Sie sind auf Seite 1von 6

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/304619735

Intraventricular Hemorrhage: the Role of Blood Components in Secondary


Injury and Hydrocephalus

Article · June 2016


DOI: 10.1007/s12975-016-0480-8

CITATIONS READS

23 406

7 authors, including:

Thomas Garton Richard F Keep


Johns Hopkins University University of Michigan
9 PUBLICATIONS   172 CITATIONS    448 PUBLICATIONS   17,704 CITATIONS   

SEE PROFILE SEE PROFILE

David Andrew Wilkinson Jennifer Strahle


University of Michigan Washington University in St. Louis
44 PUBLICATIONS   327 CITATIONS    56 PUBLICATIONS   967 CITATIONS   

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Pediatric Hydrocephalus View project

Validation View project

All content following this page was uploaded by Thomas Garton on 24 October 2017.

The user has requested enhancement of the downloaded file.


Transl. Stroke Res.
DOI 10.1007/s12975-016-0480-8

COMMENTARY

Intraventricular Hemorrhage: the Role of Blood Components


in Secondary Injury and Hydrocephalus
Thomas Garton 1 & Richard F. Keep 1 & D. Andrew Wilkinson 1 & Jennifer M. Strahle 1 &
Ya Hua 1 & Hugh J. L. Garton 1 & Guohua Xi 1,2

Received: 20 June 2016 / Accepted: 22 June 2016


# Springer Science+Business Media New York 2016

Intraventricular hemorrhage (IVH) is characterized by an in- occurs in 20–30 % of such patients [13]. In addition, blood
flux of blood into the ventricles of the brain. It has a highly in the ventricle may contribute to hydrocephalus after traumat-
morbid prognosis and develops in more than 12,000 prema- ic brain injury [14].
ture infants every year in the USA [1, 2]. Within the last two Despite this, there is a scarcity of research into the field of
decades, about 31 % of successfully resuscitated very preterm IVH when compared to ICH or SAH. Studies of ICH and
infants—born prior to 30-week gestational age—experienced SAH have shown increasing recognition of the importance
IVH, one third of which were of grade 3 or 4 severity [3]. of particular blood components in brain damage [6, 15–17].
Furthermore, the incidence of severe IVH has increased over Although the body of literature regarding the role of blood
the last 20 years. In addition to the severe neurological deficits components in IVH is far smaller than in ICH or SAH, a
associated with the disease, high-grade IVH can lead to post- similar focus is apparent—the roles of hemoglobin and its
hemorrhagic hydrocephalus (PHH), with 48 % of patients degradation products, plasma components and the coagulation
sustaining grades 3 and 4 IVH developing PHH. When com- cascade, platelets, and leukocytosis in the pathogenesis of
bined, IVH-PHH is a dauntingly critical condition, and one IVH are becoming more apparent (Fig. 1).
whose pathology is not completely understood.
In addition to infants, IVH is also seen in patients with
intracerebral hemorrhage (ICH) and subarachnoid hemor- Hemoglobin and Its Degradation Products
rhage (SAH), two major types of hemorrhagic stroke with
high morbidity and mortality [4–10]. IVH occurs in up to After hemorrhage and hemolysis, hemoglobin is released into
50 % of patients with primary ICH and 45 % of patients with the extracellular space and subsequently exhibits cytotoxic
aneurysmal SAH [11–13]. Recent studies have found IVH is a effects, depleting nitric oxide levels and increasing the inflam-
predictor of poor outcome after ICH [11, 12]. The internation- matory response [18]. Consequently, the role of hemoglobin
al surgical trial in intracerebral hemorrhage showed that in following hemolysis after IVH has recently become a topic of
ICH patients, IVH lowers the rate of favorable outcomes and interest. Gram et al. showed that intraventricular hemoglobin
is associated with hydrocephalus in more than 50 % of adult induces a pro-inflammatory response characterized by in-
patients [11]. IVH is also an independent prognostic factor for creased cerebrospinal fluid (CSF) levels of the pro-
poor outcome in SAH patients, and acute hydrocephalus inflammatory cytokine tumor necrosis factor-α (TNF-α), as
well as by periventricular brain damage [19, 20].
Intraventricular hemoglobin has been shown to induce signif-
* Guohua Xi
icant neurodegeneration in the hippocampus via JNK signal-
guohuaxi@med.umich.edu ing pathways within 3 days of hemorrhage [21].
In addition to playing a role in cellular damage, hemoglo-
1
Department of Neurosurgery, University of Michigan, Ann bin is also involved in the development of PHH. In a neonatal
Arbor, MI, USA rat model, intraventricular injection of hemoglobin results in a
2
Department of Neurosurgery, R5018 Biomedical Science Research significant (3.6-fold) enlargement of the ventricular system
Building, 109 Zina Pitcher Place, Ann Arbor, MI 48109-2200, USA compared to an injection with artificial CSF [22]. Although
Transl. Stroke Res.

