Download as pdf or txt
Download as pdf or txt
You are on page 1of 6

Brain Hemorrhages 1 (2020) 13–18

Contents lists available at ScienceDirect

Brain Hemorrhages
CHINESE ROOTS
GLOBAL IMPACT
journal homepage: www.keaipublishing.com/en/journals/brain-hemorrhages/

Review article

Intracerebral hemorrhage in translational research


Ruiyi Zhang a, Qian Bai a, Yang Liu a, Yan Zhang a, Zhaofu Sheng a, Mengzhou Xue a,⇑, V. Wee Yong b,⇑
a
The Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Henan Joint International Laboratory of Intracerebral Hemorrhagic Brain
Injury and Henan Medical Key Laboratory of Translational Cerebrovascular Diseases, Zhengzhou, Henan, China
b
Hotchkiss Brain Institute and Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada

a r t i c l e i n f o a b s t r a c t

Article history: Intracerebral hemorrhage (ICH) is a serious stroke subtype with high morbidity and mortality. The prog-
Received 25 December 2019 nosis of ICH is poor. In recent years, there have been many studies on how to improve the prognosis of
Received in revised form 10 February 2020 ICH. This article mainly summarizes the research progress of ICH in translational research, including its
Accepted 11 February 2020
risk factors and pathogenesis, course of disease, primary and secondary ICH brain injury, its prevention
Available online 28 February 2020
and treatment strategies.
Ó 2020 Production and hosting by Elsevier B.V. on behalf of KeAi Communications Co., Ltd. This is an open
Keywords:
access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
Intracerebral hemorrhage
Brain injury
Neuroinflammation
Neuroimmunology
Neuronal death

Intracerebral hemorrhage (ICH) can be induced by a variety of uals, especially in the elderly, amyloid angiopathy is an important
causes, including hypertension, cerebral amyloid angiopathy, brain cause of ICH. The deposition of amyloid protein in the media and
trauma, vascular malformations and drugs. The study of global dis- adventitia of capillaries, arterioles, cortex and pia meningeal arter-
ease burden showed that the number of ICH increased by 47% in ies causes vascular wall brittleness. The unique features of this
past 20 years, mostly gathered in low income and middle-income lesion are that hemorrhage tends to occur in the lobes of the brain
countries.1 Understanding the pattern of brain injury induced by (especially in the posterior part of the brain), multifocal and recur-
ICH is a great significance to find the effective treatment. Here rent.3 The traditional view believes that the difference between
we summarize the research progress of ICH from the view of trans- hypertensive vascular disease and amyloid vascular disease lies
lational medicine. in the site of bleeding. ICH associated with amyloid angiopathy is
usually located in the lobe, and the hypertensive ICH is usually
located in the deep part of the brain. However, hypertension can
1. Risk factors and pathogenesis of ICH
also lead to lobar hemorrhage, and hypertensive vascular disease
and amyloid vascular disease may be a co-disease, so the patho-
Hypertension is the most common risk factor for ICH. About 80%
genesis of ICH is sometimes difficult to determine.
of ICH patients have elevated blood pressure on admission, and
Smoking, alcohol abuse, low total and low-density lipoprotein
most of them have a history of hypertension.2 Long-term hyperten-
cholesterol levels all increased the risk of ICH. Warfarin increases
sion makes cerebral arterioles suffer higher stress of blood flow,
the risk of ICH by 2–5 times, and the risk of bleeding depends on
triggers smooth muscle cell proliferation and death followed by
anticoagulant strength. The increase in the number of elderly peo-
collagen replacement, weakens the strength of artery wall, and
ple taking oral anticoagulant drugs has led to an increasing number
makes it easy to occlusion and rupture. Perforating arteries such
of anticoagulant-related ICH. Antiplatelet therapy also increases
as bean striation, thalamus and brainstem perforating artery bifur-
the risk of ICH. Although several case-control studies did not prove
cation are the most common sites of bleeding, and the degree of
that the use of antiplatelet drugs increased the risk of ICH, Meta-
bleeding mainly depends on the size of arteriole wall space, blood
analysis showed that platelet therapy slightly significantly
pressure and hemostatic mechanism. In non-hypertensive individ-
increased the risk of ICH. In addition, Meta-analysis showed that
the history of antiplatelet drugs increased the risk of death after
⇑ Corresponding authors at: 2 Jingba Road, Zhengzhou, Henan 450001, China
ICH, and another study showed that the history of antiplatelet
(M. Xue).
drugs was associated with an increase in early hematoma in ICH.
E-mail addresses: xuemengzhou@zzu.edu.cn (M. Xue), vyong@ucalgary.ca
(V.W. Yong). Compared with antiplatelet monotherapy, double anti-platelet

https://doi.org/10.1016/j.hest.2020.02.003
2589-238X/Ó 2020 Production and hosting by Elsevier B.V. on behalf of KeAi Communications Co., Ltd.
This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
14 R. Zhang et al. / Brain Hemorrhages 1 (2020) 13–18

