更全的杂志信息网

The impact of hypoxic-ischemic brain injury on stem cell mobilization, migration, adhesion, and proliferation

更新时间:2016-07-05

Introduction

Neonatal hypoxic-ischemic encephalopathy (HIE) affects between 1 and 8 per 1000 live births in the United States each year, and up to 26 per 1000 live births in developing countries (Kurinczuk et al., 2010). Despite the standard use of therapeutic hypothermia, moderate to severe HIE continues to result in death or significant neurodevelopmental delays in 40–50% of patients (Douglas-Escobar and Weiss, 2015;Parikh and Juul, 2018). The use of stem cell transplantation has recently emerged as a promising supplemental therapy to further improve the outcomes of infants with HIE.

鄂中生态副总经理代中旭表示,鄂中生态科教短视频不仅降低了农化服务成本,还让技术服务离田间地头越来越近。目前鄂中生态已经在微信等自媒体开通了专家绿色通道,农民有任何的技术服务需求,都可以通过科教短视频、图片等迅速传送到专家团队,在1小时内解决技术需求,实行远程诊断,远程给出营养方案。

A recent pilot study assessing infants with HIE treated with hypothermia plus autologous umbilical cord blood(UCB) versus infants treated with hypothermia alone demonstrated that 74% of the UCB-treated infants survived to one year with Bayley Scales of Infant and Toddler Development III scores ≥ 85 versus 41% of the infants receiving hypothermia alone (Cotten et al., 2014). The study size was small,but it was the fi rst to demonstrate feasibility and short-term safety of UCB transplantation in this population. Although UCB transplantation has shown promise in this population,and the use of UCB avoids the ethical concerns that are raised by the use of fetal stem cells, the availability of trained staff to safely and successfully collect UCB is often limited. In addition to access concerns, the risk pro fi le of UCB transplantation has not been fully evaluated (Ballen, 2017).

As with any new therapy, the promise of stem cell transplantation to improve outcomes of neonatal HIE carries with it the need to establish the underlying mechanisms of action. Several recent studies have demonstrated that the upregulation or overexpression of factors on exogenous stem cells prior to injection can improve their migration and therapeutic effect in models of lung injury, liver failure, limb ischemia, and stroke (Cui et al., 2017; Wang et al., 2017;Xiang et al., 2017; Jimenez et al., 2018). As demonstrated by these studies, understanding the signaling mechanisms between the injured tissue and the stem cells may provide the opportunity to modify the signals through manipulation of the exogenous stem cells, allowing for improved efficacy and safety. As the types of active factors vary over time, therapies utilizing modi fi ed stem cell expression may take advantage of these variations to allow for different treatment approaches depending on the phase of injury.

Endogenous mesenchymal stem cells (MSCs) have been found to mobilize into the peripheral circulation after tissue ischemia. After mobilization, or when exogenous cells are transplanted, the cells must then migrate to the injured tissue. At the site of the injured tissue, the MSCs aid in tissue repair via paracrine mechanisms, local progenitor cell proliferation, and/or directly undergo adhesion and integration into the injured tissues (Deng et al., 2011; Rennert et al., 2012). In this paper, we review the biomarkers that have been found to be elevated in HIE (summarized in Table 1),and evaluate their roles in the mobilization, migration, cell adhesion, and proliferation of stem cells. Altering the ability of exogenous stem cells to home to injured tissue by manipulating their expression profiles could potentially improve the safety and efficacy of exogenous stem cell transplantation for neonatal brain injury.

Stem Cell Mobilization

Stem cells are localized in microenvironments known as“niches” that exist throughout the body, including the bone marrow (BM), where stem cells are maintained in undifferentiated and self-renewable states. Stem cell mobilization is the process by which stem cells are released from these niches into the peripheral circulation. Although transplanted stem cells do not require mobilization, as they are commonly injected directly into the circulation, the process of mobilization is discussed here to support the possibilitythat upregulation of certain factors on the transplanted cells could lead to increased mobilization of endogenous cells.This would be especially important in allogeneic transplants,to attempt to minimize the dose of foreign cells that would need to be used.