Fig. 1 An overview of blood


components and brain injury after
intraventricular hemorrhage

the underlying mechanism is still not fully elucidated, intra- released is being degraded into free iron [27]. Moreover, this
ventricular hemoglobin has been shown to induce structural iron may then be released into CSF. In a small study of infants
and cellular damage to the choroid plexus, which is not only with post-hemorrhagic ventricular dilatation, 75 % had non-
thought to be the primary site of CSF production, but which is protein-bound iron in their CSF, compared to 0 % in controls
also a crucial component of the blood-CSF barrier [19]. Such [28]. In addition, iron accumulation following ICH/IVH is
damage is accompanied by upregulation of cellular activation, correlated with ventricular dilation and brain damage [29].
inflammatory response, and oxidative stress in the choroid In a germinal matrix hemorrhage-IVH model, iron accumula-
plexus. Furthermore, hemoglobin alters the expression of tion was correlated with lateral ventricular expansion, brain
aquaporins (AQPs), which play a major role in the production edema, neuronal degeneration in the basal ganglia, and a de-
and circulation of CSF and have been implicated in the for- crease in long-term motor function [30]. These effects were
mation of hydrocephalus. Exposure of choroid plexus epithe- significantly reduced by minocycline, although whether this
lial cells to hemoglobin results in a significant changes in was due to the ability of that compound to chelate iron and/or
mRNA and protein levels of AQP1 and AQP5 [23]. AQP5 inhibit microglia is uncertain. Intraventricular injection of iron
overexpression is linked to increased cell proliferation via the also leads to significant ventriculomegaly, while treatment
RAS signaling pathway [24], which, if occurring in the cho- with the iron chelator def eroxamine ameliorates
roid plexus may contribute to increased CSF production, al- hemoglobin-induced effects on ventricle size and hippocam-
though this has not been investigated. Thus, the ability of pal damage [21, 22, 27]. Injection of protoporphyrin IX (es-
hemoglobin to affect the expression of AQPs in the choroid sentially an iron-less heme compound) does not induce hydro-
plexus may be linked to the pathogenesis of PHH. cephalus [22], implicating iron as the key component of the
The mechanism by which hemoglobin induces heme moiety in the mechanism behind PHH. It has also been
periventricular tissue damage and hydrocephalus is closely suggested that iron induces hydrocephalus by activating the
tied to the process of its degradation. After hemolysis, hemo- WNT signaling pathway after IVH, which is closely implicat-
globin dissociates into αβ dimers, which are quickly scav- ed in subarachnoid fibrosis in chronic hydrocephalus [31].
enged from the extracellular space by haptoglobin, the plasma Thus, there is considerable evidence that hemoglobin and
protein with the highest binding affinity for hemoglobin [25]. hemoglobin degradation products play an important role in the
From there, the hemoglobin heterodimers undergo receptor- pathogenesis of hydrocephalus and brain injury following
mediated endocytosis via CD163, a receptor expressed in IVH. While much of the effort to target this process therapeu-
monocytes and macrophages, and potentially neurons (though tically has focused on iron chelation, Gram et al. suggested
this finding warrants further investigation) [21]. Once inside injecting the scavenger protein haptoglobin as a way to reduce
the cell, the heme moieties are degraded by either heme oxy- the effects of free hemoglobin [19]. In plasma, haptoglobin
genase (HO)-1 or HO-2 to form biliverdin, carbon monoxide, plays a key role in clearing free hemoglobin. Although a small
and free iron [26]. The iron released by HO has the ability to amount of haptoglobin in circulating blood will enter the brain
participate in oxidative Fenton reactions and, therefore, has with the IVH, normal adult CSF levels are very low suggest-
been a primary subject of studies examining hemoglobin- ing that there may be insufficient haptoglobin to bind hemo-
mediated hydrocephalus and brain injury following IVH. globin released after hemolysis [32]. In addition, in the neo-
Intraventricular injection of lysed red blood cells increases nate, plasma haptoglobin expression is extremely low, which
HO-1 activity in the brain and induces expression of ferritin, might contribute to the high morbidity of neonatal hemor-
an endogenous iron chelator, indicating that the hemoglobin rhage [33]. Haptoglobin is limited as a therapy as it is a high
Transl. Stroke Res.

