Review Open Access
Copyright ©The Author(s) 2015. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Stem Cells. Apr 26, 2015; 7(3): 618-629
Published online Apr 26, 2015. doi: 10.4252/wjsc.v7.i3.618
Stem cell therapy in intracerebral hemorrhage rat model
Marcos F Cordeiro, Ana P Horn, Programa de Pós-graduação em Ciências Fisiológicas-Fisiologia Animal Comparada, Universidade Federal do Rio Grande, Rio Grande RS 96210-900, Brazil
Marcos F Cordeiro, Ana P Horn, Laboratório de Histologia, Instituto de Ciências Biológicas, Universidade Federal do Rio Grande, Rio Grande RS 96210-900, Brazil
Marcos F Cordeiro, Ana P Horn, Laboratório de Neurociências, Instituto de Ciências Biológicas, Universidade Federal do Rio Grande, Rio Grande RS 96210-900, Brazil
Author contributions: Cordeiro MF and Horn AP contributed to this paper.
Conflict-of-interest: The authors have no conflicts of interest to declare.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Marcos F Cordeiro, BSc, Laboratório de Histologia, Instituto de Ciências Biológicas, Universidade Federal do Rio Grande, Av Itália, Km 8, Rio Grande RS 96210-900, Brazil. cordeiromf@yandex.com
Telephone: +55-53-32935182 Fax: +55-53-32936848
Received: July 29, 2014
Peer-review started: July 29, 2014
First decision: November 18, 2014
Revised: December 5, 2014
Accepted: December 18, 2014
Article in press: December 19, 2014
Published online: April 26, 2015

Abstract

Intracerebral hemorrhage (ICH) is a very complex pathology, with many different not fully elucidated etiologies and prognostics. It is the most severe subtype of stroke, with high mortality and morbidity rates. Unfortunately, despite the numerous promising preclinical assays including neuroprotective, anti-hypertensive, and anti-inflammatory drugs, to this moment only symptomatic treatments are available, motivating the search for new alternatives. In this context, stem cell therapy emerged as a promising tool. However, more than a decade has passed, and there is still much to be learned not only about stem cells, but also about ICH itself, and how these two pieces come together. To date, rats have been the most widely used animal model in this research field, and there is much more to be learned from and about them. In this review, we first summarize ICH epidemiology, risk factors, and pathophysiology. We then present different methods utilized to induce ICH in rats, and examine how accurately they represent the human disease. Next, we discuss the different types of stem cells used in previous ICH studies, also taking into account the tested transplantation sites. Finally, we summarize what has been achieved in assays with stem cells in rat models of ICH, and point out some relevant issues where attention must be given in future efforts.

Key Words: Cell therapy, Intracerebral hemorrhage, Preclinical assays, Rat, Stem cells

Core tip: In this review, we first summarize intracerebral hemorrhage (ICH) epidemiology, risk factors, and pathophysiology. We then present different methods utilized to induce ICH in rats, and examine how accurately they represent the human disease. Next, we discuss the different types of stem cells used in previous ICH studies, also taking into account the tested transplantation sites. Finally, we summarize what has been achieved in assays with stem cells in rat models of ICH, and point out some relevant issues where attention must be given in future efforts.



INTRODUCTION

Marginal or no success was achieved from decades of research for therapeutic alternatives for intracerebral hemorrhage (ICH)[1,2]. Despite the numerous promising preclinical assays including neuroprotective, anti-hypertensive, and anti-inflammatory drugs, only symptomatic treatments are currently available[2,3], which motivates the search for new alternatives. In this context, cell therapy emerged as a promising tool[4-6]. However, more than a decade has passed, and there is still much to be learned not only about stem cells, but also about ICH itself, and how these two pieces come together. It is clear that achieving a more detailed knowledge of each involved element is crucial to obtain better results.

INTRACEREBRAL HEMORRHAGE: A SEVERE AND HETEROGENEOUS PATHOLOGY
Epidemiology

Cardiovascular diseases (CVDs) are the leading cause of morbidity and mortality worldwide. In 2008, nearly 17.3 million people lost their lives because of CVDs (30% of all deaths); of these, 6.2 million (35.8%) were to cerebrovascular accidents[7], alternatively referred as stroke. Among CVDs, coronary artery disease is responsible for the highest mortality rate on a worldwide basis, followed by stroke[7]. However, stroke mortality rates surpass those of the coronary artery disease in some world regions, like East Asia, Africa, and South America[8].

There are two types of stroke: ischemic and hemorrhagic. This review will focus on nontraumatic ICH, which corresponds to spontaneous leakage of blood within the brain parenchyma or ventricular spaces[1]. ICH is the most lethal subtype of stroke, often causing immediate death[9]. Secondary brain injuries may lead to delayed fatality when death does not occur shortly after the onset. One-year and five-year survival are estimated to be around merely 46% and 29%, respectively[10]. Beyond that, surviving patients are usually left with many limitations in motor and cognitive functions. Overall, ICH is less frequent than its ischemic counterpart, but the proportion fluctuates depending on the ethnic and racial group. Among Chinese people, for example, ICH accounts for about 33% of stroke cases, vs 12% in white populations[11]. On the same hand, global incidence of ICH per 100000 person-years is estimated to be around 24.6, reaching 51.8 in Asian populations[12].

Risk factors

High blood pressure is the major risk factor for CVDs[13], particularly ICH[14,15]. As a matter of fact, it is estimated that about 50% of ICH cases are caused by chronic hypertension[1]. There is a well established liaison between regular consumption of alcoholic beverages and high blood pressure, being this effect independent of age, body mass, smoking habits and physical activity[16-18]. Hypertension and ICH incidence are also linked to other lifestyle-related risk factors, such as smoking[19,20], physical inactivity[21], and high dietary sodium and/or fat consumption[22,23]. About 20% of cases are due to amyloid angiopathy, being this the second cause of ICH[24]. Amyloid deposition incidence is primarily associated with increasing age[25-27], but genetic factors also play an important role[28]. Oral anticoagulant intake is other important risk factor, which not only increases ICH risk, but also has higher intrinsic death rates[29,30]. Other relevant risk factors are obesity, diabetes mellitus, high blood cholesterol and other lipids, brain tumors, aneurysms, cerebrovascular malformations, cavernous angiomas, and arteriovenous fistulae[23,31].

Pathophysiology

As previously stated, hemorrhagic events in brain parenchyma or ventricular spaces are included in ICH. Bleeding associated with chronic hypertension usually originates from microaneurysms near or at bifurcations of penetrating brain arterioles which emanate from basilar arteries or anterior, middle, or posterior cerebral arteries[1,32,33]. In most cases, intraventricular hemorrhage is a consequence of ICH, resulting from hematoma expansion to the ventricular space[1]. This phenomenon increases mortality in nearly five times[34]. In more rare occasions, hematoma may extend to the subarachnoid space[31]. Amyloid angiopathy associated ICH is commonly lobar, often extending into the subarachnoid space and ventricles[35]. Size and location of hematoma are determinant factors in the disease outcome[36]. For example, even though patients commonly experience continued bleeding or rebleeding[37], the latter phenomenon is more recurrent in amyloid angiopathy-derived ICH[38].

Initial injuries are consequent of mechanical disruption of adjacent brain tissue by physical compression due to hematoma formation[1,31,39]. Most hematoma enlargement occurs in the first hours following ictus, but this growth can extend for many hours[40,41]. Disruption of the blood-brain barrier implies in the formation of plasma-derived perihematomal edema[42], which grows rapidly in the first two or three days and reaches full extension about fourteen days after ICH onset[43,44]. Edema evolution can be described in three phases[45]. In the first few hours, hematoma expands and retracts as a result of coagulation, depositing serum molecules in the surrounding tissue. In two to three days, inflammatory mediators come off from circulation. In the third and last phase, erythrocytes suffer degradation, which culminates in hemoglobin toxicity. Thrombin and iron from erythrocytes play major roles in secondary injuries[9,46]. In fact, iron overload seems to be related to the formation of the perihematomal zone[47]. This area surrounds the edema and includes viable but very vulnerable tissue[45].

CELL THERAPY IN INTRACEREBRAL HEMORRHAGE ANIMAL MODELS
Rat: The animal model of choice

Most of the present ICH pathophysiology knowledge derives from studies in animal models. After all, animals assays offer many research advantages, such as the possibility to work with homogeneous groups by the control of variables concerning the subjects (e.g., age, weight, feeding, activity, genetics, etc.), and the ICH itself (e.g., time and site of the injury onset and its intensity). Moreover, this kind of study permits various types of biological assays from surviving subjects at any stage of the pathology.

By far, the majority of ICH studies on animals are performed on rodents, mostly on rats. This choice is understandable, once rats are cheap and convenient to house, easy to manipulate, have a plethora of related products for the most diverse purposes, and have a well described anatomophysiology, which, overall, shares considerable similarities with humans in cerebrovascular parameters[48]. However, there are some important differences that should not be underestimated or neglected.

