; 55:257–70. 43:302–4. 96. Dalkara T, Moskowitz MA. The
76. Hata R, Maeda K, Hermann D, 84. del Zoppo GJ, Sharp FR, Heiss complex role of nitric oxide in the
Mies G, Hossmann KA. Dynamics WD, Albers GW. Heterogeneity in pathophysiology of focal cerebral
30 of regional brain metabolism and the penumbra. J Cereb Blood Flow ischemia. Brain Pathol 1994;
gene expression after middle Metab 2011; 31:1836–51. 4:49–57.

Downloaded from https://www.cambridge.org/core. University of Sussex Library, on 17 Apr 2018 at 16:27:57, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781107239340.002
Chapter 1: Neuropathology and pathophysiology of stroke

97. Shuttleworth CW, Weiss JH. Zinc: 108. Badaut J, Lasbennes F, Magistretti and 13NH3. Ann Neurol 1980;
new clues to diverse roles in brain PJ, Regli L. Aquaporins in brain: 8:47–60.
ischemia. Trends Pharmacol Sci distribution, physiology, and 118. Baron JC, Frackowiak RS, Herholz
2011; 32:480–6. pathophysiology. J Cereb Blood K, et al. Use of PET methods for
98. Sensi SL, Jeng JM. Rethinking the Flow Metab 2002; 22:367–78. measurement of cerebral energy
excitotoxic ionic milieu: the 109. Kerr JF, Wyllie AH, Currie AR. metabolism and hemodynamics in
emerging role of Zn(2+) in Apoptosis: a basic biological cerebrovascular disease. J Cereb
ischemic neuronal injury. Curr phenomenon with wide-ranging Blood Flow Metab 1989; 9:723–42.
Mol Med 2004; 4:87–111. implications in tissue kinetics. Br 119. Heiss WD, Grond M, Thiel A,
99. Paschen W. Endoplasmic J Cancer 1972; 26:239–57. et al. Permanent cortical damage
reticulum dysfunction in brain 110. Niizuma K, Yoshioka H, Chen H, detected by flumazenil positron
pathology: critical role of protein et al. Mitochondrial and apoptotic emission tomography in acute
synthesis. Curr Neurovasc Res neuronal death signaling stroke. Stroke 1998; 29:454–61.
2004; 1:173–81. pathways in cerebral ischemia. 120. Baird AE, Benfield A, Schlaug G,
100. Youle RJ, van der Bliek AM. Biochim Biophys Acta 2010; et al. Enlargement of human
Mitochondrial fission, fusion, and 1802:92–9. cerebral ischemic lesion volumes
stress. Science 2012; 337:1062–5. 111. MacManus JP, Buchan AM. measured by diffusion-weighted
Apoptosis after experimental magnetic resonance imaging. Ann
101. Gouriou Y, Demaurex N, Bijlenga
stroke: fact or fashion? Neurol 1997; 41:581–9.
P, De Marchi U. Mitochondrial
calcium handling during J Neurotrauma 2000; 17:899–914. 121. Kidwell CS, Alger JR, Saver JL.
ischemia-induced cell death in 112. Dirnagl U, Simon RP, Hallenbeck Beyond mismatch: evolving
neurons. Biochimie 2011; JM. Ischemic tolerance and paradigms in imaging the
93:2060–7. endogenous neuroprotection. ischemic penumbra with
Trends Neurosci 2003; 26:248–54. multimodal magnetic resonance
102. Norenberg MD, Rao KV. The
imaging. Stroke 2003; 34:2729–35.
mitochondrial permeability 113. Zhao H, Sapolsky RM, Steinberg
transition in neurologic disease. GK. Interrupting reperfusion as a 122. Heiss WD, Sobesky J, Smekal U,
Neurochem Int 2007; 50:983–97. stroke therapy: ischemic et al. Probability of cortical
postconditioning reduces infarct infarction predicted by flumazenil
103. Rothwell NJ, Luheshi GN.
size after focal ischemia in rats. binding and diffusion-weighted
Interleukin 1 in the brain: biology,
J Cereb Blood Flow Metab 2006; imaging signal intensity: a
pathology and therapeutic
26:1114–21. comparative positron emission
target. Trends Neurosci 2000;
tomography/magnetic resonance
23:618–25. 114. Wiltrout C, Lang B, Yan Y, imaging study in early ischemic
104. Planas AM, Gorina R, Chamorro Dempsey RJ, Vemuganti R. stroke. Stroke 2004; 35:1892–8.
A. Signalling pathways mediating Repairing brain after stroke: a
123. Kane I, Carpenter T, Chappell F,
inflammatory responses in brain review on post-ischemic
et al. Comparison of 10 different
ischaemia. Biochem Soc Trans neurogenesis. Neurochem Int
magnetic resonance perfusion
2006; 34:1267–70. 2007; 50:1028–41.
imaging processing methods in
105. Wang CX, Shuaib A. Critical role 115. Kamouchi M, Ago T, Kuroda J, acute ischemic stroke: effect on
of microvasculature basal lamina Kitazono T. The possible roles of lesion size, proportion of patients
in ischemic brain injury. Prog brain pericytes in brain ischemia with diffusion/perfusion
Neurobiol 2007; 83:140–8. and stroke. Cell Mol Neurobiol mismatch, clinical scores, and
2012; 32:159–65. radiologic outcomes. Stroke 2007;
106. Walz B, Zimmermann C, Bottger
S, Haberl RL. Prognosis of 116. Abe K, Yamashita T, Takizawa S, 38:3158–64.
patients after hemicraniectomy in et al. Stem cell therapy for cerebral 124. Sobesky J, Weber OZ, Lehnhardt
malignant middle cerebral artery ischemia: from basic science to FG, et al. Does the mismatch
infarction. J Neurol 2002; clinical applications. J Cereb match the penumbra? Magnetic
249:1183–90. Blood Flow Metab 2012; 32: resonance imaging and positron
1317–31. emission tomography in early
107. Lansberg MG, Thijs VN, O’Brien
MW, et al. Evolution of apparent 117. Kuhl DE, Phelps ME, Kowell AP, ischemic stroke. Stroke 2005;
diffusion coefficient, diffusion- et al. Effects of stroke on local 36:980–5.
weighted, and T2-weighted signal cerebral metabolism and 125. Olivot JM, Albers GW. Diffusion-
intensity of acute stroke. AJNR Am perfusion: mapping by emission perfusion MRI for triaging 31
J Neuroradiol 2001; 22:637–44. computed tomography of 18FDG transient ischemic attack and