molecular weight protein and would likely require direct ad- Thrombin’s downstream target fibrinogen has also been of
ministration into the brain. interest in intracranial hemorrhage, mostly in patients on an-
In addition to hemoglobin and its degradation products, ticoagulants who are at risk for hemorrhage because of inad-
there may be other erythrocyte components that play a role equate fibrin formation or in the use of fibrinolytics to clear
in IVH-induced injury. For example, in ICH-induced brain hematomas. An example of the latter is the Clot lysis: evalu-
injury, there is evidence that red blood cell carbonic anhydrase ating accelerated resolution of intraventricular hemorrhage
can have deleterious effects [34]. Further study into both he- phase III (CLEAR III) trial which is investigating tissue plas-
moglobin and other red blood cell breakdown products is minogen activator (tPA) as an effective intraventricular fibri-
warranted including treatments specific to these pathways. nolytic agent. However, recent evidence indicates that extra-
vascular fibrinogen is a powerful pro-inflammatory mediator
[44, 45] and may also play a role in mediating brain injury in
Plasma Components and the Coagulation Cascade IVH. The role of fibrinogen in IVH-induced brain injury and
hydrocephalus merits investigation.
Plasma-derived factors also have a role in IVH-induced brain It should be noted that one of the proposed functions of the
injury, with most of the existing evidence focusing on the role blood–brain barrier is to prevent the entry of potentially neu-
of the coagulation cascade. Thrombin is a key factor in the roactive compounds from blood to the brain. However, there
coagulation cascade. Also known as factor IIa, thrombin is may also be plasma components in addition to prothrombin
created by the cleavage of prothrombin by the prothrombinase (converted to thrombin) and fibrinogen (converted to fibrin)
complex (factors Xa/Va). As a serine protease, the primary that enter the brain during IVH and induce or modulate brain
function of thrombin in the coagulation cascade is to limit injury. As noted previously, haptoglobin may be one such
further hemorrhage by cleaving fibrinogen into insoluble fibrin. component that warrants further study, but this has received
However, thrombin plays a variety of other distinctly detrimen- little attention.
tal roles upon release into the ventricles following IVH. Some
of the actions of thrombin are via a family of protease-activated
receptors (PARs) and most significantly PAR-1. PAR-1 is a G- Platelets
protein-coupled receptor activated directly by thrombin proteo-
lytic activity which initiates a wide range of intracellular cas- Platelets are an integral component of the hemostatic system,
cades [35, 36]. Thrombin production in IVH activates PAR-1 but in general, the role of platelets in IVH-induced injury has
and thereby induces both significant ependymal wall damage received little attention compared to the role of erythrocytes.
and hydrocephalus [37]. One PAR-1-mediated pathway is ac- Indeed, this is the case for all forms of cerebral hemorrhage.
tivation of the Src family of kinases. These proteins can phos- Platelet aggregation (platelet plug) is involved in hemostasis.
phorylate metalloproteinases, known to be a significant cause Activated platelets release a range of potent chemical media-
of damage in other types of intracranial hemorrhage, although tors, including adenosine diphosphate, serotonin, and throm-
their specific roles in IVH and clot clearance are unknown [38]. boxane A2. As noted above, one mediator released by platelets,
Indeed, SCH79797, a PAR-1 antagonist, has been shown to TGF β, has been implicated in inducing hydrocephalus [43].
significantly reduce this damage [37], while inhibiting Src fam-
ily kinases with PP2 blocks brain edema and blood brain barrier
disruption [38]. Thrombin-induced hydrocephalus is reduced Leukocytes
by CSF production inhibitors such as acetazolamide (a carbon-
ic anhydrase inhibitor), but even after this treatment, rats Leukocytes are the body’s mobile immune cells, tasked with
injected with thrombin had ventricular volumes double those protecting against infection and foreign bodies. Leukocytes
of vehicle controls [39]. are broadly categorized into two main types, the lymphoid
In SAH animal models, transforming growth factor β1 cells (B cells, T cells, NK cells) and myeloid cells (basophils,
(TGF β) has been shown to be a mediator of thrombin- neutrophils, eosinophils, and monocytes). All of these cells
induced inflammation [40]. TGF β is released into CSF by can be found circulating through the blood and migrating to-
extravasated platelets following IVH, and some have theo- wards the site of injury during an inflammatory response [46,
rized that it may play a role in the pathogenesis of obstructive 47]. The brain’s resident immune cell is the microglia, a dif-
hydrocephalus [41]. While there is some controversy, recent ferentiated monocyte that can be activated classically or alter-
studies of kaolin-induced hydrocephalus suggest that the TGF natively. The classical M1 pathway is pro-inflammatory and
β antagonist, decorin, reduces ventricle size and white matter has been linked to increased brain injury following hemor-
injury [42, 43]. Further research into this topic is necessary to rhage, while the alternative M2 pathway is involved in reso-
elucidate the role of TGF β and downstream signaling in lution of inflammation [48]. In the neonatal brain, microglial
hydrocephalus, including PHH. density is already the highest in periventricular white matter.
Transl. Stroke Res.