White matter paucity in rat brains limits the similarities with the human condition[32]. Human brain proportion of white/gray matter shares more similitude with that of porcines and primates, but both are incomparably more expensive and complicated to handle if compared to rats[49]. In addition, if compared to humans, rats also have anatomical disparities in brain perforating arteries, as well as anatomical and biochemical differences in the basal ganglia[50]. It is important to notice that both structures have fundamental involvement in ICH model, especially considering that the commonest experimentally induced ICH is intrastriatal. Actually, this fact is probably included among the reasons why ICH prognostic differs so much in some parameters between rats and humans (more on these matters in the next section). Thus, even though rodents provide advantages as experimental models, multiple species should be used for more trustworthy results[51]. Additionally, rats and mice are also different in many anatomophysiological aspects[52], and tests should not be limited to either of the two.

Intracerebral hemorrhage replication

Bacterial collagenase injection is the most widespread method to induce ICH in rats, followed by autologous blood injection (ABI). The latter is one of the earliest methods developed to replicate ICH in rats. In this model, blood is collected from a superficial vessel and is injected directly into the brain, usually in the striatum[53]. The application of this protocol should be limited to blood toxicity and edema formation studies, once very important aspects of ICH pathology are absent, like small vessel rupture and hematoma expansion[54]. Moreover, especially in rats, ABI implies in exaggerated inflammatory response as a consequence of hemoglobin crystallization[55].

Collagenases are enzymes that break collagen peptide bonds[56]. Consequently, intracerebral injection of bacterial collagenase leads to the breakdown of the basal lamina of blood vessels, establishing the leakage of blood within the brain[57]. Through this method, rebleeding is present and functional impairments are more long lasting than that of ABI model[57]. Also, application is simpler[49]. However, dissolution of endothelial basal lamina causes ICH in an unnatural manner[32].

Even though collagenase injection model mimics ICH with more success than ABI, both show relevant limitations[54]. As previously stated, the standard collagenase and blood site of injection is the striatum, which differs importantly from the human counterpart in biochemical and anatomical features[50]. In top of that, ICH is often not restricted to single anatomical regions in humans, while this is what is achieved with both models[58]. Lastly, but certainly not least, both models comprise intracerebral injections. Therefore, some significant ICH unrelated injuries must be done in order to access the target structures: animal scalp and periosteum must be cut open, a hole must be drilled in the skull, and most importantly, a needle must perforate all brain superjacent structures[59].

As tacit in the Pathophysiology section of this review, ICH is a disease with multiple etiologies, which directly affect the prognostic. More than that, the role played by the ICH causing factors is probably unrestricted to the way the pathology is going to have a start, but is also determinant how it is going to evolve[49]. For example, it is deductible that ICH disrupted by anticoagulant intake develops differently, as hematoma might probably include uncoagulated blood[60].

Even though ICH affects mostly humans with chronic hypertension and advanced age, preclinical studies are predominantly conducted using young and healthy animals. The absence of comorbidities presumably implies in unrealistic outcomes and weaker translational power. Aware of this limitation, many efforts have been made on the last years to develop and use models with characteristic risk factors. ICH related functional outcomes are known to be more severe and long lasting in older[61-63] and chronic hypertensive[64] rats. The latter develop spontaneous ICH with location and distribution consistent to what is observed in hypertensive humans[32]. However, spontaneous chronic hypertension-derived ICH is hard to model, once animals tend to develop ischemic stroke instead of ICH[65].

Prognostic analysis of experimentally induced ICH

ICH in rats and humans have remarkably different prognostics. Human ICH is generally followed by important long lasting or permanent cognitive and motor impairments. In contrast, rats subjected to ICH exhibit no (or at least not long lasting) cognitive deficits[58,66]. Brain structural disparities might play a crucial role in this matter, implying in different affected structures[58]. Motor function impairments in rats are present and well described, but undergo notable recovery in few weeks[67]. Moreover, in rats, edema reaches full extension three to four days after ICH onset[68], whereas in humans it takes weeks[69]. Although there is much to be discovered in this matter, rat superior neurogenesis and/or neuroplasticity may explain these dissimilarities[70].

Many behavioral tests are used to assess ICH outcome. Among the most used are the modified limb placement test (mLPT)[71] and the modified neurological severity score (mNSS)[72]. In both tests, rats are subjected to a set of simple sensorimotor tasks that evaluate different aspects of neurological function. A score is attributed to the performance in each task, resulting in a final sum that ranges from 0 to 14. Both mLPT and mNSS are theoretically consistent, simple to perform, and cost-free, dismissing the need for complex training and special equipments. However, both tests should be always applied by blinded investigators, preferably the same ones for every session. If such attention is present, it is commonly unknown, as it is not always mentioned. Evaluating rat behaviors through mLPT and mNSS can be considerably subjective and interpretative tasks, relying strongly on the observer sensitivity and judging parameters. Importantly, the small score amplitude (0-2 in mLPT and 0-1 in mNSS) attributed to each parameter may lead to significant variations in the final result. Thus, expectations of any nature and level should be absent in the assigned investigator for trustworthy results. Additionally, more than one single test should be applied for more solid assertions.

Last but not least, pathophysiological features of ischemic and hemorrhagic stroke diverge in many ways[73]. Thus, great caution should be taken when comparing findings from each disease.

Stem cell types and clinical applications: An ever-emerging science

Stem cells are characterized by ability of self-renewal (production of identical copies of themselves) and capability to differentiate into distinct functional cells. Cells with capability to differentiate into any body cell are called totipotent. These cells are only found in the earliest stages following fertilization. Embryonic stem cells (ESCs) are mostly pluripotent, meaning that they are able to maturate into cells of the three germ layers (endoderm, mesoderm, and ectoderm). Unfortunately, ESCs are very difficult to isolate, and their use is very limited and even avoided due to ethical, social and religious concerns[74]. Induced pluripotent stem cells (iPSCs) were developed to work around ESCs major issues. iPSCs are generated from somatic cells reprogrammed by the introduction of few defined factors[75]. However, as embryonic stem cells, iPSCs can potentially generate teratomas after transplantation[76-78]. It happens as a result of their propensity for uncontrolled self-renewal and triploblastic differentiation[79].

Somatic stem cells are multipotent, thus possessing the ability to differentiate into only certain cell types. As they can be found in adult tissues, they are sometimes referred as “adult stem cells”. This term, however, is misleading and should be avoided, once the embryo starts containing somatic stem cells shortly after implantation[80]. Mesenchymal stem cells (MSCs) are somatic stem cells present in virtually all organs[81]. They are able to differentiate into mesodermal cell lineages, like osteoblasts, chondrocytes, adipocytes, and myoblasts[82,83]. Some groups, however, showed that MSCs can be induced to differentiate into neurons, but is debatable if they are functional[84]. Bone marrow and adipose tissue have been the preferred sources for MSCs, and cells from each origin are biologically different[82,85]. Bone marrow-derived stem cells (BMSCs) were the first to be discovered, and are the most tested in preclinical assays. However, if compared to BMSCs, adipose-derived stem cells (ASCs) are simpler and less invasive to harvest, as large amounts of these cells can be obtained via liposuction with local anesthesia only[86,87]. If ASC allogeneic applications become someday proven to be safe, fat from liposuctions can be utilized for stem cell cultures instead of being discarded. Moreover, adipose tissue yields more stem cells than any other source[88]. For example, ASCs are more frequent in adipose tissue than BMSCs are in bone marrow in a 100 to 500 fold difference[86]. Although considered very promising[89,90], ASC use is relatively recent if compared to BMSCs. Thus, more studies are needed in order to effectively evaluate advantages of one over another on particular purposes, concerning efficacy, safety, etc. Neural stem cells (NSCs) are multipotent and can differentiate into neurons, astrocytes, and oligodendrocytes. Even though NSCs might initially sound as the most appropriate for nervous system diseases like ICH, the isolation of these cells is a complicated and delicate procedure[91].

Umbilical cord-derived stem cells (UCSCs) are the most used cells from perinatal tissues. Controversy still exists if UCSCs are MSCs or MSC precursors, as they differ in morphological and behavioral aspects[83]. UCSCs are harvested from umbilical cord or umbilical cord blood (UCB) from newborns[92,93], which are presently treated as medical waste. Thus, the isolation method completely avoids donor morbidity[94]. UCSCs are very abundant and show high proliferative capacity[94,95]. UCB contains mononuclear cells that are mostly leucocytes, but it also contains UCSCs. Importantly, 0.6% of umbilical cord blood mononuclear cells are multipotent stress-enduring (Muse) cells. Muse cells are a non teratogenic pluripotent cell type that was recently isolated not only from UCB, but also from dermal fibroblasts, bone marrow, and adipose tissue[79,96,97].

Autologous stem cell transplantation guarantees the absence of immune rejection. However, depending on the circumstances, this might not be feasible. For instance, somatic stem cells require several weeks in culture to achieve sufficient proliferation[91]. Additionally, the harvested cells themselves may not be appropriate in number and/or quality for therapeutic purposes (e.g., in case of elder patients). More than that, harvesting might be a painful and complicated procedure to the patient (e.g., BMSCs). Cells from perinatal tissues could be used for autologous administration, but this practice would be very resource demanding, comprising long term storage of the isolated cells in freezers until they are eventually needed[98].