therapy may further increase the risk of ICH. In patients with atrial resulting in cerebral hernia and slow blood flow velocity, which
fibrillation, the risk of ICH in the aspirin combined with clopidogrel may lead to a further decrease in blood flow if the supply of collat-
group was almost twice as high as that in the aspirin alone eral circulation in the brain area supplied by ruptured blood ves-
group.4,5 sels is poor. However, few clinical and animal studies have
There have been many reports of associations between sympa- reported whether changes in blood flow around the hematoma
thetic drugs such as cocaine, heroin, amphetamine and ephedrine can lead to ischemic injury. It should be noted that the changes
with ICH, especially in young patients. Especially in women, rela- of blood flow velocity around the hematoma mix metabolism
tively high doses of phenylpropanolamine are an independent fac- and hematoma formation and other factors. In ICH patients, the
tor for ICH, and low doses of phenylpropanolamine in cold cures is blood flow and oxygen metabolic rate around the hematoma
also associated with ICH risk.6,7 In addition, chronic kidney disease decreased, resulting in the decrease of oxygen uptake fraction,
increases the risk of ICH, and chronic kidney disease may be a mar- indicating that there is a low perfusion area around the hematoma.
ker of cerebral microvascular disease.4,8 Recent data have shown that the decline in brain metabolism may
reflect mitochondrial damage rather than cerebral ischemia.14
ICH may also trigger the protective defense mechanism of the
2. Progression of ICH body. Up-regulation of a variety of endogenous proteins after ICH
may help protect the brain from damage. For example, ferritin
After vascular rupture, the formation of hematoma begins and and nuclear factor 2 related factor 2 (NF-E2 related factor 2, Nrf
progresses within 60 min. The sudden influx of blood flow into 2) were significantly up-regulated and participated in the mecha-
the brain leads to mechanical destruction of the brain parenchyma, nism of antioxidant defense. In Nrf2 knockout mice, collagenase
distortion and displacement of the brain tissue, increases the risk leads to more brain injury by inducing ICH than that in wild-type
of cerebral hernia and ischemia, and triggers brain cell death mice.14
including necrosis and apoptosis, inflammation and edema.9 About
1/3 of the patients had enlarged hematoma within 3 h, and about
2/3 of them occurred within 1 h after ICH.10 Even without the coag- 3. Primary brain injury induced by ICH and its prevention and
ulation disorders, the hematoma will increase too, and the mecha- treatment strategy
nism of hematoma increase is not clear, which may be related to
the continued bleeding at the initial bleeding site and the satellite ICH firstly destroys the brain structure. Hematoma may
bleeding around the blood clot caused by the destruction of adja- increase intracranial pressure, oppress brain areas, potentially
cent small blood vessels. Alcohol abuse, irregular bleeding, low affect blood flow and cause cerebral hernia. In view of the physical
levels of fibrinogen and prothrombin, diabetes and liver diseases compression of hematoma, many clinical trials have explored the
are all risk factors for hematoma enlargement.3 Computed tomo- effect of surgical removal of blood clots, but no clinical trials have
graphic angiography (CTA) can predict the risk of hematoma confirmed the benefits of ICH patients. One possible explanation is
enlargement. The spot sign of CTA is highly correlated with hema- that the side effects of surgery offset the beneficial effects of blood
toma enlargement.10 CTA spot sign was first described in 2007, it clot removal. It is worth noting that the location of ICH affects the
was defined as focal or multifocal enhancer spillover from normal prognosis, and surgical decompression can save lives in patients
or abnormal blood vessels around hematoma after ICH, which pre- with cerebellar hemorrhage. Because it is more difficult to remove
dicted hematoma enlargement and became an independent indica- hematoma in small animals, there are relatively few preclinical
tor of poor clinical prognosis.11 Subsequent large clinical studies studies on the effect of removing blood clots, and most studies
found that about 30% of ICH patients had spot sign. Multiple CTA use pigs, which confirms that early removal of hematoma may
tests could further help predict hematoma enlargement, and the benefit, but there are many problems with ultra-early clearance
clinical prognosis of patients with spot sign within 23 s of enhancer of blood clots. In clinical, some patients will continue to bleed or
injection was poor.12 Intraventricular hemorrhage (IVH) can occur easily bleed again.10 Moreover, a clinical trial shows that removing
with ICH or within 24–72 h after ICH, accounting for about 20%– acute intracerebral hematomas in early phase by minimally inva-
55% of patients with ICH, which is another important factor in sive surgery which has been widely used recently is safe and effec-
the poor prognosis of ICH. Compared with patients without IVH, tive to bring better neurological prognosis in ICH patients.15
the prognosis of patients with IVH was worse.13 Preventing hematoma enlargement may be another way to
ICH patients developed brain edema within a few hours after reduce the space occupying effect. Many patients suffered rapid
the onset of symptoms and reached the peak at 1–2 weeks. The elevated blood pressure after ICH which could result in further
blood composition is closely related to the brain edema around bleeding and edema. Some clinical trials exhibit that although
the hematoma. there are no differences between intensive lowering of systolic
Within 1 h after ICH, the blood clot contracts and the plasma pressure under 140 mmHg and under 180 mmHg as guideline in
move out from the clot to the tissue around the hematoma, form- death or severe disability, rapid reduction of the blood pressure
ing edema adjacent to hematoma. 24 h after bleeding, thrombin may contributes to improved functional outcomes.16,17 And many
cascade reaction and thrombin production are involved in the for- studies have focused on finding drugs that change the process of
mation of early edema. Erythrocyte lysis, hemoglobin and its coagulation cascade or fibrinolysis. Clinical trials have confirmed
decomposition products, and carbonic anhydrase-1, another com- that activated coagulation factor VII (VIIa), which can reduce
ponent of erythrocytes, lead to late edema. The blood–brain barrier hematoma enlargement but cannot improve prognosis and may
(BBB) remained intact within a few hours after ICH, but the perme- induce thromboembolic complications. There are also some clini-
ability of BBB increased after 8–12 h. Although there are many cal trials to explore which patients are suitable for the treatment
forms of edema after ICH, angiogenic edema is the main form of of activated coagulation factor VIIa.18 Some studies have shown
ICH.10 The imaging findings of brain edema were low density on that patients with enlarged hematoma or patients with ICH who
CT scan and high signal on T2 weighted or Flair of magnetic reso- take anticoagulant or antiplatelet drugs benefit the most. Other
nance imaging (MRI). The most severe edema was located around methods are also being tested clinically, such as platelet transfu-
the clot, mainly along the white matter.3 sion in patients receiving antiplatelet therapy, or the use of amino-
The degree of cerebral ischemia around hematoma after ICH is caproic acid, an antifibrinolytic agent. Early platelet transfusion
still controversial. Large hematoma increases intracranial pressure, can benefit from low platelet activity or antiplatelet therapy. In
R. Zhang et al. / Brain Hemorrhages 1 (2020) 13–18 15