Following IVH, microglia and neutrophils accumulate in the References


periventricular area to a greater extent than in the cortex with a
time course beginning at about 48-h post-hemorrhage [49]. 1. Ballabh P. Intraventricular hemorrhage in premature infants: mech-
This infiltration and activation of microglial cells may result anism of disease. Pediatr Res. 2010;67(1):1–8. Pubmed Central
PMCID: 2799187, Epub 2009/10/10. eng.
in increased phagocytic activity [50]. Moreover, M1-activated
2. Strahle J, Garton HJ, Maher CO, Muraszko KM, Keep RF, Xi G.
microglia release pro-inflammatory cytokines, which can be Mechanisms of hydrocephalus after neonatal and adult intraventric-
cytotoxic to the surrounding brain matter [48]. ular hemorrhage. Transl Stroke Res. 2012;3 Suppl 1:25–38.
While attention has almost entirely focused on the role of Pubmed Central PMCID: 3750748.
leukocytes infiltrating into the brain after cerebral hemor- 3. Radic JA, Vincer M, McNeely PD. Temporal trends of intraventric-
ular hemorrhage of prematurity in Nova Scotia from 1993 to 2012.
rhage, it should be noted that they are also a component of J Neurosurg Pediatr. 2015;15(6):573–9.
the initial hemorrhage. They have a relatively short half-life 4. Schlunk F, Greenberg SM. The pathophysiology of intracerebral
[51] but are a potential source of inflammatory mediators; hemorrhage formation and expansion. Transl Stroke Res.
whether or not they impact the brain inflammatory response 2015;6(4):257–63.
5. Zhang JH. Vascular neural network in subarachnoid hemorrhage.
after an IVH has not been examined directly.
Transl Stroke Res. 2014;5(4):423–8. Pubmed Central PMCID:
4127639.
6. Xi G, Keep RF, Hoff JT. Mechanisms of brain injury after intrace-
Conclusions rebral hemorrhage. Lancet Neurol. 2006;5(1):53–63.
7. Qureshi AI, Mendelow AD, Hanley DF. Intracerebral haemorrhage.
Lancet. 2009;373(9675):1632–44. Epub 2009/05/12. eng.
The current state of research investigating the roles of specific 8. Cahill J, Calvert JW, Zhang JH. Mechanisms of early brain injury
blood components following IVH leaves vast gaps for future after subarachnoid hemorrhage. J Cereb Blood Flow Metab.
studies to fill. More research is needed on the mechanisms by 2006;26(11):1341–53. Epub 2006/02/17. eng.
which the blood components such as hemoglobin, plasma prod- 9. Macdonald RL, Pluta RM, Zhang JH. Cerebral vasospasm after
subarachnoid hemorrhage: the emerging revolution. Nat Clin
ucts, platelets, and leukocytes induce brain damage. In particu- Pract Neurol. 2007;3(5):256–63. Epub 2007/05/05. eng.
lar, the mechanism by which hemoglobin induces hydrocepha- 10. Tso MK, Macdonald RL. Subarachnoid hemorrhage: a review of
lus is not fully understood. While the release of free iron is experimental studies on the microcirculation and the neurovascular
clearly a significant factor, it probably does not account for unit. Transl Stroke Res. 2014;5(2):174–89.
11. Bhattathiri PS, Gregson B, Prasad KS, Mendelow AD.
the entirety of the damage found, suggesting that other poten-