It is not consensual if allogeneic delivery of MSCs would lead to immune rejection. Actually, many findings with somatic stem cells showed that they possess immunossupressive action, not being rejected by the host organism[99-102]. If lack of immune rejection becomes confirmed for allogeneic somatic stem cell transplants, this would certainly represent the preferred option, making it possible that cultured cells are readily available for the patients. ESCs, by the other hand, are known to trigger immune responses following allogeneic transplantation. Taking that into consideration, research is being conducted in order to inhibit donor-host immune reactions[103].

Stem cell therapeutic effects and underlying mechanisms

It was in the beginning of the last decade that stem cells started to be considered as a treatment for neurological disorders. Back then, the expected therapeutic mechanism was very simple: stem cells were expected to repopulate the damaged tissue, differentiating into functional neurons and glial cells[104,105]. Actually, a considerable number of earlier studies have mistakenly concluded that MSCs and UCBCs could in fact differentiate into functional mature neurons and astrocytes, but these findings were contested[84,106]. NSCs are the only type of somatic stem cell capable to differentiate into functional mature nervous cells. But again, probably it is not the main action underlying NSC therapeutic effects. The underlying mechanisms are presently recognized to be far more complex, and are still not fully understood.

Evidence show that therapeutic effects primarily emerge from the release of trophic factors, cytokines, and microRNAs that stimulate endogenous mechanisms of repair[106-109]. In consonance with this, in a recent work, Jeon et al[110] reported a modest, yet significant recovery in rats subjected to ICH treated only with ASC extract. Additionally, different research groups already reported that endogenous NSCs are stimulated and recruited to the injury site in response to exogenous stem cell injection[111,112]. In fact, the injury itself seems to stimulate stem cells, exogenous and endogenous, to migrate to the perihematoma perimeter[113-117] and to promote proliferation and plasticity[108,111,118,119]. These restorative reactions are probably consequent of the secretion of trophic factors, which is the most likely mechanism underlying apoptosis prevention as well[111,113,119-121]. The same is true to the intensified angiogenesis, presumably due to elevated VEGF release[66,113,122,123]. Hematoma area reduction is also often observed[66,115,119,121,124], but not always[112-114,117]. Likewise, decrement of brain edema volume is a commonly stated benefice[113,125,126]. Stem cells are also seen to exert important anti-inflammatory effects in the injury site[66,113,125,126], being known to modulate the action of dendritic cells, and B and T lymphocytes[127-129]. Stem cells were linked to astrocyte and microglia response modulation as well[130]. In fact, recent findings showed that some glial cells within the oligodendrocytic and astrocytic lineages present progenitor and neural stem cell functions, with active response to brain injury[131,132].

Preclinical studies: an heterogeneous approach for an heterogeneous disease

Present available data concerning stem cell therapy for ICH derives from investigations conducted with notably different approaches. Many stem cell administration routes have been tested, but it remains unclear which is the best option. Findings in ICH model support that cervical vein delivery is inefficient[133], whereas good outcomes were already observed following stem cell delivery in brain parenchyma[66,111,113-116,121,126,134-137], lateral ventricle[120,133], tail vein[117,119,122,124-126,138,139], and carotid artery[133,140,141]. However, each one of these alternatives has drawbacks. Intracerebral and intracerebroventricular routes are very invasive alternatives, hardly representing the best clinical options. In this sense, intravascular injections are incomparably more feasible and less troublesome, and allows injections with higher cell doses[142]. However, due to their size, stem cells were reported to get trapped in the lungs, liver and spleen and to cause embolisms, following tail vein and intracarotid injections, respectively[143-145]. Importantly, the latter adversity can be surpassed with slower injection rates[143], a generally overlooked variable. It was reported that carotid artery delivery favors cell migration to the brain if compared to tail vein delivery route[146]. However, no differences in this aspect were seen when comparing carotid artery and jugular vein stem cell injections[147]. Additionally, concerning intravascular transplantation, the potential of other systemic interactions is of unknown impact, being the safety of this route debatable.

Recently, efforts have been devoted to the development of new cell administration routes. Among these, intranasal cell delivery represents a promising alternative for the treatment of neurological disorders. Not only intranasal administration is simple and noninvasive, but also avoids potentially dangerous systemic interactions[148-151]. Positive results were reported following the delivery of growth factors[152-154] and MSCs[155-158] following experimentally-induced ischemic stroke in rodents. To our knowledge to date, no intranasal delivery trials were conducted on the treatment of ICH using MSCs.

Some comparative studies using different routes have been made on ischemic stroke models. Findings comparing cell migration and possible systemic adversities resulting from different stem cell injection routes represent potential lessons to apply in future ICH model investigations.

Stem cells have been delivered at many different times after experimental ICH onset, ranging from one hour to two months following ictus (Table 1). Apparently, earlier NSC transplantation (acute phase) is particularly important in intravenous delivery[126]. Interestingly, in the same study, time matter seemed to be indifferent when stem cell injection was intracerebral. Also working with NSCs, other group tested the effects of different carotid artery injection times. Acute phase delivery, in this case, returned the worst therapeutic effects[140]. In late transplantation studies, intracerebral stem cell implantation was performed only two months after ictus. Even so, good functional and morphological outcomes could be observed[111,136]. It is also important to keep in mind that ICH dynamics in rats and humans, as previously discussed, are clearly different in many aspects, including temporality. Thus, time windows in these different organisms are proportionally divergent, e.g., 24 h after ICH onset, rodents and humans suffer from different pathological actions, due to the different paces in the development of the pathology.

Table 1 Methodologies used in preclinical trials of stem cell therapy for intracerebral hemorrhage.
Ref.ICH modelCellsTime after ICHAmountVol (μL)Delivery site
Bao et al[113]Col VIIhBMSC (Flk-1+)24 h2 × 10515BP
Chen et al[120]Col IVASC48 h3 × 10610LV
Fatar et al[112]Col IVhASC24 h3 × 106500TV
Jeon et al[110]Col VIIASC extract1 h--IP
Jeong et al[138]Col IVhNSC24 h5 × 106500TV
Kim et al[125]Col VIIhASC24 h3 × 1062000TV
Lee et al[126]Col VIIhNSC2 h; 24 h5 × 106 (TV); 1 × 106 (IC)500 (TV); 2 (BP)TV; BP
Li et al[139]Col VIINSC2 h; 7 h; 14 h; 21 h; 27 h4 × 106400CA
Liang et al[114]Col IVBMSC24 h1 × 10610BP
Liao et al[66]Col VIIhUCSC24 h2 × 10510BP
Nan et al[158]Col VIIhUCB24 h2.4 × 106-3.2 × 106500SV
Otero et al[134]Col IVBMSC72 h2 × 10610BP
Otero et al[115]Col IVBMSC2 m5 × 10615BP
Otero et al[111]Col IVBMSC2 h2 × 10615BP
Qin et al[116]Col VIIiPSC24 h2 × 10610BP
Qin et al[135]Col VIIiPSC24 h1 × 10610BP
Seghatoleslam et al[124]Col IVhUCBMC24 h4 × 106; 8 × 106; 16 × 1061000TV
Seyfried et al[140]ABIBMSC24 h1 × 106100CA
Seyfried et al[122]ABIBMSC24 h5 × 105; 1 × 1061000; 2000TV
Vaquero et al[136]Col IVhBMSC12 m5 × 10630BP
Wang et al[119]Col VIIBMSC1 h1 × 1061000TV
Wang et al[137]Col VIINSC72 h1 × 106500 (TV); 2 (BP)TV; BP
Yang et al[121]Col IBMSC (overexpressing GDNF)72 h5 × 10520BP
Yang et al[117]ABIhUCSC24 h; 72 h; 7 d3 × 1062000TV
Yang et al[159]Col VIIhASC24 h1 × 106200FV
Zhang et al[133]Col VIIBMSC24 h; 72 h; 5 d; 7 d2 × 10620CA; CV; LV

The ideal amount of cells and vehicle to be injected is another unanswered question. The only studies performed with variations in that parameters used the tail vein route[122,122,124]. BMSC injection of 1 million cells was more therapeutically effective than half of it[122,122]. UCMCs were also more effective in the highest tested amount (16 million cells), reducing injury area with more success[124]. However, apart from this studies, the most common quantity injected in the tail vein is of 3 million cells, suspended in 1 or 2 mL of saline solution[117,119,124,125]. The amount of intracerebrally grafted cells varied up to 25 fold among studies[66,113,115,121,136]. Interestingly, in general, the vehicle volume is not necessarily proportional with the number of cells in suspension (Table 1).

Most of the aforementioned findings, however, are very hard to counterweigh and take as conclusive. Not only the time window after ICH varied between studies, but also the cell type, amount, and injection site. Moreover, it must be never forgotten that rats and humans are different organisms, and studies in more models must be performed before calculating the extrapolation to human applications. As stated in previous sections, different stem cell types have different attributes, and knowledge is in its youth about how all these divergences may affect ICH outcome.

CONCLUSION

In this review, we highlighted that there is still much to be learned about ICH and stem cells, both individually as interacting with each other. The approaches adopted by different groups were diverse, concerning not only the outcome evaluation, but also ICH induction and stem cell treatment protocols. Although it is important to perform tests under diverse conditions, more comparative studies should be motivated, including different times for interventions, stem cell types, quantities, and delivery sites. Homogeneity of all implicated variables except the one to be tested is imperative for the improvement of our understanding, preventing waste of hopes and resources in forthcoming human ICH trials.