the rat model of ICH induced by collagenase, activated coagulation activated receptors (PARs). PAR-1, PAR-3 and PAR-4 are thrombin
factor VIIa reduced early hematoma enlargement.19 Vasodilator in receptors.24 It has been found that neurons and astrocytes express
plasma can inhibit platelet aggregation and inhibit vasodilator can mRNA of thrombin receptors and the immunoreactivity of which in
reduce the enlargement of hematoma. In the mouse ICH model human brain. Studies have shown that PAR-1 mediates some
induced by warfarin anticoagulant and collagenase, prothrombin pathological effects of thrombin and participates in the pathophys-
complex concentrate and frozen plasma could reduce bleeding, iological mechanism induced by ICH.25 Infusion of high doses of
and the effects of activated coagulation factor VIIa and tranexamic thrombin directly into the brain can cause inflammatory cell infil-
acid were poor.14,20 tration, mesenchymal cell proliferation, scar formation, brain
The increase of blood pressure after ICH is the result of the edema and seizures.23,24,26,27 Thrombin acts on many cell types,
superposition of many mechanisms, including pre-onset hyperten- including endothelial cells, which leads to the destruction of BBB
sion, increased intracranial pressure, autonomic nerve and neu- and the formation of brain edema. Neurons and astrocytes are acti-
roendocrine activation. Clinical trials to reduce hematoma also vated by Src kinase and can be killed by high concentration of
include lowering blood pressure. There is a dilemma in the treat- thrombin in vitro. Microglia can be activated by thrombin. Animal
ment of elevated blood pressure. On the one hand, acute hyperten- experiments showed that inhibition of thrombin by hirudin, a
sion may be protective, ensuring cerebral blood flow supply in the specific thrombin inhibitor,23,24,27 could reduce the damage
case of elevated intracranial pressure to prevent ischemic stroke, induced by ICH. Clinical trials have also confirmed that thrombin
on the other hand, hypertension increases the risk of edema. Pro- mediated brain injury. Argatroban, a thrombin inhibitor, can signif-
mote hematoma enlargement by continuing bleeding and rebleed- icantly reduce brain edema around the hematoma following ICH.
ing. According to the American Heart and Stroke Association In addition, there is evidence that thrombin is associated with
guidelines, blood pressure is safe at 150–220 mmHg after ICH. brain recovery and neuronal regeneration after ICH.27–29
Some clinical trials have confirmed that the target value of blood There are obvious inflammatory reactions after ICH, including
pressure reduction within 1 h after ICH is not less than 140 mmHg, microglia activation, leukocyte infiltration and inflammatory
which has no obvious negative effect on neural state and has noth- mediator formation, which play an important role in ICH-induced
ing to do with serious adverse events. Recent clinical trials have injury and brain recovery.30 In ICH animal model, the activation
shown that the target value of hypotension at 110 mmHg is still of microglia was an early reaction, which appeared within 1 h after
safe.21 It is not known whether antihypertensive treatment has ICH, reached the peak at 3–7 days after ICH, and maintained for 3–
long-term benefits, but there is evidence that it can reduce hema- 4 weeks.23,24,26 Tuftsin or minocycline plays a protective role after
toma enlargement. There are few preclinical studies on the effect ICH by inhibiting microglia activation.31–33 A meta-analysis includ-
of blood pressure on hematoma enlargement. After injection of col- ing 4 clinical trials and 14 rodent studies displays therapeutic
lagenase, there was no significant difference in bleeding volume effects on acute ischemic stroke. But the study about minocycline
between spontaneously hypertensive rats and normal blood pres- treatment on ICH remains less. A pilot study of minocycline in
sure rats. However, the injury induced by ICH in spontaneously ICH patients shows that 400 mg dose can produce neuroprotective
hypertensive rats is more serious, indicating that hypertension effects,34 but further study in different doses or usage still need to
does not mediate ICH-induced injury by regulating the volume of be done. For brain injury after ICH, microglia/macrophage are a
hematoma. Rapid changes in blood pressure may have a greater double-edged sword, which can promote inflammation reactions
impact on hematoma enlargement than persistent hypertension. leading secondary injury or mediate inflammation and dissolve
Pathological examination of ICH rat model showed that there blood clots,22 and the net effect may change with time.35 Neu-
was no significant difference in bleeding volume between sponta- trophils are the first leukocytes to enter the brain after ICH. Neu-
neously hypertensive rats and normal hypertensive rats, but there trophils participate in ICH-induced brain injury by producing
was more bleeding in normotensive rats with sharply elevated reactive oxygen species (ROS), pro-inflammatory proteases and
blood pressure.14 destroying BBB. Monocytes also enter the brain after ICH, and
Many studies have focused on the application of drug-enhanced recent studies have shown that consumption of neutrophils
defense mechanisms. Nrf2 can be up-regulated by a range of drugs, reduces the number of monocytes entering the brain. Toll-like
including sulforaphane, a component of broccoli, which reduces receptor 4 on leukocytes mediates the infiltration of neutrophils
ICH-induced brain injury in rats and mice through a Nrf2- and monocytes. The two types of cells that have received little
dependent mechanism. Peroxisome proliferator-activated attention in the ICH study are mast cells and infiltrating lympho-
receptor-c (PPAR-c) agonists can also play a beneficial role by cytes in the brain. Recent studies have shown that the both may
up-regulating cellular defense mechanisms, including hydrogen mediate ICH-induced brain injury.31 In animals and humans, brain
peroxide.14 injury induced by ICH is associated with the significant upregula-
tion of a variety of inflammatory mediators, such as tumor necrosis
factor-a (TNF-a) and interleukin-1 b (IL-1b), chemokines, adhesion
4. Secondary brain injury induced by ICH and its prevention and molecules, and matrix metalloproteinase (MMPs) such as MMP-9
treatment strategy and -3.27,29,36,37 The secondary reaction of inflammation after ICH
leads to the destruction of BBB, which can aggravate inflammation
Secondary brain injury after ICH may be caused by a series of by promoting leukocyte infiltration. ROS, pro-inflammatory cytoki-
events induced by primary injury, including body/tissue responses nes, chemokines and MMPs from leukocytes are all involved in the
to hematoma, such as inflammation, microglia/macrophage activa- destruction of BBB.38 In addition to promoting inflammation, the
tion and neutrophil infiltration,22 release of clot components (e.g. destruction of BBB also leads to the formation of vascular edema
hemoglobin/iron).14 Following factors are involved in secondary after ICH. There have been clinical trials to verify whether
brain injury induced by ICH (Fig. 1). cyclooxygenase inhibitor celecoxib, anti-inflammatory hypo-
The initial response of the body to ICH is the activation of the glycemic drug pioglitazone can reduce brain injury after ICH, but
hemostatic mechanism to limit bleeding. Thrombin plays an no clear conclusion has been drawn. Some clinical trials focused
important role in homeostasis, but there is also a lot of evidence on two kinds of lipid-lowering drugs, rosuvastatin and simvastatin,
that thrombin can be involved in ICH-induced brain injury.23 The which had anti-inflammatory effects. Rosuvastatin had a positive
most important role of thrombin is to break down fibrinogen into effect, but simvastatin trial could not continue because of difficulty
fibrin, while the other effects are mediated by three protease- in recruiting volunteers.39,40
16 R. Zhang et al. / Brain Hemorrhages 1 (2020) 13–18