Intraventricular hemorrhage and hydrocephalus after spontaneous
tially non-oxidative mechanisms play a role. Furthermore, intracerebral hemorrhage: results from the STICH trial. Acta
while deferoxamine’s efficacy against iron-induced ventricular Neurochir Suppl. 2006;96:65–8.
damage in animal and clinical studies of ICH has been prom- 12. Hanley DF. Intraventricular hemorrhage: severity factor and treat-
ising, clinical trials in ICH are still ongoing [52], and no trial ment target in spontaneous intracerebral hemorrhage. Stroke.
2009;40(4):1533–8. Pubmed Central PMCID: 2744212, Epub
investigating deferoxamine in IVH has been proposed. 2009/02/28. eng.
Overall, few studies have examined the role of extravasated 13. Rosen D, Macdonald R, Huo D, Goldenberg F, Novakovic R, Frank
blood in IVH and PHH. IVH is an extremely morbid disease J, et al. Intraventricular hemorrhage from ruptured aneurysm: clin-
but, as of yet, there is no effective treatment as is evidenced by ical characteristics, complications, and outcomes in a large, pro-
spective, multicenter study population. J Neurosurg. 2007;107(2):
its unchanging incidence and morbidity over the past decades 261–5.
[3, 53]. For infants with grade 4 IVH, mortality rates remain as 14. Zhao J, Chen Z, Xi G, Keep RF, Hua Y. Deferoxamine attenuates
high as 40 % in the USA [53]. Preclinical IVH research is acute hydrocephalus after traumatic brain injury in rats. Transl
beginning to indicate that blood components play a significant Stroke Res. 2014;5:586–94.
role in IVH-induced brain injury and PHH, but the volume of 15. Pandey AS, Xi G. Intracerebral hemorrhage: a multimodality ap-
proach to improving outcome. Transl Stroke Res. 2014;5(3):313–5.
literature and replication is still low. Future studies investigat- 16. Lee J, Keep RF, He Y, Sagher O, Hua Y, Xi G. Hemoglobin and
ing these topics could yield significant advancements towards iron handling in brain following subarachnoid hemorrhage and the
finding effective therapeutic treatments. effect of deferoxamine on early brain injury. J Cereb Blood Flow
Metab. 2010;30(11):1793–803.
17. Keep RF, Hua Y, Xi G. Intracerebral haemorrhage: mechanisms of
Acknowledgments This study was supported by grants NS-073595, injury and therapeutic targets. Lancet Neurol. 2012;11(8):720–31.
NS-079157, NS-084049, NS-090925, NS-091545, NS-096917, and Epub 2012/06/16. eng.
NS-007222 from the National Institutes of Health (NIH) and 973 18. Xiong XY, Wang J, Qian ZM, Yang QW. Iron and intracerebral
Program-2014CB541600. hemorrhage: from mechanism to translation. Transl Stroke Res.
2014;5(4):429–41.
19. Gram M, Sveinsdottir S, Cinthio M, Sveinsdottir K, Hansson SR,
Compliance with Ethical Standards Morgelin M, et al. Extracellular hemoglobin—mediator of inflam-
mation and cell death in the choroid plexus following preterm in-
Conflict of Interest The authors declare that they have no conflict of traventricular hemorrhage. J Neuroinflammation. 2014;11:200.
interest. Pubmed Central PMCID: 4269927.
Transl. Stroke Res.