Footnotes

P- Reviewer: Kim MS, Lee T, Zaminy A S- Editor: Ji FF L- Editor: A E- Editor: Lu YJ

References
1.  Provencio JJ, Da Silva IR, Manno EM. Intracerebral hemorrhage: new challenges and steps forward. Neurosurg Clin N Am. 2013;24:349-359.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 26]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
2.  Katsuki H. Exploring neuroprotective drug therapies for intracerebral hemorrhage. J Pharmacol Sci. 2010;114:366-378.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 46]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
3.  Qureshi AI, Mendelow AD, Hanley DF. Intracerebral haemorrhage. Lancet. 2009;373:1632-1644.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1159]  [Cited by in F6Publishing: 1088]  [Article Influence: 72.5]  [Reference Citation Analysis (0)]
4.  Bibber B, Sinha G, Lobba AR, Greco SJ, Rameshwar P. A review of stem cell translation and potential confounds by cancer stem cells. Stem Cells Int. 2013;2013:241048.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 12]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
5.  Bliss T, Guzman R, Daadi M, Steinberg GK. Cell transplantation therapy for stroke. Stroke. 2007;38:817-826.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 281]  [Cited by in F6Publishing: 260]  [Article Influence: 15.3]  [Reference Citation Analysis (0)]
6.  Kan I, Melamed E, Offen D. Autotransplantation of bone marrow-derived stem cells as a therapy for neurodegenerative diseases. Handb Exp Pharmacol. 2007;219-242.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 46]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
7.  World Health Organization Global atlas on cardiovascular disease prevention and control. Geneva: Shandi Mendis, Pekka Puska and Bo Norrving 2011; .  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Kim AS, Johnston SC. Global variation in the relative burden of stroke and ischemic heart disease. Circulation. 2011;124:314-323.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 243]  [Cited by in F6Publishing: 272]  [Article Influence: 20.9]  [Reference Citation Analysis (0)]
9.  Hua Y, Keep RF, Hoff JT, Xi G. Brain injury after intracerebral hemorrhage: the role of thrombin and iron. Stroke. 2007;38:759-762.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 203]  [Cited by in F6Publishing: 204]  [Article Influence: 12.0]  [Reference Citation Analysis (0)]
10.  Poon MT, Fonville AF, Al-Shahi Salman R. Long-term prognosis after intracerebral haemorrhage: systematic review and meta-analysis. J Neurol Neurosurg Psychiatry. 2014;85:660-667.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 356]  [Cited by in F6Publishing: 381]  [Article Influence: 38.1]  [Reference Citation Analysis (0)]
11.  Tsai CF, Thomas B, Sudlow CL. Epidemiology of stroke and its subtypes in Chinese vs white populations: a systematic review. Neurology. 2013;81:264-272.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 313]  [Cited by in F6Publishing: 336]  [Article Influence: 30.5]  [Reference Citation Analysis (0)]
12.  van Asch CJ, Luitse MJ, Rinkel GJ, van der Tweel I, Algra A, Klijn CJ. Incidence, case fatality, and functional outcome of intracerebral haemorrhage over time, according to age, sex, and ethnic origin: a systematic review and meta-analysis. Lancet Neurol. 2010;9:167-176.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1514]  [Cited by in F6Publishing: 1691]  [Article Influence: 120.8]  [Reference Citation Analysis (0)]
13.  Hozawa A. Attributable fractions of risk factors for cardiovascular diseases. J Epidemiol. 2011;21:81-86.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 16]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
14.  Rapsomaniki E, Timmis A, George J, Pujades-Rodriguez M, Shah AD, Denaxas S, White IR, Caulfield MJ, Deanfield JE, Smeeth L. Blood pressure and incidence of twelve cardiovascular diseases: lifetime risks, healthy life-years lost, and age-specific associations in 1·25 million people. Lancet. 2014;383:1899-1911.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1150]  [Cited by in F6Publishing: 1028]  [Article Influence: 102.8]  [Reference Citation Analysis (0)]
15.  Perkovic V, Huxley R, Wu Y, Prabhakaran D, MacMahon S. The burden of blood pressure-related disease: a neglected priority for global health. Hypertension. 2007;50:991-997.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 227]  [Cited by in F6Publishing: 241]  [Article Influence: 14.2]  [Reference Citation Analysis (0)]
16.  Klatsky AL, Friedman GD, Siegelaub AB, Gérard MJ. Alcohol consumption and blood pressure Kaiser-Permanente Multiphasic Health Examination data. N Engl J Med. 1977;296:1194-1200.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Klatsky AL, Friedman GD, Armstrong MA. The relationships between alcoholic beverage use and other traits to blood pressure: a new Kaiser Permanente study. Circulation. 1986;73:628-636.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 196]  [Cited by in F6Publishing: 202]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
18.  Marchi KC, Muniz JJ, Tirapelli CR. Hypertension and chronic ethanol consumption: What do we know after a century of study? World J Cardiol. 2014;6:283-294.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 61]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
19.  Kurth T, Kase CS, Berger K, Schaeffner ES, Buring JE, Gaziano JM. Smoking and the risk of hemorrhagic stroke in men. Stroke. 2003;34:1151-1155.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 114]  [Cited by in F6Publishing: 94]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
20.  Kurth T, Kase CS, Berger K, Gaziano JM, Cook NR, Buring JE. Smoking and risk of hemorrhagic stroke in women. Stroke. 2003;34:2792-2795.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 102]  [Cited by in F6Publishing: 113]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
21.  Lee CD, Folsom AR, Blair SN. Physical activity and stroke risk: a meta-analysis. Stroke. 2003;34:2475-2481.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 517]  [Cited by in F6Publishing: 474]  [Article Influence: 22.6]  [Reference Citation Analysis (0)]
22.  Strazzullo P, D’Elia L, Kandala NB, Cappuccio FP. Salt intake, stroke, and cardiovascular disease: meta-analysis of prospective studies. BMJ. 2009;339:b4567.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 977]  [Cited by in F6Publishing: 1007]  [Article Influence: 67.1]  [Reference Citation Analysis (0)]
23.  Go AS, Mozaffarian D, Roger VL, Benjamin EJ, Berry JD, Blaha MJ, Dai S, Ford ES, Fox CS, Franco S. Heart disease and stroke statistics--2014 update: a report from the American Heart Association. Circulation. 2014;129:e28-e292.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3046]  [Cited by in F6Publishing: 3492]  [Article Influence: 349.2]  [Reference Citation Analysis (0)]
24.  Manno EM, Atkinson JL, Fulgham JR, Wijdicks EF. Emerging medical and surgical management strategies in the evaluation and treatment of intracerebral hemorrhage. Mayo Clin Proc. 2005;80:420-433.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 72]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
25.  Yamada M. Brain hemorrhages in cerebral amyloid angiopathy. Semin Thromb Hemost. 2013;39:955-962.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 8]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
26.  Mendel TA, Wierzba-Bobrowicz T, Lewandowska E, Stępień T, Szpak GM. The development of cerebral amyloid angiopathy in cerebral vessels. A review with illustrations based upon own investigated post mortem cases. Pol J Pathol. 2013;64:260-267.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 13]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
27.  Viswanathan A, Greenberg SM. Cerebral amyloid angiopathy in the elderly. Ann Neurol. 2011;70:871-880.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 375]  [Cited by in F6Publishing: 406]  [Article Influence: 33.8]  [Reference Citation Analysis (0)]
28.  Rost NS, Greenberg SM, Rosand J. The genetic architecture of intracerebral hemorrhage. Stroke. 2008;39:2166-2173.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 66]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
29.  Rosand J, Eckman MH, Knudsen KA, Singer DE, Greenberg SM. The effect of warfarin and intensity of anticoagulation on outcome of intracerebral hemorrhage. Arch Intern Med. 2004;164:880-884.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 443]  [Cited by in F6Publishing: 419]  [Article Influence: 21.0]  [Reference Citation Analysis (0)]
30.  Flaherty ML, Haverbusch M, Sekar P, Kissela B, Kleindorfer D, Moomaw CJ, Sauerbeck L, Schneider A, Broderick JP, Woo D. Long-term mortality after intracerebral hemorrhage. Neurology. 2006;66:1182-1186.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 276]  [Cited by in F6Publishing: 289]  [Article Influence: 16.1]  [Reference Citation Analysis (0)]
31.  Qureshi AI, Tuhrim S, Broderick JP, Batjer HH, Hondo H, Hanley DF. Spontaneous intracerebral hemorrhage. N Engl J Med. 2001;344:1450-1460.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1182]  [Cited by in F6Publishing: 1090]  [Article Influence: 47.4]  [Reference Citation Analysis (0)]
32.  Xi G, Keep RF, Hoff JT. Mechanisms of brain injury after intracerebral haemorrhage. Lancet Neurol. 2006;5:53-63.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 977]  [Cited by in F6Publishing: 1032]  [Article Influence: 57.3]  [Reference Citation Analysis (0)]
33.  Takebayashi S, Kaneko M. Electron microscopic studies of ruptured arteries in hypertensive intracerebral hemorrhage. Stroke. 1983;14:28-36.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 81]  [Cited by in F6Publishing: 78]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
34.  Tuhrim S, Horowitz DR, Sacher M, Godbold JH. Volume of ventricular blood is an important determinant of outcome in supratentorial intracerebral hemorrhage. Crit Care Med. 1999;27:617-621.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 311]  [Cited by in F6Publishing: 296]  [Article Influence: 11.8]  [Reference Citation Analysis (0)]