Fig. 1. In the earliest stage of ICH, the primary brain injury made by hematoma and edema causes blood products (Fe2+, Hb, thrombin) to leak into the damage area to
activate microglia/macrophages and other peripheral immune cells to express high levels of IL-6, IL-1b, TNFa, GMCSF, INFc, ROS, RNS, CCLs, HO-1, and MMPs. These changes
result in the secondary brain injury, which extend the brain damage such as brain edema, cell death, blood–brain barrier disruption, and neurologic deficits.

The complement system involves immune responses, including injuries in ICH rats and piglets,44,45 and deferoxamine mesylate
cell lysis and inflammation. The plasma protein components of the has been used in clinical trials.46 Heme oxygenase breaks down
complement system are usually blocked out of the brain by the blood into bilirubin, carbon dioxide and iron. Inhibition of heme
BBB, but they may enter the brain after ICH as part of the blood oxygenase or knockout serum heme oxygenase-1 can reduce brain
or after BBB is damaged. Membrane attack complex (MAC, C5b- damage caused by ICH.47 One possible mechanism of tissue dam-
9) is formed after ICH and complement activation. MAC partici- age caused by iron is the production of free radicals. Free radical
pates in the cleavage of red blood cells and mediates the release mediated ICH injury, animal experiments showed that free radical
of hemoglobin and iron, resulting in damage to surrounding tis- scavengers reduce the damage induced by ICH. However, it should
sues. It may also cause direct damage to neurons, glial cells and be pointed out that there may be a variety of sources of free radi-
blood vessels around the hematoma. In addition, complement cas- cals after ICH. NXY-059 is a free radical spin trap used in clinical
cade activation produces C3a and C5a, leukocyte chemokines, trials in patients with ICH.48 It has not been confirmed whether
microglia and mast cell activators, which promotes inflammation patients benefit or not. The cause of this negative result has not
after ICH. Several studies have confirmed that consumption of yet been determined, but it may reflect that the increase in the per-
complements using antagonists or gene knockout can reduce meability of BBB after ICH is not sufficient to neutralize a large
ICH-induced brain injury.41 However, the conclusions on C5 are number of free radicals.49 Although hemoglobin is the main com-
not consistent. C5a inhibitors are protective, while the brain injury ponent of red blood cells, it is not the only one. A recent study
induced by ICH in C5 gene deficient mice is significantly increased, has shown that intracerebral injection of carbonic anhydrase 1,
which may reflect the compensatory changes in C5 gene deficient another major component of red blood cells, may lead to brain
mice. Similar to inflammation, although complement activation damage, and carbonic anhydrase inhibitors reduce ICH-induced
after ICH promotes brain damage, there is also evidence that com- damage in rats.50
plement is beneficial to the long-term recovery of the brain.42 Although glutamate-induced excitability plays an important
There is growing evidence that hemoglobin and iron released role in cell death after cerebral ischemia, there is some evidence
from hematoma are the main causes of brain damage caused by that glutamate may also be involved in ICH-induced brain injury
ICH. Injection of lysed red blood cells into the rodent brain induces and through specific potential mechanisms. The primary hemor-
brain damage, and infusion of hemoglobin and iron into the brain rhage leads to glutamate outflow, and the production of thrombin
has a similar effect. After ICH, iron accumulates in the tissues after ICH leads to the activation of Src kinase. Src kinase can phos-
around the hematoma. In rat and pig models, the use of iron chela- phorylate NMDA receptor and enhance its function. Compared
tor deferoxamine can reduce brain damage induced by ICH.43 with cerebral ischemia, there are few clinical trials to verify the
Deferoxamine has shown protective effects and reduction of role of glutamate receptor antagonists in ICH, except for a small
R. Zhang et al. / Brain Hemorrhages 1 (2020) 13–18 17