Table 1 Key features of the factors elevated after neonatal hypoxic-ischemic brain injury

*Derived from studies of blood samples, except where noted as in vitro or cerebrospinal fluid (CSF). BDNF: Brain-derived neurotrophic factor; EPO: erythropoietin; HIF-1: hypoxia inducible factor 1; ICAM-1: intracellular adhesion molecule 1; IL: interleukin; MCP-1: monocyte chemoattractant protein-1; MIP-1α: macrophage in fl ammatory protein-1α; MMP-9: matrix metalloproteinase 9; SCF: stem cell factor; SDF-1:stromal cell-derived factor 1; t-PA: tissue-type plasminogen activator; TNFα: tumor necrosis factor α; u-PA: urinary-type plasminogen activator;VCAM-1: vascular cell adhesion molecule 1; VEGF: vascular endothelial growth factor.

Biomarker Released from Timing of elevation post-hypoxic injury*Primary effects on stem cellsCitation SDF-1Astrocytes,endothelial cells Qiu et al., 1988; Peled et al., 1999; Petit et al., 2002; Miller et al.,2005; Cashen et al., 2007; Wang et al., 2008; Rosenkranz et al.,2010; Gong et al., 2011; Yu et al., 2015 SCFNeurons4–24 hours (rodent neuronal cultures)1–7 days (mice & rats)Mobilization,migration,adhesion Mobilization,migration Heissig et al., 2002; Jin et al., 2002a; Sun et al., 2004; Serfozo et al.,2006; Zgheib et al., 2015 MMP-9Vascular endothelial cells, neutrophils,microglia Pruijt et al., 1999; Heissig et al., 2002; Kolev et al., 2003; Avigdor et al., 2004; Shirvaikar et al., 2004; Rosell et al., 2006; Sunagawa et al., 2009; Vagima et al., 2009; Marquez-Curtis et al., 2011;Bednarek et al., 2012 t-PA &u-PA 24 hours (rats)1–6 hours (humans)Mobilization,adhesion≤ 24 hours (rats)MobilizationWang et al., 2000; Heissig et al., 2007; Gong et al., 2011 EPOLiver, kidney,astrocytes Neurons, astrocytes macrophages MigrationMarti et al., 1996; Juul et al., 1999; Li et al., 2017 BDNFNeurons, astrocytes72 hours (humans)MigrationHohn et al., 1990; Benraiss et al., 2001; Chmielnicki et al., 2004;Imam et al., 2009; Douglas-Escobar et al., 2012 VEGFAstrocytes, neuronsBirth (humans) 8 hours–14 days (rats)Birth (humans) 1 hour(mice)Jin et al., 2002b; Sun et al., 2003; Huang et al., 2004; Aly et al.,2009; Sun et al., 2010; Vasiljevic et al., 2011; Chen et al., 2012;Alvarez et al., 2013; Dzietko et al., 2013; Wang et al., 2015;Ramirez-Rodriguez et al., 2017 TNFαMicroglia, astrocytes,neurons Migration,proliferation MigrationSza fl arski et al., 1995; Silverstein et al., 1997; Oygur et al., 1998;Silveira and Procianoy, 2003; Aly et al., 2006; Belmadani et al.,2006; Fu et al., 2009; Liu and Feng, 2010; Xiao et al., 2012 MCP-1Astrocytes, microglia 2.5–48 hours (rats)MigrationIvacko et al., 1997; Belmadani et al., 2006; Jiang et al., 2008 MIP-1αAstrocytes, neurons4 hours–5 days (rats)MigrationCowell et al., 2002; Jiang et al., 2008; Pimentel-Coelho et al., 2012 IL-1βAstrocytes,macrophages 24 hours (humans)6–48 hours (human CSF)MigrationOrzylowska et al., 1999; Lau and Yu, 2001; Guo et al., 2004; Aly et al., 2006; Jiang et al., 2008; Liu and Feng, 2010; Sullivan et al.,2014 ICAM-1Vascular endothelial cells 24 hours(human serum and CSF)3–7 days (humans)AdhesionPeled et al., 1999; Huseynova et al., 2012, 2014 VCAM-1Vascular endothelial cells ProliferationSaliba and Henrot, 2001; Chiesa et al., 2003; Aly et al., 2006;Covey et al., 2011; Pimentel-Coelho et al., 2012; Lv et al., 2015;Orrock et al., 2016 AdhesionPapayannopoulou and Craddock, 1997; Vermeulen et al., 1998;Ruster et al., 2006; Ip et al., 2007; Steingen et al., 2008; Huseynova et al., 2012; Li et al., 2015 IL-6Microglia, astrocytes,neurons 1–7 days (humans)48 hours (rats)Birth–72 hours (humans)24 hours (human CSF)