Downloaded from https://www.cambridge.org/core. University of Sussex Library, on 17 Apr 2018 at 16:27:57, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781107239340.002
Section 1: Etiology, pathophysiology, and imaging

acute cerebrovascular syndromes. M, eds. Neuroenergetics: Relevance 130. Hossmann KA, Mies G.
Curr Opin Neurol 2011; 24:44–9. for Functional Brain Imaging. Multimodal Mapping of the
126. Garcia JH, Liu KF, Ho KL. Strasbourg: HFSP Workshop XI; Ischemic Penumbra in Animal
Neuronal necrosis after middle 2001: 133–42. Models. New York: Marcel
cerebral artery occlusion in Wistar 128. Attwell D, Laughlin SB. An energy Dekker, Inc.; 2007.
rats progresses at different time budget for signaling in the grey 131. Hossmann KA. Periinfarct
intervals in the caudoputamen matter of the brain. J Cereb Blood depolarizations. Cerebrovasc
and the cortex. Stroke 1995; Flow Metab 2001; 21:1133–45. Brain Metab Rev 1996; 8:
26:636–42. 129. Heiss WD, Rosner G. Functional 195–208.
127. Magistretti PJ. Coupling synaptic recovery of cortical neurons as 132. Hossmann KA. Pathophysiology
activity to glucose metabolism. In: related to degree and duration of and therapy of experimental
Frackowiak RSJ, Magistretti PJ, ischemia. Ann Neurol 1983; stroke. Cell Mol Neurobiol 2006;
Shulman RG, Altman JS, Adams 14:294–301. 26:1057–83.

32

Downloaded from https://www.cambridge.org/core. University of Sussex Library, on 17 Apr 2018 at 16:27:57, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781107239340.002

You might also like