clinical trial of NMDA antagonist CP-101606, which focuses on 6. Conclusion


traumatic brain injury.51,52
ICH causes brain cell death and brain atrophy around the hema- In summary, our understanding of the mechanism of brain
toma. The pathway of cell death involves apoptosis, necrosis and damage caused by ICH has increased over the past 20 years, and
autophages, which the dominant way is controversial. The necrosis the conduct of many clinical trials offers hope for the relief of sev-
of the brain around the blood clot may be related to the mechanical ere ICH. However, it should be noted that ICH covers a variety of
pressure of hematoma, the composition of blood clot and its degra- hematoma in different locations and sizes. As a result, there is
dation. Although apoptosis is associated with peri-hematoma brain probably no treatment that is most suitable for all patients.77 For
cell death, the importance of apoptosis in ICH-induced brain injury different individuals, the combination of different treatments
is not clear.10 A large number of studies have used ICH model to may provide the best way to alleviate ICH brain injury.77,78
find possible ways to reduce apoptosis and brain injury, such as
taurodeoxycholic acid, membrane stabilizer cytidine diphosphate Declaration of Competing Interest
choline.14 There is also evidence that autophagy occurs after
ICH.14 Autophagy is the degradation process of cells. Proteins and The authors declare that they have no known competing finan-
organelles are isolated in bilayer vesicles and transported to lyso- cial interests or personal relationships that could have appeared
somes, which are digested by lysosomal hydrolases. ICH can induce to influence the work reported in this paper.
programmed cell death in the form of autophagy, and iron plays an
important role in the process of autophagy.53 As with other neuro- Acknowledgements
logical diseases, it is necessary to further confirm whether autop-
hagy is protective (removing dead cells) or nociceptive (inducing The authors acknowledge operating grant support from the
multiple cell deaths).54 In clinical and animal experiments, cell National Natural Science Foundation of China (grants nos.:
death after ICH can lead to brain atrophy.10,55 81870942, 81471174 and 81520108011), National Key Research
and Development Program of China (grant no.: 2018
YFC1312200), and Innovation Scientists and Technicians Troop
5. From bench to clinic Constructions Projects of Henan Province of China (for MX); and
from the Canadian Institutes of Health Sciences (VWY).
Due to absence of effective treatment for ICH,56 many research-
ers devote into discover new therapeutic targets and pathophysiol-
ogy with the help of different animal models including References
microballoon insertion,57 autologous whole blood injection,58 col-
lagenase,59 thrombin,23 hypertensive stroke model,60 neonatal 1. Kang D-W. Intracerebral hemorrhage: large disease burden but less therapeutic
PVH/IVH61 and other newly developed models. But none of these progress. J Stroke. 2017;19:1–2.
2. Sessa M. Intracerebral hemorrhage and hypertension. Neurol Sci. 2008;29:258.
animal models can completely mimic the condition of human 3. Godoy DA, Piñero GR, Koller P, Masotti L, Di Napoli M. Steps to consider in the
ICH. Each of them has advantages and disadvantages. Autologous approach and management of critically ill patient with spontaneous
blood injection model may imitate the effects of hematoma most intracerebral hemorrhage. World J Crit Care Med. 2015;4:213.
4. An SJ, Kim TJ, Yoon B-W. Epidemiology, risk factors, and clinical features of
accurately but cannot reproduce the process of bleeding.62 Collage- intracerebral hemorrhage: an update. J Stroke. 2017;19:3.
nase can disrupt capillary basal lamina and cause bleeding,63 but it 5. Suzuki Y, Sato T, Sakuma J, et al. Intracranial hemorrhage and platelet
is artificial and may lead to direct damage of brain tissue besides transfusion after administration of anti-platelets agents: Fukushima
Prefecture. Fukushima J Med Sci. 2016;2015–2026.