Plasmin is a serine protease that acts in fi brinolysis, extracellular matrix degradation, and activation of MMPs, and may further brain damage in ischemic injury via extracellular proteolysis and cell degradation (Oranskii et al., 1977;Junge et al., 2003). Tissue-type and urinary-type PAs (t-PA& u-PA) are elevated after injury in a rat model of neonatal cerebral hypoxia (Adhami et al., 2008). This may be unique to neonatal hypoxic injury, as the same study demonstrated that elevations of t-PA and u-PA concentrations were not observed in adult rats after the same injury. Although PA concentrations increase in the fi rst four hours after injury,corresponding to the increased fi brin deposition seen, t-PA and u-PA levels remain elevated well beyond that period, up to 24 hours after injury. Due to expression of t-PA and u-PA by macrophages and astrocytes outside of the vasculature,it has been suggested that the prolonged period of elevation may result in plasmin-mediated cytotoxicity. Supporting this theory, investigators have demonstrated that plasmin inactivation by alpha-2-antiplasmin within two hours of hypoxic-ischemic (HI) injury leads to a dose-dependent decrease in brain injury after 1 week, and t-PA knockout mice have a 69% increase in short term mortality after HI injury.Additionally, increase HIE-associated brain damage was seen with ventricular injection of t-PA (Adhami et al., 2008).Similar to MMP-9, plasmin’s pro-injury roles in HIE may be balanced by its involvement in stem cell mobilization.Plasmin is closely related to MMP-9, as activation of plasminogen and the fibrinolytic cascade promotes MMP-mediated release of SCF (Heissig et al., 2007). Granulocyte colony-stimulating factor (G-CSF) is also involved in this pathway, as G-CSF induces HSC mobilization via direct plasminogen-mediated downregulation of SDF-1 and an MMP-9 dependent upregulation of SCF (Heissig et al.,2002; Gong et al., 2011). Further, a plasminogen antagonist,aprotinin, inhibited G-CSF induced HSC mobilization and MMP-9 activation (Gong et al., 2011) (Figure 2).

SDF-1 or CXCL12

SDF-1 is a member of the CXC chemokine family that is primarily released by bone marrow stromal cells and is a key component in stem cell mobilization (Matthys et al., 2001).After hypoxic-ischemic injury, hypoxia-inducible factor-1(HIF-1) increases the gene expression of SDF-1 (Ceradini et al., 2004). SDF-1 release by the astrocytes and endothelial cells is proportionate to the level of hypoxia and is elevated one to seven days following HIE. The time to peak con-centration appears to vary depending on the model used,between about three days after injury in neonatal mice and seven days after injury in neonatal rats (Miller et al., 2005;Yu et al., 2015).

一是在选择选择精准农业应用系统时应选择大品牌、大企业、服务质量好的企业产品。二是举办专业的精准农业应用系统培训班,对农机操作人员和服务人员进行定期和不定期的理论和实践培训[1]。

Figure 1 Receptor-ligand binding to maintain hematopoietic stem cells (HSC) quiescent in the bone marrow niche.