20. Gram M, Sveinsdottir S, Ruscher K, Hansson SR, Cinthio M, Flow Metab. 2014;34(3):489–94. Pubmed Central PMCID:
Akerstrom B, et al. Hemoglobin induces inflammation after pre- 3948129.
term intraventricular hemorrhage by methemoglobin formation. J 38. Liu DZ, Ander BP, Xu H, Shen Y, Kaur P, Deng W, et al. Blood–
Neuroinflammation. 2013;10:100. Pubmed Central PMCID: brain barrier breakdown and repair by Src after thrombin-induced
3750409. injury. Ann Neurol. 2010;67(4):526–33. Pubmed Central PMCID:
21. Garton TP, He Y, Garton HJ, Keep RF, Xi G, Strahle JM. 2919346, Epub 2010/05/04. eng.
Hemoglobin-induced neuronal degeneration in the hippocampus 39. Gao F, Zheng M, Hua Y, Keep RF, Xi G. Acetazolamide attenuates
after neonatal intraventricular hemorrhage. Brain Res. 2016;1635: thrombin-induced hydrocephalus. Acta Neurochir Suppl.
86–94. Pubmed Central PMCID: 4801173. 2016;121:373–7.
22. Strahle JM, Garton T, Bazzi AA, Kilaru H, Garton HJ, Maher CO, 40. Li T, Zhang P, Yuan B, Zhao D, Chen Y, Zhang X. Thrombin-
et al. Role of hemoglobin and iron in hydrocephalus after neonatal induced TGF-beta1 pathway: a cause of communicating hydro-
intraventricular hemorrhage. Neurosurgery. 2014;75(6):696–705. cephalus post subarachnoid hemorrhage. Int J Mol Med.
Pubmed Central PMCID: 4237659, discussion 6. 2013;31(3):660–6.
23. Sveinsdottir S, Gram M, Cinthio M, Sveinsdottir K, Morgelin M, 41. Whitelaw A, Christie S, Pople I. Transforming growth factor-beta1:
Ley D. Altered expression of aquaporin 1 and 5 in the choroid a possible signal molecule for posthemorrhagic hydrocephalus?
plexus following preterm intraventricular hemorrhage. Dev Pediatr Res. 1999;46(5):576–80.
Neurosci. 2014;36(6):542–51. 42. Aojula A, Botfield H, McAllister 2nd JP, Gonzalez AM, Abdullah O,
24. Direito I, Madeira A, Brito MA, Soveral G. Aquaporin-5: from Logan A, et al. Diffusion tensor imaging with direct cytopathological
structure to function and dysfunction in cancer. Cellular and molec- validation: characterisation of decorin treatment in experimental ju-
ular life sciences : CMLS. 2016;73(8):1623–40. venile communicating hydrocephalus. Fluids and barriers of the
25. Ascenzi P, Bocedi A, Visca P, Altruda F, Tolosano E, Beringhelli T, CNS. 2016;13(1):9. Pubmed Central PMCID: 4888658.
et al. Hemoglobin and heme scavenging. IUBMB life. 2005;57(11): 43. Botfield H, Gonzalez AM, Abdullah O, Skjolding AD, Berry M,
749–59. McAllister 2nd JP, et al. Decorin prevents the development of ju-
26. Maines MD. Heme oxygenase: function, multiplicity, regulatory venile communicating hydrocephalus. Brain. 2013;136(Pt 9):
mechanisms, and clinical applications. FASEB J. 1988;2(10): 2842–58.
2557–68. 44. Davalos D, Ryu JK, Merlini M, Baeten KM, Le Moan N, Petersen
27. Gao C, Du H, Hua Y, Keep RF, Strahle J, Xi G. Role of red blood MA, et al. Fibrinogen-induced perivascular microglial clustering is
cell lysis and iron in hydrocephalus after intraventricular hemor- required for the development of axonal damage in neuroinflamma-
rhage. J Cereb Blood Flow Metab. 2014;34(6):1070–5. Pubmed tion. Nat Commun. 2012;3:1227. Pubmed Central PMCID:
Central PMCID: 4050252. 3514498.
28. Savman K, Nilsson UA, Blennow M, Kjellmer I, Whitelaw A. Non- 45. Ryu JK, Petersen MA, Murray SG, Baeten KM, Meyer-Franke A,
protein-bound iron is elevated in cerebrospinal fluid from preterm Chan JP, et al. Blood coagulation protein fibrinogen promotes au-