the effects of hemorrhage.64 Thrombin released after ICH activates 6. Tormoehlen LM, Blatsioris AD, Moser EA, et al. Disparities and guideline
microglia and promotes cytokine production that causes neuroin- adherence in drugs of abuse screening in intracerebral hemorrhage. Neurology.
flammation and brain cell death, but this model may be not suit- 2017;88:252–258.
7. Klys M, Konopka T, Rojek S. Intracerebral hemorrhage associated with
able for research other than effects of thrombin. Hence, an amphetamine. J Anal Toxicol. 2005;29:577–581.
animal model needs to be selected deliberately according to the 8. Kim JK, Shin JJ, Park SK, Hwang YS, Kim TH, Shin HS. Prognostic factors and
purpose and condition of a preclinical research of ICH. clinical outcomes of acute intracerebral hemorrhage in patients with chronic
kidney disease. J Korean Neurosurg Soc. 2013;54:296.
Many preclinical studies focused on inflammation response
9. Brouwers HB, Greenberg SM. Hematoma expansion following acute
after ICH and as the most important immune cells in central nerval intracerebral hemorrhage. Cerebrovasc Dis. 2013;35:195–201.
system, microglia/macrophage are very popular and promising.65 10. Xi G, Strahle J, Hua Y, Keep RF. Progress in translational research on
As a double side’s sword, activated pro-inflammatory microglia intracerebral hemorrhage: is there an end in sight?. Prog Neurobiol.
2014;115:45–63.
increase their production of numerous proinflammatory and 11. Wada R, Aviv RI, Fox AJ, et al. CT angiography ‘‘spot sign” predicts hematoma
potentially neurotoxic mediators which may contribute to ICH expansion in acute intracerebral hemorrhage. Stroke. 2007;38:1257–1262.
neuronal injury while increasing evidence indicates that activated 12. Bates T, Phatourous C, Van Heerden J. Spot sign, prognosis and intracerebral
haemorrhage. QJM. 2017;110:51–52.
microglia in the later phase of ICH phagocytose/resorb hematoma 13. Chen S, Zeng L, Hu Z. Progressing haemorrhagic stroke: categories, causes,
and resolve edema, contributing to improved white matter integ- mechanisms and managements. J Neurol. 2014;261:2061–2078.
rity, repair and functional recovery.66 Some promising drugs tar- 14. Keep RF, Hua Y, Xi G. Intracerebral haemorrhage: mechanisms of injury and
therapeutic targets. Lancet Neurol. 2012;11:720–731.
geting on shift the balance between pro-inflammatory and 15. Vespa P, Hanley D, Betz J, et al. ICES (intraoperative stereotactic computed
regulatory phenotypes of microglia/macrophage after ICH achieved tomography-guided endoscopic surgery) for brain hemorrhage: a multicenter
therapeutic effects in preclinical or clinical trials. Some of these randomized controlled trial. Stroke. 2016;47:2749–2755.
16. Anderson CS, Heeley E, Huang Y, et al. Rapid blood-pressure lowering in
medications focus on inhibit the generation of pro-inflammatory patients with acute intracerebral hemorrhage. N Engl J Med.
cells including minocycline,67 C5aRA, rCTRP9, NLRP3 blocker,68 2013;368:2355–2365.
LTB4 receptor antagonist, TLR4 inhibitors,69 Fingolimod70 and 17. Qureshi AI, Palesch YY, Barsan WG, et al. Intensive blood-pressure lowering in
patients with acute cerebral hemorrhage. N Engl J Med. 2016;375:1033–1043.
bortezomib71 while others dedicate to promote the transformation
18. Diringer MN, Skolnick BE, Mayer SA, et al. Thromboembolic events with
of regulatory microglia/macrophage such as SIPR agonists,72 recombinant activated factor VII in spontaneous intracerebral hemorrhage:
Statins,73 CB2R agonist, PPARc activators,74 mTOR inhibiters75 results from the factor seven for acute hemorrhagic stroke (FAST) trial. Stroke.
and sinomenine.76 All of these promising candidates still need to 2010;41:48–53.
19. Kawai N, Nakamura T, Early Nagao S. hemostatic therapy using recombinant
be further discovered in mechanism and tested in usage and doses factor VIIa in a collagenase-induced intracerebral hemorrhage model in
in both laboratory and clinical situation. ratsBrain Edema XIII. Springer; 2006:212–217.
18 R. Zhang et al. / Brain Hemorrhages 1 (2020) 13–18