(A) HSCs are maintained in the bone marrow niche through molecular interactions including CXCR4 binding to SDF-1 and c-kit binding SCF. AMD3100 is a CXCR4 antagonist which induces stem cell mobilization. (B) When these interactions are disrupted, SDF-1 and SCF are released and HSCs are mobilized. CXCR4: CXC chemokine receptor 4; HSC: hematopoietic stem cell; SCF: stem cell factor; SDF-1: stromal cell-derived factor 1.

Human BM endothelium constitutively expresses SDF-1 which binds to the CXCR4 receptor on the cell surface of CD34+ stem cells (Figure 1) (Peled et al., 1999). A reduction in the SDF-1 concentration within the bone marrow niche results in mobilization of stem cells despite stimulating upregulation of the BM CXCR4 receptors (Petit et al., 2002).Although both of the previous studies assessed HSCs, several studies have also investigated the role of SDF-1 in MSCs.CXCR4 expression has been shown to be upregulated in MSCs pretreated with SDF-1 as well as after hypoxic conditions (Yu et al., 2015). Additionally, inhibition of CXCR4 increased the number of circulating MSCs in a mouse model of bone injury (Kumar and Ponnazhagan, 2012).

SCF

SCF (a.k.a. steel factor, kit ligand, mast cell growth factor)is a hemopoietic cytokine that binds to the tyrosine kinase receptor c-kit (Qiu et al., 1988). SCF in the bone marrow niche binds to c-kit on stem cells to maintain their quiescent state, and when SCF is solubilized and the bond is disrupted,stem cells are mobilized. SCF and c-kit expression are elevated throughout development and adulthood in neural cells,hematopoietic stem cells, and germ cells (Matsui et al., 1990;Motro et al., 1991; Manova et al., 1992), and SCF is involved in the regulation of the activity of astrocytes, oligodendroglia,and microglia (Ida et al., 1993; Zhang and Fedoroff, 1997,1999). SCF expression is upregulated in central nervous system injury and in fl ammation, and has been found to be increased 4 to 24 hours after hypoxic injury in rodent forebrain neuronal cultures (Jin et al., 2002a). One of the factors leading to the release of soluble SCF is matrix metalloproteinase(MMP)-9, which induces HSC mobilization and hematopoietic recovery through its effects on SCF (Heissig et al., 2002).

Figure 2 Stem cell mobilization.

†G-SCF causes stem cell mobilization through direct plasminogen-mediated downregulation of SDF-1 and MMP-9 dependent release of SCF.‡Tissue and urinary plasminogen activators promote MMP-9 mediated release of SCF and decrease SDF-1 levels in the bone marrow. ◊SDF-1 and VEGF stimulate release of pro-MMP-9 which results in increased SCF and stem cell mobilization. ★Aprotinin is a plasminogen inhibitor which consequently inhibits MMP-9 activation and stem cell mobilization. G-CSF: Granulocyte colony-stimulating factor; MMP-9: matrix metalloproteinase 9; SCF: stem cell factor; SDF-1: stromal cell-derived factor 1; t-PA: tissue-type plasminogen activator; u-PA: urinary-type plasminogen activator; VEGF: vascular endothelial growth factor.