35.  Miller JH, Wardlaw JM, Lammie GA. Intracerebral haemorrhage and cerebral amyloid angiopathy: CT features with pathological correlation. Clin Radiol. 1999;54:422-429.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 38]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
36.  Broderick JP, Brott TG, Duldner JE, Tomsick T, Huster G. Volume of intracerebral hemorrhage. A powerful and easy-to-use predictor of 30-day mortality. Stroke. 1993;24:987-993.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1119]  [Cited by in F6Publishing: 1102]  [Article Influence: 35.5]  [Reference Citation Analysis (0)]
37.  Fujii Y, Tanaka R, Takeuchi S, Koike T, Minakawa T, Sasaki O. Hematoma enlargement in spontaneous intracerebral hemorrhage. J Neurosurg. 1994;80:51-57.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 246]  [Cited by in F6Publishing: 254]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
38.  Flibotte JJ, Hagan N, O’Donnell J, Greenberg SM, Rosand J. Warfarin, hematoma expansion, and outcome of intracerebral hemorrhage. Neurology. 2004;63:1059-1064.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 439]  [Cited by in F6Publishing: 419]  [Article Influence: 22.1]  [Reference Citation Analysis (0)]
39.  Rosell A, Vilalta A, García-Berrocoso T, Fernández-Cadenas I, Domingues-Montanari S, Cuadrado E, Delgado P, Ribó M, Martínez-Sáez E, Ortega-Aznar A. Brain perihematoma genomic profile following spontaneous human intracerebral hemorrhage. PLoS One. 2011;6:e16750.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 48]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
40.  Brott T, Broderick J, Kothari R, Barsan W, Tomsick T, Sauerbeck L, Spilker J, Duldner J, Khoury J. Early hemorrhage growth in patients with intracerebral hemorrhage. Stroke. 1997;28:1-5.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 935]  [Cited by in F6Publishing: 997]  [Article Influence: 36.9]  [Reference Citation Analysis (0)]
41.  Davis SM, Broderick J, Hennerici M, Brun NC, Diringer MN, Mayer SA, Begtrup K, Steiner T. Hematoma growth is a determinant of mortality and poor outcome after intracerebral hemorrhage. Neurology. 2006;66:1175-1181.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 766]  [Cited by in F6Publishing: 762]  [Article Influence: 42.3]  [Reference Citation Analysis (0)]
42.  Butcher KS, Baird T, MacGregor L, Desmond P, Tress B, Davis S. Perihematomal edema in primary intracerebral hemorrhage is plasma derived. Stroke. 2004;35:1879-1885.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 139]  [Cited by in F6Publishing: 143]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
43.  Inaji M, Tomita H, Tone O, Tamaki M, Suzuki R, Ohno K. Chronological changes of perihematomal edema of human intracerebral hematoma. Acta Neurochir Suppl. 2003;86:445-448.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 50]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
44.  Venkatasubramanian C, Mlynash M, Finley-Caulfield A, Eyngorn I, Kalimuthu R, Snider RW, Wijman CA. Natural history of perihematomal edema after intracerebral hemorrhage measured by serial magnetic resonance imaging. Stroke. 2011;42:73-80.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 136]  [Cited by in F6Publishing: 153]  [Article Influence: 10.9]  [Reference Citation Analysis (0)]
45.  Xi G, Keep RF, Hoff JT. Pathophysiology of brain edema formation. Neurosurg Clin N Am. 2002;13:371-383.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 154]  [Cited by in F6Publishing: 161]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
46.  Xi G, Reiser G, Keep RF. The role of thrombin and thrombin receptors in ischemic, hemorrhagic and traumatic brain injury: deleterious or protective? J Neurochem. 2003;84:3-9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 266]  [Cited by in F6Publishing: 272]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
47.  Bakhshayesh B, Hosseininezhad M, Seyedeh Saadat SN, Ansar MM, Ramezani H, Seyed Saadat SM. Iron Overload is Associated with Perihematoma Edema Growth following Intracerebral Hemorrhage that May Contribute to in-Hospital Mortality and Long-Term Functional Outcome. Curr Neurovasc Res. 2014;11:248-253.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 16]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
48.  Durukan A, Tatlisumak T. Acute ischemic stroke: overview of major experimental rodent models, pathophysiology, and therapy of focal cerebral ischemia. Pharmacol Biochem Behav. 2007;87:179-197.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 468]  [Cited by in F6Publishing: 501]  [Article Influence: 29.5]  [Reference Citation Analysis (0)]
49.  Kirkman MA, Allan SM, Parry-Jones AR. Experimental intracerebral hemorrhage: avoiding pitfalls in translational research. J Cereb Blood Flow Metab. 2011;31:2135-2151.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 52]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
50.  Hardman CD, Henderson JM, Finkelstein DI, Horne MK, Paxinos G, Halliday GM. Comparison of the basal ganglia in rats, marmosets, macaques, baboons, and humans: volume and neuronal number for the output, internal relay, and striatal modulating nuclei. J Comp Neurol. 2002;445:238-255.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 177]  [Cited by in F6Publishing: 182]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
51.  Fisher M, Feuerstein G, Howells DW, Hurn PD, Kent TA, Savitz SI, Lo EH. Update of the stroke therapy academic industry roundtable preclinical recommendations. Stroke. 2009;40:2244-2250.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 919]  [Cited by in F6Publishing: 977]  [Article Influence: 65.1]  [Reference Citation Analysis (0)]
52.  Chopp M, Steinberg GK, Kondziolka D, Lu M, Bliss TM, Li Y, Hess DC, Borlongan CV. Who’s in favor of translational cell therapy for stroke: STEPS forward please? Cell Transplant. 2009;18:691-693.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 45]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
53.  Bullock R, Mendelow AD, Teasdale GM, Graham DI. Intracranial haemorrhage induced at arterial pressure in the rat. Part 1: Description of technique, ICP changes and neuropathological findings. Neurol Res. 1984;6:184-188.  [PubMed]  [DOI]  [Cited in This Article: ]
54.  James ML, Warner DS, Laskowitz DT. Preclinical models of intracerebral hemorrhage: a translational perspective. Neurocrit Care. 2008;9:139-152.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 72]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
55.  Kleinig TJ, Helps SC, Ghabriel MN, Manavis J, Leigh C, Blumbergs PC, Vink R. Hemoglobin crystals: a pro-inflammatory potential confounder of rat experimental intracerebral hemorrhage. Brain Res. 2009;1287:164-172.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 13]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
56.  Harris ED, Krane SM. Collagenases (third of three parts). N Engl J Med. 1974;291:652-661.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 90]  [Cited by in F6Publishing: 104]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
57.  Rosenberg GA, Mun-Bryce S, Wesley M, Kornfeld M. Collagenase-induced intracerebral hemorrhage in rats. Stroke. 1990;21:801-807.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 416]  [Cited by in F6Publishing: 426]  [Article Influence: 12.5]  [Reference Citation Analysis (0)]
58.  MacLellan CL, Langdon KD, Churchill KP, Granter-Button S, Corbett D. Assessing cognitive function after intracerebral hemorrhage in rats. Behav Brain Res. 2009;198:321-328.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 36]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
59.  McCluskey L, Campbell S, Anthony D, Allan SM. Inflammatory responses in the rat brain in response to different methods of intra-cerebral administration. J Neuroimmunol. 2008;194:27-33.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 32]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
60.  Levine JM, Snider R, Finkelstein D, Gurol ME, Chanderraj R, Smith EE, Greenberg SM, Rosand J. Early edema in warfarin-related intracerebral hemorrhage. Neurocrit Care. 2007;7:58-63.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 37]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
61.  Gong Y, Hua Y, Keep RF, Hoff JT, Xi G. Intracerebral hemorrhage: effects of aging on brain edema and neurological deficits. Stroke. 2004;35:2571-2575.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 110]  [Cited by in F6Publishing: 116]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
62.  Gong Y, He Y, Gu Y, Keep RF, Xi G, Hua Y. Effects of aging on autophagy after experimental intracerebral hemorrhage. Acta Neurochir Suppl. 2011;111:113-117.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 21]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
63.  Wasserman JK, Yang H, Schlichter LC. Glial responses, neuron death and lesion resolution after intracerebral hemorrhage in young vs. aged rats. Eur J Neurosci. 2008;28:1316-1328.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in F6Publishing: 86]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
64.  Wu G, Bao X, Xi G, Keep RF, Thompson BG, Hua Y. Brain injury after intracerebral hemorrhage in spontaneously hypertensive rats. J Neurosurg. 2011;114:1805-1811.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 23]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
65.  Sadoshima S, Busija D, Brody M, Heistad D. Sympathetic nerves protect against stroke in stroke-prone hypertensive rats. A preliminary report. Hypertension. 1981;3:I124-I127.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 53]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