20. Illanes S, Zhou W, Schwarting S, Heiland S, Veltkamp R. Comparative 50. Guo F, Hua Y, Wang J, Keep RF, Xi G. Inhibition of carbonic anhydrase reduces
effectiveness of hemostatic therapy in experimental warfarin-associated brain injury after intracerebral hemorrhage. Transl Stroke Res. 2012;3:130–137.
intracerebral hemorrhage. Stroke. 2011;42:191–195. 51. Sharp F, Liu DZ, Zhan X, Ander BP. Intracerebral hemorrhage injury
21. Lattanzi S, Silvestrini M. Blood pressure in acute intra-cerebral hemorrhage. mechanisms: glutamate neurotoxicity, thrombin, and Src. Acta
Ann Transl Med. 2016;4. Neurochirurgica Suppl. 2008;105:43–46.
22. Bai Q, Xue M, Yong VW. Microglia and macrophage phenotypes in intracerebral 52. Bullock MR, Merchant RE, Carmack CA, et al. An open-label study of CP-101,606
haemorrhage injury: therapeutic opportunities. Brain. 2020. https://doi.org/ in subjects with a severe traumatic head injury or spontaneous intracerebral
10.1093/brain/awz393, pii: awz393 [Epub ahead of print]. hemorrhage. Ann NY Acad Sci. 1999;890:51–58.
23. Xue M, Del Bigio MR. Injections of blood, thrombin, and plasminogen more 53. Chen TY, Lin CL, Wang LF, Tsai KL, Lin JY, Hsu C. Targeting GPER1 to suppress
severely damage neonatal mouse brain than mature mouse brain. Brain Pathol. autophagy as a male-specific therapeutic strategy for iron-induced striatal
2005;15:273–280. injury. Sci Rep. 2019;9:6661.
24. Xue M, Balasubramaniam J, Parsons KA, McIntyre IW, Peeling J, Del Bigio MR. 54. Durocher M, Ander BP, Jickling G, et al. Inflammatory, regulatory, and
Does thrombin play a role in the pathogenesis of brain damage after autophagy co-expression modules and hub genes underlie the peripheral
periventricular hemorrhage?. Brain Pathol. 2005;15:241–249. immune response to human intracerebral hemorrhage. J Neuroinflammation.
25. Xue M, Hollenberg MD, Demchuk A, Yong VW. Relative importance of 2019;16:56.
proteinase-activated receptor-1 versus matrix metalloproteinases in 55. Li Q, Weiland A, Chen X, et al. Ultrastructural characteristics of neuronal death
intracerebral hemorrhage-mediated neurotoxicity in mice. Stroke. and white matter injury in mouse brain tissues after intracerebral hemorrhage:
2009;40:2199–2204. coexistence of ferroptosis, autophagy, and necrosis. Front Neurol. 2018;9:581.
26. Xue M, Del Bigio MR. Acute tissue damage after injections of thrombin and 56. Qureshi AI, Tuhrim S, Broderick JP, Batjer HH, Hondo H, Hanley DF.
plasmin into rat striatum. Stroke. 2001;32:2164–2169. Spontaneous intracerebral hemorrhage. N Engl J Med. 2001;344:1450–1460.
27. Xue M, Hollenberg MD, Yong VW. Combination of thrombin and matrix 57. Sinar E, Mendelow AD, Graham DI, Teasdale GM. Experimental intracerebral
metalloproteinase-9 exacerbates neurotoxicity in cell culture and intracerebral hemorrhage: effects of a temporary mass lesion. J Neurosurg. 1987;66:568–576.
hemorrhage in mice. J Neurosci. 2006;26:10281–10291. 58. Xue M, Del Bigio MR. Intracerebral injection of autologous whole blood in rats:
28. Babu R, Bagley JH, Di C, Friedman AH, Adamson C. Thrombin and hemin as time course of inflammation and cell death. Neurosci Lett. 2000;283:230–232.
central factors in the mechanisms of intracerebral hemorrhage–induced 59. Rosenberg GA, Mun-Bryce S, Wesley M, Kornfeld M. Collagenase-induced
secondary brain injury and as potential targets for intervention. Neurosurg intracerebral hemorrhage in rats. Stroke. 1990;21:801–807.
Focus. 2012;32:E8. 60. Liard J-F, Cowley Jr AW, McCaa RE, McCaa CS, Guyton AC. Renin, aldosterone,
29. Xue M, Fan Y, Liu S, Zygun DA, Demchuk A, Yong VW. Contributions of multiple body fluid volumes, and the baroreceptor reflex in the development and
proteases to neurotoxicity in a mouse model of intracerebral haemorrhage. reversal of Goldblatt hypertension in conscious dogs. Circ Res.
Brain. 2009;132(Pt 1):26–36. 1974;34:549–560.
30. Aronowski J, Zhao X. Molecular pathophysiology of cerebral hemorrhage: 61. Goddard J, Lewis RM, Armstrong DL, Zeller RS. Moderate, rapidly induced
secondary brain injury. Stroke. 2011;42:1781–1786. hypertension as a cause of intraventricular hemorrhage in the newborn beagle
31. Xue M, Mikliaeva EI, Casha S, Zygun D, Demchuk A, Yong VW. Improving model. J Pediatr. 1980;96(6):1057–1060.
outcomes of neuroprotection by minocycline: guides from cell culture and 62. Xi G, Wagner KR, Keep RF, et al. Role of blood clot formation on early edema
intracerebral hemorrhage in mice. Am J Pathol. 2010;176:1193–1202. development after experimental intracerebral hemorrhage. Stroke.
32. Wang G, Li Z, Li S, et al. Minocycline preserves the integrity and permeability of 1998;29:2580–2585.
BBB by altering the activity of DKK1-Wnt signaling in ICH model. Neuroscience. 63. Rosenberg GA, Estrada E, Kelley RO, Kornfeld M. Bacterial collagenase disrupts
2019;415:135–146. extracellular matrix and opens blood-brain barrier in rat. Neurosci Lett.
33. Sheng Z, Liu Y, Li H, et al. Efficacy of minocycline in acute ischemic stroke: a 1993;160:117–119.
systematic review and meta-analysis of rodent and clinical studies. Front 64. Xue M, Del Bigio MR. Comparison of brain cell death and inflammatory reaction
Neurol. 2018;9:1103. in three models of intracerebral hemorrhage in adult rats. J Stroke Cerebrovasc