MMP-9

Client/Server架构的数据库,可根据Client查询请求返回相应的结果集,避免了在网络上传播无效数据,有效地利用了网络带宽,提高了系统性能。本系统采用了流处理机制(Stream)对数据集对象(ADO)序列化和反序列化,利用AdoDataSet数据集SaveToFile方法将数据集保存到文件中,再通过内存流(Mem⁃oryStream)缓存在内存中,使用ZIP压缩算法在压缩内存流中的数据后返回给Client,Client收到响应数据后进行反序列化即可还原出该数据集对象(ADO)。由于网络上传输的数据是经过压缩的,进一步提高了系统性能。

In human neonates, MMP-9 has been found elevated up to six hours after birth, with higher levels in asphyxiated neonates with neurologic sequela compared to those without neurologic sequela. This significant difference in MMP-9 level resolved by day 1 of life, suggesting that MMP-9 may play an early role in humans (Sunagawa et al., 2009). In the later phases of recovery, MMPs appear to take on more bene fi cial roles. MMP-9 was found to be elevated 7–14 days after stroke in a rat model, and inhibition of MMP in this model caused suppression of neurovascular remodeling, impaired functional recovery, and increased brain injury (Cunningham et al., 2005; Zhao et al., 2006; Yang and Rosenberg, 2015). MMP-9 may have limitations as a biomarker of HIE in premature infants, as plasma MMP-9 levels are signi fi cantly lower in infants born at 25–27 weeks gestation(Bednarek et al., 2012).

MMP-9 also functions in promoting release of extracellular matrix-bound or cell-surface-bound cytokines that activate stem cell mobilization. One of these cytokines is SCF,as MMP knockout mice demonstrate significantly lower levels of SCF than wildtype. Additionally, MMP-9 has been shown to induce HSC mobilization via interleukin (IL)-8 in non-human primates (Pruijt et al., 1999). Conversely,MMP-9 can be upregulated by other factors that are active in stem cell regulation, such as SDF-1 and vascular endothelial growth factor (VEGF), which stimulate release of pro-MMP-9, resulting in increased SCF plasma levels and mobilization of CD34+ stem cells (Heissig et al., 2002).

Figure 3 Stem cell migration.

†HIF-1 induces expression of EPO. EPO cau ses MSC migration through an EPO receptor on MSCs and through increasing levels of VEGF and BDNF. ‡IL-1β released at the injured site in HIE causes secretion of MCP-1 and MIP-1α. ◊TNFα causes MCP-1 dependent migration of neural progenitor cells. *SDF-1, SCF, BDNF, MCP-1, and MIP-1α all induce migration of MSCs. BDNF: Brain-derived neurotrophic factor; EPO: erythropoietin; HIF-1: hypoxia inducible factor 1; IL-1β:interleukin 1β; MCP-1: monocyte chemoattractant protein-1; MIP-1α:macrophage inflammatory protein-1α; MSC: mesenchymal stem cell;SCF: stem cell factor; SDF-1: stromal cell-derived factor 1; TNFα: tumor necrosis factor α; VEGF: vascular endothelial growth factor.

PAs

There are several signaling molecules involved in maintaining stem cells in niches that can be modi fi ed to allow for stem cell mobilization. Most of the research on these signaling molecules has been done in hematopoietic stem cell(HSC) lines, and there remains a paucity of data on mesenchymal stem cell (MSC) niches. Because of this, much of the data presented in this section will represent studies in HSCs,with the likelihood that many of these signaling mechanisms may similarly affect MSCs. Two receptors involved in stem cell mobilization include CXC chemokine receptor 4 (CXCR4) and c-kit. CXCR4 and c-kit are expressed by HSCs and bind to stromal cell-derived factor 1 (SDF-1) and stem cell factor (SCF), respectively, on the BM endothelium(Figure 1). In addition, both MMP-9 and plasminogen activators (PAs) have been found to be elevated after neonatal HIE and are factors involved in the process of stem cell mobilization (Figure 2).

Figure 4 Chemotactic ligands at the site of injured tissues and stem cell receptors involved in migration.

EPO: erythropoietin; MCP-1: monocyte chemoattractant protein-1;MIP-1α: macrophage inflammatory protein-1α; MSC: mesenchymal stem cell; SCF: stem cell factor; SDF-1: stromal cell-derived factor 1.