66.  Liao W, Zhong J, Yu J, Xie J, Liu Y, Du L, Yang S, Liu P, Xu J, Wang J. Therapeutic benefit of human umbilical cord derived mesenchymal stromal cells in intracerebral hemorrhage rat: implications of anti-inflammation and angiogenesis. Cell Physiol Biochem. 2009;24:307-316.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 100]  [Cited by in F6Publishing: 107]  [Article Influence: 7.1]  [Reference Citation Analysis (0)]
67.  Hua Y, Schallert T, Keep RF, Wu J, Hoff JT, Xi G. Behavioral tests after intracerebral hemorrhage in the rat. Stroke. 2002;33:2478-2484.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 393]  [Cited by in F6Publishing: 401]  [Article Influence: 18.2]  [Reference Citation Analysis (0)]
68.  Xi G, Keep RF, Hoff JT. Erythrocytes and delayed brain edema formation following intracerebral hemorrhage in rats. J Neurosurg. 1998;89:991-996.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 259]  [Cited by in F6Publishing: 270]  [Article Influence: 10.4]  [Reference Citation Analysis (0)]
69.  Zazulia AR, Diringer MN, Derdeyn CP, Powers WJ. Progression of mass effect after intracerebral hemorrhage. Stroke. 1999;30:1167-1173.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 254]  [Cited by in F6Publishing: 275]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
70.  Nishibe M, Barbay S, Guggenmos D, Nudo RJ. Reorganization of motor cortex after controlled cortical impact in rats and implications for functional recovery. J Neurotrauma. 2010;27:2221-2232.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 65]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
71.  Puurunen K, Jolkkonen J, Sirviö J, Haapalinna A, Sivenius J. An alpha(2)-adrenergic antagonist, atipamezole, facilitates behavioral recovery after focal cerebral ischemia in rats. Neuropharmacology. 2001;40:597-606.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 83]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
72.  Chen J, Li Y, Wang L, Lu M, Zhang X, Chopp M. Therapeutic benefit of intracerebral transplantation of bone marrow stromal cells after cerebral ischemia in rats. J Neurol Sci. 2001;189:49-57.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 422]  [Cited by in F6Publishing: 446]  [Article Influence: 19.4]  [Reference Citation Analysis (0)]
73.  Bliss TM, Andres RH, Steinberg GK. Optimizing the success of cell transplantation therapy for stroke. Neurobiol Dis. 2010;37:275-283.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 138]  [Cited by in F6Publishing: 152]  [Article Influence: 10.1]  [Reference Citation Analysis (0)]
74.  Ishii T, Eto K. Fetal stem cell transplantation: Past, present, and future. World J Stem Cells. 2014;6:404-420.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 52]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
75.  Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126:663-676.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17989]  [Cited by in F6Publishing: 17070]  [Article Influence: 948.3]  [Reference Citation Analysis (0)]
76.  Blum B, Benvenisty N. The tumorigenicity of human embryonic stem cells. Adv Cancer Res. 2008;100:133-158.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 300]  [Cited by in F6Publishing: 167]  [Article Influence: 10.4]  [Reference Citation Analysis (0)]
77.  Nishimori M, Yakushiji H, Mori M, Miyamoto T, Yaguchi T, Ohno S, Miyake Y, Sakaguchi T, Ueda M, Ohno E. Tumorigenesis in cells derived from induced pluripotent stem cells. Hum Cell. 2014;27:29-35.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 24]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
78.  Miura K, Okada Y, Aoi T, Okada A, Takahashi K, Okita K, Nakagawa M, Koyanagi M, Tanabe K, Ohnuki M. Variation in the safety of induced pluripotent stem cell lines. Nat Biotechnol. 2009;27:743-745.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 669]  [Cited by in F6Publishing: 701]  [Article Influence: 46.7]  [Reference Citation Analysis (0)]
79.  Simerman AA, Perone MJ, Gimeno ML, Dumesic DA, Chazenbalk GD. A mystery unraveled: nontumorigenic pluripotent stem cells in human adult tissues. Expert Opin Biol Ther. 2014;14:917-929.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 16]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
80.  Monti M, Perotti C, Del Fante C, Cervio M, Redi CA. Stem cells: sources and therapies. Biol Res. 2012;45:207-214.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 15]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
81.  da Silva Meirelles L, Chagastelles PC, Nardi NB. Mesenchymal stem cells reside in virtually all post-natal organs and tissues. J Cell Sci. 2006;119:2204-2213.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1715]  [Cited by in F6Publishing: 1652]  [Article Influence: 91.8]  [Reference Citation Analysis (0)]
82.  Augello A, De Bari C. The regulation of differentiation in mesenchymal stem cells. Hum Gene Ther. 2010;21:1226-1238.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 250]  [Cited by in F6Publishing: 253]  [Article Influence: 19.5]  [Reference Citation Analysis (0)]
83.  Murray IR, West CC, Hardy WR, James AW, Park TS, Nguyen A, Tawonsawatruk T, Lazzari L, Soo C, Péault B. Natural history of mesenchymal stem cells, from vessel walls to culture vessels. Cell Mol Life Sci. 2014;71:1353-1374.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 206]  [Cited by in F6Publishing: 188]  [Article Influence: 18.8]  [Reference Citation Analysis (0)]
84.  Barnabé GF, Schwindt TT, Calcagnotto ME, Motta FL, Martinez G, de Oliveira AC, Keim LM, D’Almeida V, Mendez-Otero R, Mello LE. Chemically-induced RAT mesenchymal stem cells adopt molecular properties of neuronal-like cells but do not have basic neuronal functional properties. PLoS One. 2009;4:e5222.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 67]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
85.  Wegmeyer H, Bröske AM, Leddin M, Kuentzer K, Nisslbeck AK, Hupfeld J, Wiechmann K, Kuhlen J, von Schwerin C, Stein C. Mesenchymal stromal cell characteristics vary depending on their origin. Stem Cells Dev. 2013;22:2606-2618.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 148]  [Cited by in F6Publishing: 156]  [Article Influence: 14.2]  [Reference Citation Analysis (0)]
86.  Casteilla L, Planat-Benard V, Laharrague P, Cousin B. Adipose-derived stromal cells: Their identity and uses in clinical trials, an update. World J Stem Cells. 2011;3:25-33.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 164]  [Cited by in F6Publishing: 401]  [Article Influence: 30.8]  [Reference Citation Analysis (0)]
87.  Baer PC. Adipose-derived mesenchymal stromal/stem cells: An update on their phenotype in vivo and in vitro. World J Stem Cells. 2014;6:256-265.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 126]  [Cited by in F6Publishing: 121]  [Article Influence: 12.1]  [Reference Citation Analysis (0)]
88.  Kim EH, Heo CY. Current applications of adipose-derived stem cells and their future perspectives. World J Stem Cells. 2014;6:65-68.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 55]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
89.  Gutiérrez-Fernández M, Rodríguez-Frutos B, Otero-Ortega L, Ramos-Cejudo J, Fuentes B, Díez-Tejedor E. Adipose tissue-derived stem cells in stroke treatment: from bench to bedside. Discov Med. 2013;16:37-43.  [PubMed]  [DOI]  [Cited in This Article: ]
90.  Ong WK, Sugii S. Adipose-derived stem cells: fatty potentials for therapy. Int J Biochem Cell Biol. 2013;45:1083-1086.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 94]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
91.  Banerjee S, Williamson DA, Habib N, Chataway J. The potential benefit of stem cell therapy after stroke: an update. Vasc Health Risk Manag. 2012;8:569-580.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 47]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
92.  Kim JY, Jeon HB, Yang YS, Oh W, Chang JW. Application of human umbilical cord blood-derived mesenchymal stem cells in disease models. World J Stem Cells. 2010;2:34-38.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 65]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
93.  Nagamura-Inoue T, He H. Umbilical cord-derived mesenchymal stem cells: Their advantages and potential clinical utility. World J Stem Cells. 2014;6:195-202.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 258]  [Cited by in F6Publishing: 251]  [Article Influence: 25.1]  [Reference Citation Analysis (4)]
94.  Ilancheran S, Moodley Y, Manuelpillai U. Human fetal membranes: a source of stem cells for tissue regeneration and repair? Placenta. 2009;30:2-10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 189]  [Cited by in F6Publishing: 183]  [Article Influence: 11.4]  [Reference Citation Analysis (0)]
95.  Kern S, Eichler H, Stoeve J, Klüter H, Bieback K. Comparative analysis of mesenchymal stem cells from bone marrow, umbilical cord blood, or adipose tissue. Stem Cells. 2006;24:1294-1301.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2248]  [Cited by in F6Publishing: 2224]  [Article Influence: 123.6]  [Reference Citation Analysis (0)]
96.  Heneidi S, Simerman AA, Keller E, Singh P, Li X, Dumesic DA, Chazenbalk G. Awakened by cellular stress: isolation and characterization of a novel population of pluripotent stem cells derived from human adipose tissue. PLoS One. 2013;8:e64752.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in F6Publishing: 98]  [Article Influence: 8.9]  [Reference Citation Analysis (0)]
97.  Ogura F, Wakao S, Kuroda Y, Tsuchiyama K, Bagheri M, Heneidi S, Chazenbalk G, Aiba S, Dezawa M. Human adipose tissue possesses a unique population of pluripotent stem cells with nontumorigenic and low telomerase activities: potential implications in regenerative medicine. Stem Cells Dev. 2014;23:717-728.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 100]  [Cited by in F6Publishing: 108]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
98.  Bieback K, Brinkmann I. Mesenchymal stromal cells from human perinatal tissues: From biology to cell therapy. World J Stem Cells. 2010;2:81-92.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in F6Publishing: 78]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