34. Fouda AY, Newsome AS, Spellicy S, et al. Minocycline in acute cerebral Dis. 2003;12(3):152–159.
hemorrhage: an early phase randomized trial. Stroke. 2017;48:2885–2887. 65. Poon CC, Sarkar S, Yong VW, Kelly JJ. Glioblastoma-associated microglia and
35. Yong HYF, Rawji KS, Ghorbani S, Xue M, Yong VW. The benefits of macrophages: targets for therapies to improve prognosis. Brain.
neuroinflammation for the repair of the injured central nervous system. Cell 2017;140:1548–1560.
Mol Immunol. 2019;16:540–546. 66. Wan S, Cheng Y, Jin H, et al. Microglia activation and polarization after
36. Dang B, Duan X, Wang Z, He W, Chen G. A therapeutic target of cerebral intracerebral hemorrhage in mice: the role of protease-activated receptor-1.
hemorrhagic stroke: matrix metalloproteinase-9. Curr Drug Targets. Transl Stroke Res. 2016;7:478–487.
2017;18:1358–1366. 67. Kobayashi K, Imagama S, Ohgomori T, et al. Minocycline selectively inhibits M1
37. Chang JJ, Emanuel BA, Mack WJ, Tsivgoulis G, Alexandrov AV. Matrix polarization of microglia. Cell Death Dis. 2013;4. e525–e525.
metalloproteinase-9: dual role and temporal profile in intracerebral 68. Yang X, Sun J, Kim TJ, et al. Pretreatment with low-dose fimasartan ameliorates
hemorrhage. J Stroke Cerebrovasc Dis. 2014;23:2498–2505. NLRP3 inflammasome-mediated neuroinflammation and brain injury after
38. Xue M, Yong VW. Matrix metalloproteinases in intracerebral hemorrhage. intracerebral hemorrhage. Exp Neurol. 2018;310:22–32.
Neurol Res. 2008;30:775–782. 69. Lan X, Han X, Li Q, et al. Pinocembrin protects hemorrhagic brain primarily by
39. Wang J, Dore S. Inflammation after intracerebral hemorrhage. J Cereb Blood Flow inhibiting toll-like receptor 4 and reducing M1 phenotype microglia. Brain
Metab. 2007;27:894–908. Behav Immun. 2017;61:326–339.
40. Zhou Y, Wang Y, Wang J, Anne Stetler R, Yang QW. Inflammation in 70. Yang Z, Dong S, Zheng Q, et al. FTY720 attenuates iron deposition and glial
intracerebral hemorrhage: from mechanisms to clinical translation. Prog responses in improving delayed lesion and long-term outcomes of collagenase-
Neurobiol. 2014;115:25–44. induced intracerebral hemorrhage. Brain Res. 2019;1718:91–102.
41. Garrett MC, Otten ML, Starke RM, et al. Synergistic neuroprotective effects of 71. Sinn D-I, Lee S-T, Chu K, et al. Proteasomal inhibition in intracerebral
C3a and C5a receptor blockade following intracerebral hemorrhage. Brain Res. hemorrhage: neuroprotective and anti-inflammatory effects of bortezomib.
2009;1298:171–177. Neurosci Res. 2007;58:12–18.
42. Ducruet AF, Zacharia BE, Hickman ZL, et al. The complement cascade as a 72. Sun N, Shen Y, Han W, et al. Selective sphingosine-1-phosphate receptor 1
therapeutic target in intracerebral hemorrhage. Exp Neurol. 2009;219:398–403. modulation attenuates experimental intracerebral hemorrhage. Stroke.
43. Sun YM, Wang YT, Jiang L, Xue MZ. The effects of deferoxamine on inhibition for 2016;47:1899–1906.
microglia activation and protection of secondary nerve injury after 73. Wang Y, Chen Q, Tan Q, et al. Simvastatin accelerates hematoma resolution
intracerebral hemorrhage in rats. Pak J Pharm Sci. 2016;29(3 after intracerebral hemorrhage in a PPARc-dependent manner.
Suppl):1087–1093. Neuropharmacology. 2018;128:244–254.
44. Hatakeyama T, Okauchi M, Hua Y, Keep RF, Xi G. Deferoxamine reduces 74. Chang C-F, Wan J, Li Q, Renfroe SC, Heller NM, Wang J. Alternative activation-
neuronal death and hematoma lysis after intracerebral hemorrhage in aged skewed microglia/macrophages promote hematoma resolution in
rats. Transl Stroke Res. 2013;4:546–553. experimental intracerebral hemorrhage. Neurobiol Dis. 2017;103:54–69.
45. Xie Q, Gu Y, Hua Y, Liu W, Keep RF, Xi G. Deferoxamine attenuates white matter 75. Lu Q, Gao L, Huang L, et al. Inhibition of mammalian target of rapamycin
injury in a piglet intracerebral hemorrhage model. Stroke. 2014;45:290–292. improves neurobehavioral deficit and modulates immune response after
46. Yeatts SD, Palesch YY, Moy CS, Selim M. High dose deferoxamine in intracerebral hemorrhage in rat. J Neuroinflamm. 2014;11:44.
intracerebral hemorrhage (HI-DEF) trial: rationale, design, and methods. 76. Shi H, Zheng K, Su Z, et al. Sinomenine enhances microglia M2 polarization and
Neurocrit Care. 2013;19:257–266. attenuates inflammatory injury in intracerebral hemorrhage. J Neuroimmunol.
47. LeBlanc 3rd RH, Chen R, Selim MH, Hanafy KA. Heme oxygenase-1-mediated 2016;299:28–34.
neuroprotection in subarachnoid hemorrhage via intracerebroventricular 77. Jiang YB, Wei KY, Zhang XY, Feng H, Hu R. White matter repair and treatment
deferoxamine. J Neuroinflammation. 2016;13:244. strategy after intracerebral hemorrhage. CNS Neurosci Ther.
48. Lyden PD, Shuaib A, Lees KR, et al. Safety and tolerability of NXY-059 for acute 2019;25:1113–1125.
intracerebral hemorrhage: the CHANT trial. Stroke. 2007;38:2262–2269. 78. Shao A, Zhu Z, Li L, Zhang S, Zhang J. Emerging therapeutic targets associated
49. Righy C, Bozza MT, Oliveira MF, Bozza FA. Molecular, cellular and clinical with the immune system in patients with intracerebral haemorrhage (ICH):
aspects of intracerebral hemorrhage: are the enemies within?. Curr from mechanisms to translation. EBioMedicine. 2019;45:615–623.
Neuropharmacol. 2016;14:392–402.

You might also like