infants with posthemorrhagic ventricular dilatation. Pediatr Res. toimmunity and demyelination via chemokine release and antigen
2001;49(2):208–12. presentation. Nat Commun. 2015;6:8164. Pubmed Central PMCID:
29. Chen Q, Tang J, Tan L, Guo J, Tao Y, Li L, et al. Intracerebral 4579523.
hematoma contributes to hydrocephalus after intraventricular hem- 46. Zhao X, Sun G, Zhang H, Ting SM, Song S, Gonzales N, et al.
orrhage via aggravating iron accumulation. Stroke. 2015;46(10): Polymorphonuclear neutrophil in brain parenchyma after experimen-
2902–8. tal intracerebral hemorrhage. Transl Stroke Res. 2014;5(5):554–61.
30. Guo J, Chen Q, Tang J, Zhang J, Tao Y, Li L, et al. Minocycline- 47. Wang J, Dore S. Inflammation after intracerebral hemorrhage. J
induced attenuation of iron overload and brain injury after experi- Cereb Blood Flow Metab. 2007;27(5):894–908. Epub 2006/10/
mental germinal matrix hemorrhage. Brain Res. 2015;1594:115–24. 13. eng.
31. Meng H, Li F, Hu R, Yuan Y, Gong G, Hu S, et al. Deferoxamine 48. Zhao H, Garton T, Keep RF, Hua Y, Xi G. Microglia/macrophage
alleviates chronic hydrocephalus after intraventricular hemorrhage polarization after experimental intracerebral hemorrhage. Transl
through iron chelation and Wnt1/Wnt3a inhibition. Brain Res. Stroke Res. 2015;6(6):407–9. Pubmed Central PMCID: 4628553.
2015;1602:44–52. 49. Georgiadis P, Xu H, Chua C, Hu F, Collins L, Huynh C, et al.
32. Galea J, Cruickshank G, Teeling JL, Boche D, Garland P, Perry VH, Characterization of acute brain injuries and neurobehavioral pro-
et al. The intrathecal CD163-haptoglobin-hemoglobin scavenging files in a rabbit model of germinal matrix hemorrhage. Stroke.
system in subarachnoid hemorrhage. J Neurochem. 2012;121(5): 2008;39(12):3378–88.
785–92. Pubmed Central PMCID: 3412209. 50. Supramaniam V, Vontell R, Srinivasan L, Wyatt-Ashmead J,
33. Kessel I, Leib M, Levy A, Miller-Lotan R, Waisman D, Jacobson E, Hagberg H, Rutherford M. Microglia activation in the extremely
et al. Does haptoglobin phenotype influence postnatal morbidity in preterm human brain. Pediatr Res. 2013;73(3):301–9.
preterm neonates? Am J Perinatol. 2016;33(2):130–5. 51. Summers C, Rankin SM, Condliffe AM, Singh N, Peters AM,
34. Guo F, Hua Y, Wang J, Keep RF, Xi G. Inhibition of carbonic Chilvers ER. Neutrophil kinetics in health and disease. Trends
anhydrase reduces brain injury after intracerebral hemorrhage. Immunol. 2010;31(8):318–24. Pubmed Central PMCID: 2930213.
Translational Stroke Research. 2012;3:130–7. 52. Yeatts SD, Palesch YY, Moy CS, Selim M. High dose deferox-
35. Cheng Y, Xi G, Jin H, Keep RF, Feng J, Hua Y. Thrombin-induced amine in intracerebral hemorrhage (HI-DEF) trial: rationale, design,
cerebral hemorrhage: role of protease-activated receptor-1. Transl and methods. Neurocrit Care. 2013;19(2):257–66. Pubmed Central
Stroke Res. 2014;5(4):472–5. Pubmed Central PMCID: 3962522. PMCID: 3932442.
36. Coughlin SR. Thrombin signalling and protease-activated recep- 53. Christian EA, Jin DL, Attenello F, Wen T, Cen S, Mack WJ, et al.
tors. Nature. 2000;407(6801):258–64. Trends in hospitalization of preterm infants with intraventricular
37. Gao F, Liu F, Chen Z, Hua Y, Keep RF, Xi G. Hydrocephalus after hemorrhage and hydrocephalus in the United States, 2000–2010.
intraventricular hemorrhage: the role of thrombin. J Cereb Blood J Neurosurg Pediatr. 2016;17(3):260–9.

View publication stats

Das könnte Ihnen auch gefallen