99.  Puissant B, Barreau C, Bourin P, Clavel C, Corre J, Bousquet C, Taureau C, Cousin B, Abbal M, Laharrague P. Immunomodulatory effect of human adipose tissue-derived adult stem cells: comparison with bone marrow mesenchymal stem cells. Br J Haematol. 2005;129:118-129.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 685]  [Cited by in F6Publishing: 663]  [Article Influence: 34.9]  [Reference Citation Analysis (0)]
100.  Coulson-Thomas VJ, Gesteira TF, Hascall V, Kao W. Umbilical cord mesenchymal stem cells suppress host rejection: the role of the glycocalyx. J Biol Chem. 2014;289:23465-23481.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 66]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
101.  Puissant-Lubrano B, Huynh A, Attal M, Blancher A. Evolution of peripheral blood T lymphocyte subsets after allogenic or autologous hematopoietic stem cell transplantation. Immunobiology. 2014;219:611-618.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 5]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
102.  Jeong SH, Ji YH, Yoon ES. Immunosuppressive activity of adipose tissue-derived mesenchymal stem cells in a rat model of hind limb allotransplantation. Transplant Proc. 2014;46:1606-1614.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 20]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
103.  Li SC, Zhong JF. Twisting immune responses for allogeneic stem cell therapy. World J Stem Cells. 2009;1:30-35.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 9]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
104.  Rutherford A. Stemming stroke-damaged brains. Trends Mol Med. 2001;7:150.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 2]  [Article Influence: 0.1]  [Reference Citation Analysis (0)]
105.  Savitz SI. Stem cells and stroke: are we further away than anyone is willing to admit? Int J Stroke. 2012;7:34-35.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 9]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
106.  Mir O, Savitz SI. Stem cell therapy in stroke treatment: is it a viable option? Expert Rev Neurother. 2013;13:119-121.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 6]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
107.  Carmichael ST, Krakauer JW. The promise of neuro-recovery after stroke: introduction. Stroke. 2013;44:S103.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 8]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
108.  Onteniente B. The multiple aspects of stroke and stem cell therapy. Curr Mol Med. 2013;13:821-831.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 10]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
109.  Lee HK, Finniss S, Cazacu S, Xiang C, Brodie C. Mesenchymal Stem Cells Deliver Exogenous miRNAs to Neural Cells and Induce Their Differentiation and Glutamate Transporter Expression. Stem Cells Dev. 2014;23:2851-2861.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 92]  [Cited by in F6Publishing: 93]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
110.  Jeon D, Chu K, Lee ST, Jung KH, Ban JJ, Park DK, Yoon HJ, Jung S, Yang H, Kim BS. Neuroprotective effect of a cell-free extract derived from human adipose stem cells in experimental stroke models. Neurobiol Dis. 2013;54:414-420.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 33]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
111.  Otero L, Zurita M, Bonilla C, Aguayo C, Rico MA, Rodríguez A, Vaquero J. Allogeneic bone marrow stromal cell transplantation after cerebral hemorrhage achieves cell transdifferentiation and modulates endogenous neurogenesis. Cytotherapy. 2012;14:34-44.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 33]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
112.  Fatar M, Stroick M, Griebe M, Marwedel I, Kern S, Bieback K, Giesel FL, Zechmann C, Kreisel S, Vollmar F. Lipoaspirate-derived adult mesenchymal stem cells improve functional outcome during intracerebral hemorrhage by proliferation of endogenous progenitor cells stem cells in intracerebral hemorrhages. Neurosci Lett. 2008;443:174-178.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 22]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
113.  Bao XJ, Liu FY, Lu S, Han Q, Feng M, Wei JJ, Li GL, Zhao RC, Wang RZ. Transplantation of Flk-1+ human bone marrow-derived mesenchymal stem cells promotes behavioral recovery and anti-inflammatory and angiogenesis effects in an intracerebral hemorrhage rat model. Int J Mol Med. 2013;31:1087-1096.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 26]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
114.  Liang H, Yin Y, Lin T, Guan D, Ma B, Li C, Wang Y, Zhang X. Transplantation of bone marrow stromal cells enhances nerve regeneration of the corticospinal tract and improves recovery of neurological functions in a collagenase-induced rat model of intracerebral hemorrhage. Mol Cells. 2013;36:17-24.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 21]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
115.  Otero L, Zurita M, Bonilla C, Aguayo C, Vela A, Rico MA, Vaquero J. Late transplantation of allogeneic bone marrow stromal cells improves neurologic deficits subsequent to intracerebral hemorrhage. Cytotherapy. 2011;13:562-571.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 35]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
116.  Qin J, Song B, Zhang H, Wang Y, Wang N, Ji Y, Qi J, Chandra A, Yang B, Zhang Y. Transplantation of human neuro-epithelial-like stem cells derived from induced pluripotent stem cells improves neurological function in rats with experimental intracerebral hemorrhage. Neurosci Lett. 2013;548:95-100.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 24]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
117.  Yang D, Han Y, Zhang J, Seyda A, Chopp M, Seyfried DM. Therapeutic effect of human umbilical tissue-derived cell treatment in rats with experimental intracerebral hemorrhage. Brain Res. 2012;1444:1-10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 12]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
118.  Savitz SI. Introduction to cellular therapy: the next frontier for stroke therapeutics. Stroke. 2009;40:S141-S142.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 11]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
119.  Wang SP, Wang ZH, Peng DY, Li SM, Wang H, Wang XH. Therapeutic effect of mesenchymal stem cells in rats with intracerebral hemorrhage: reduced apoptosis and enhanced neuroprotection. Mol Med Rep. 2012;6:848-854.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 52]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
120.  Chen J, Tang YX, Liu YM, Chen J, Hu XQ, Liu N, Wang SX, Zhang Y, Zeng WG, Ni HJ. Transplantation of adipose-derived stem cells is associated with neural differentiation and functional improvement in a rat model of intracerebral hemorrhage. CNS Neurosci Ther. 2012;18:847-854.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 50]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
121.  Yang C, Zhou L, Gao X, Chen B, Tu J, Sun H, Liu X, He J, Liu J, Yuan Q. Neuroprotective effects of bone marrow stem cells overexpressing glial cell line-derived neurotrophic factor on rats with intracerebral hemorrhage and neurons exposed to hypoxia/reoxygenation. Neurosurgery. 2011;68:691-704.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 39]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
122.  Seyfried DM, Han Y, Yang D, Ding J, Shen LH, Savant-Bhonsale S, Chopp M. Localization of bone marrow stromal cells to the injury site after intracerebral hemorrhage in rats. J Neurosurg. 2010;112:329-335.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 30]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
123.  Dao M, Tate CC, McGrogan M, Case CC. Comparing the angiogenic potency of naïve marrow stromal cells and Notch-transfected marrow stromal cells. J Transl Med. 2013;11:81.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 25]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
124.  Seghatoleslam M, Jalali M, Nikravesh MR, Hamidi Alamdari D, Hosseini M, Fazel A. Intravenous administration of human umbilical cord blood-mononuclear cells dose-dependently relieve neurologic deficits in rat intracerebral hemorrhage model. Ann Anat. 2013;195:39-49.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 9]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
125.  Kim JM, Lee ST, Chu K, Jung KH, Song EC, Kim SJ, Sinn DI, Kim JH, Park DK, Kang KM. Systemic transplantation of human adipose stem cells attenuated cerebral inflammation and degeneration in a hemorrhagic stroke model. Brain Res. 2007;1183:43-50.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 122]  [Cited by in F6Publishing: 117]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
126.  Lee ST, Chu K, Jung KH, Kim SJ, Kim DH, Kang KM, Hong NH, Kim JH, Ban JJ, Park HK. Anti-inflammatory mechanism of intravascular neural stem cell transplantation in haemorrhagic stroke. Brain. 2008;131:616-629.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 301]  [Cited by in F6Publishing: 312]  [Article Influence: 18.4]  [Reference Citation Analysis (0)]
127.  Chamberlain G, Fox J, Ashton B, Middleton J. Concise review: mesenchymal stem cells: their phenotype, differentiation capacity, immunological features, and potential for homing. Stem Cells. 2007;25:2739-2749.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1697]  [Cited by in F6Publishing: 1616]  [Article Influence: 95.1]  [Reference Citation Analysis (0)]
128.  Herrero C, Pérez-Simón JA. Immunomodulatory effect of mesenchymal stem cells. Braz J Med Biol Res. 2010;43:425-430.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 53]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
129.  Lotfinegad P. Immunomodulatory nature and site specific affinity of mesenchymal stem cells: a hope in cell therapy. Adv Pharm Bull. 2014;4:5.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 29]  [Reference Citation Analysis (0)]
130.  Li Y, Liu Z, Xin H, Chopp M. The role of astrocytes in mediating exogenous cell-based restorative therapy for stroke. Glia. 2014;62:1-16.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 56]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
131.  Nishiyama A, Suzuki R, Zhu X. NG2 cells (polydendrocytes) in brain physiology and repair. Front Neurosci. 2014;8:133.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 74]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
132.  Dimou L, Götz M. Glial cells as progenitors and stem cells: new roles in the healthy and diseased brain. Physiol Rev. 2014;94:709-737.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 165]  [Cited by in F6Publishing: 174]  [Article Influence: 17.4]  [Reference Citation Analysis (0)]
133.  Zhang H, Huang Z, Xu Y, Zhang S. Differentiation and neurological benefit of the mesenchymal stem cells transplanted into the rat brain following intracerebral hemorrhage. Neurol Res. 2006;28:104-112.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 59]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
134.  Otero L, Bonilla C, Aguayo C, Zurita M, Vaquero J. Intralesional administration of allogeneic bone marrow stromal cells reduces functional deficits after intracerebral hemorrhage. Histol Histopathol. 2010;25:453-461.  [PubMed]  [DOI]  [Cited in This Article: ]
135.  Qin J, Gong G, Sun S, Qi J, Zhang H, Wang Y, Wang N, Wang QM, Ji Y, Gao Y. Functional recovery after transplantation of induced pluripotent stem cells in a rat hemorrhagic stroke model. Neurosci Lett. 2013;554:70-75.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 20]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
136.  Vaquero J, Otero L, Bonilla C, Aguayo C, Rico MA, Rodriguez A, Zurita M. Cell therapy with bone marrow stromal cells after intracerebral hemorrhage: impact of platelet-rich plasma scaffolds. Cytotherapy. 2013;15:33-43.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 38]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
137.  Wang Z, Cui C, Li Q, Zhou S, Fu J, Wang X, Zhuge Q. Intracerebral transplantation of foetal neural stem cells improves brain dysfunction induced by intracerebral haemorrhage stroke in mice. J Cell Mol Med. 2011;15:2624-2633.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 16]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
138.  Jeong SW, Chu K, Jung KH, Kim SU, Kim M, Roh JK. Human neural stem cell transplantation promotes functional recovery in rats with experimental intracerebral hemorrhage. Stroke. 2003;34:2258-2263.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 290]  [Cited by in F6Publishing: 298]  [Article Influence: 14.2]  [Reference Citation Analysis (0)]
139.  Li F, Liu Y, Zhu S, Wang X, Yang H, Liu C, Zhang Y, Zhang Z. Therapeutic time window and effect of intracarotid neural stem cells transplantation for intracerebral hemorrhage. Neuroreport. 2007;18:1019-1023.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 10]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
140.  Seyfried DM, Han Y, Yang D, Ding J, Savant-Bhonsale S, Shukairy MS, Chopp M. Mannitol enhances delivery of marrow stromal cells to the brain after experimental intracerebral hemorrhage. Brain Res. 2008;1224:12-19.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 47]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
141.  Du G, Liu Y, Dang M, Zhu G, Su R, Fan Y, Tan Z, Wang LX, Fang J. Comparison of administration routes for adipose-derived stem cells in the treatment of middle cerebral artery occlusion in rats. Acta Histochem. 2014;116:1075-1084.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 27]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
142.  Janowski M, Lyczek A, Engels C, Xu J, Lukomska B, Bulte JW, Walczak P. Cell size and velocity of injection are major determinants of the safety of intracarotid stem cell transplantation. J Cereb Blood Flow Metab. 2013;33:921-927.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 106]  [Cited by in F6Publishing: 111]  [Article Influence: 10.1]  [Reference Citation Analysis (0)]
143.  Ge J, Guo L, Wang S, Zhang Y, Cai T, Zhao RC, Wu Y. The size of mesenchymal stem cells is a significant cause of vascular obstructions and stroke. Stem Cell Rev. 2014;10:295-303.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 138]  [Cited by in F6Publishing: 127]  [Article Influence: 12.7]  [Reference Citation Analysis (0)]
144.  Misra V, Yang B, Sharma S, Savitz S. Cell-based therapy for stroke. Progenitor cell therapy for neurological injury. Stem Cell Biology and Regenerative Medicine. USA: Humana Press 2011; 143-161.  [PubMed]  [DOI]  [Cited in This Article: ]
145.  Byun JS, Kwak BK, Kim JK, Jung J, Ha BC, Park S. Engraftment of human mesenchymal stem cells in a rat photothrombotic cerebral infarction model: comparison of intra-arterial and intravenous infusion using MRI and histological analysis. J Korean Neurosurg Soc. 2013;54:467-476.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 19]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
146.  Vasconcelos-dos-Santos A, Rosado-de-Castro PH, Lopes de Souza SA, da Costa Silva J, Ramos AB, Rodriguez de Freitas G, Barbosa da Fonseca LM, Gutfilen B, Mendez-Otero R. Intravenous and intra-arterial administration of bone marrow mononuclear cells after focal cerebral ischemia: Is there a difference in biodistribution and efficacy? Stem Cell Res. 2012;9:1-8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 56]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
147.  Dhuria SV, Hanson LR, Frey WH. Intranasal delivery to the central nervous system: mechanisms and experimental considerations. J Pharm Sci. 2010;99:1654-1673.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 755]  [Cited by in F6Publishing: 789]  [Article Influence: 56.4]  [Reference Citation Analysis (0)]
148.  Lochhead JJ, Thorne RG. Intranasal delivery of biologics to the central nervous system. Adv Drug Deliv Rev. 2012;64:614-628.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 658]  [Cited by in F6Publishing: 708]  [Article Influence: 59.0]  [Reference Citation Analysis (0)]
149.  Danielyan L, Schäfer R, von Ameln-Mayerhofer A, Buadze M, Geisler J, Klopfer T, Burkhardt U, Proksch B, Verleysdonk S, Ayturan M. Intranasal delivery of cells to the brain. Eur J Cell Biol. 2009;88:315-324.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 226]  [Cited by in F6Publishing: 235]  [Article Influence: 15.7]  [Reference Citation Analysis (0)]
150.  van Velthoven CT, Kavelaars A, van Bel F, Heijnen CJ. Nasal administration of stem cells: a promising novel route to treat neonatal ischemic brain damage. Pediatr Res. 2010;68:419-422.  [PubMed]  [DOI]  [Cited in This Article: ]
151.  Cheng X, Wang Z, Yang J, Ma M, Lu T, Xu G, Liu X. Acidic fibroblast growth factor delivered intranasally induces neurogenesis and angiogenesis in rats after ischemic stroke. Neurol Res. 2011;33:675-680.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 33]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
152.  Ma M, Ma Y, Yi X, Guo R, Zhu W, Fan X, Xu G, Frey WH, Liu X. Intranasal delivery of transforming growth factor-beta1 in mice after stroke reduces infarct volume and increases neurogenesis in the subventricular zone. BMC Neurosci. 2008;9:117.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 95]  [Cited by in F6Publishing: 105]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
153.  Yang JP, Liu HJ, Liu XF. VEGF promotes angiogenesis and functional recovery in stroke rats. J Invest Surg. 2010;23:149-155.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 68]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
154.  Donega V, van Velthoven CT, Nijboer CH, van Bel F, Kas MJ, Kavelaars A, Heijnen CJ. Intranasal mesenchymal stem cell treatment for neonatal brain damage: long-term cognitive and sensorimotor improvement. PLoS One. 2013;8:e51253.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 123]  [Cited by in F6Publishing: 129]  [Article Influence: 11.7]  [Reference Citation Analysis (0)]
155.  Donega V, Nijboer CH, van Tilborg G, Dijkhuizen RM, Kavelaars A, Heijnen CJ. Intranasally administered mesenchymal stem cells promote a regenerative niche for repair of neonatal ischemic brain injury. Exp Neurol. 2014;261:53-64.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 105]  [Cited by in F6Publishing: 115]  [Article Influence: 11.5]  [Reference Citation Analysis (0)]
156.  van Velthoven CT, Sheldon RA, Kavelaars A, Derugin N, Vexler ZS, Willemen HL, Maas M, Heijnen CJ, Ferriero DM. Mesenchymal stem cell transplantation attenuates brain injury after neonatal stroke. Stroke. 2013;44:1426-1432.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 144]  [Cited by in F6Publishing: 149]  [Article Influence: 13.5]  [Reference Citation Analysis (0)]
157.  van Velthoven CT, Braccioli L, Willemen HL, Kavelaars A, Heijnen CJ. Therapeutic potential of genetically modified mesenchymal stem cells after neonatal hypoxic-ischemic brain damage. Mol Ther. 2014;22:645-654.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 38]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
158.  Nan Z, Grande A, Sanberg CD, Sanberg PR, Low WC. Infusion of human umbilical cord blood ameliorates neurologic deficits in rats with hemorrhagic brain injury. Ann N Y Acad Sci. 2005;1049:84-96.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 88]  [Cited by in F6Publishing: 91]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
159.  Yang KL, Lee JT, Pang CY, Lee TY, Chen SP, Liew HK, Chen SY, Chen TY, Lin PY. Human adipose-derived stem cells for the treatment of intracerebral hemorrhage in rats via femoral intravenous injection. Cell Mol Biol Lett. 2012;17:376-392.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 31]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]