Stem Cell Migration

Once stem cells are released from the BM niche into peripheral circulation, they must then migrate to the speci fi c areas of the body where they are needed. Figure 3 demonstrates several of the factors that have been found to be elevated after HIE and are involved in stem cell migration, and Figure 4 illustrates the MSC receptors involved in migration and their corresponding ligands.

(3)提高施工标准与审核标准。山区公路桥梁勘察与设计工作必须按照国家相关的法律法规来开展,施工材料的选用与施工的过程方法也要严格按照国家标准行事。如果在施工过程中出现需要使用新工艺、新材料等情况,则需要对相关部门提前进行报备,经其确认后才能投入工程建设中去。同时在工程完成之后进行严格的验收工作,充分保证工程的质量安全。

Erythropoietin (EPO)

As circulating stem cells appears to act, at least in part,through paracrine mechanisms rather than adhesion and direct tissue integration, it is important to also discuss stem cell paracrine effects such as the effects on endogenous cell proliferation. There exists a complex interaction between circulating stem cells and the neural stem cell niche, however, that is beyond the scope of this current review. For more information on the factors involved in altering stem cell survival and differentiation, we would direct the reader to previously published manuscripts which fully review those topics (Decimo et al., 2012; Ruddy and Morshead, 2018).

Given the studies suggesting EPO may have a role in both neuroprotection and MSC migration, several investigators have begun assessing the effects of exogenous EPO administration on neonatal HIE (Parikh and Juul, 2018). Although the doses and intervals have varied, multiple clinical studies have now demonstrated that EPO administered immediately after HI injury, and in combination with therapeutic hypothermia, decreases seizure activity and endogenous nitric oxide production, and results in improved neurodevelopmental outcomes or death at 6-18 months of age (Zhu et al., 2009; Elmahdy et al., 2010). Additionally, EPO combined with therapeutic hypothermia results in lower brain injury on MRI and improved motor scores at 1 year of age over control infants who received hypothermia and placebo(Wu et al., 2016). Additional large multi-center studies are ongoing, with high-dose erythropoietin for asphyxia and encephalopathy (HEAL) hoping to recruit 500 newborns in the United States (Kawahara et al., 1997) and EPO for hypoxic ischaemic encephalopathy in newborns (PAEAN) hoping to recruit 300 newborns in Australia (Plumier et al., 1997).

SDF-1

In addition to stem cell mobilization, SDF-1 also plays a role in migration of stem cells to sites of injury. In a rat model of cerebral hypoxia, SDF-1 was found to be highly expressed at lesion sites in the hippocampus, corpus callosum, and periventricular areas, correlating with the sites of human UCB stem cell migration after intraperitoneal injection. Additionally, administration of an SDF-1 inhibitor resulted in a decrease in the number of UCB cells found in the lesioned areas of the brain (Rosenkranz et al., 2010). Similarly, treatment with a CXCR4 antagonist, AMD3100, also prevents MSC migration to HI-injured brain (Wang et al., 2008; Yu et al., 2015). Because AMD3100 also inhibits interaction of CXCR4 with SDF-1 within the bone marrow, administration results in more stem cells mobilized into peripheral circulation, but those stem cells have decreased migration to speci fi c sites along the SDF-1 gradient (Figure 1) (Cashen et al., 2007). Likely related to its role in stem cell mobilization,intracranial administration of SDF-1 decreased intracranial inflammation, induced cerebral re-myelination, and improved memory and spatial perception in a neonatal mouse model of cerebral hypoxia (Mori et al., 2015).

SCF

In addition to VEGF’s role in angiogenesis during hypoxia, it also increases NPC migration in response to interaction with VEGF receptor 2 (Ramirez-Rodriguez et al.,2017). Although VEGF stimulates migration of stem cells in a dose dependent manner in vitro, in vivo studies have been unable to show this phenomenon with VEGF signaling alone (Schmidt et al., 2005). VEGF also induces migration of MSCs, although the increased migration appears to be associated with decreased MSC differentiation (Wang et al.,2015).

Matrix metalloproteinases are enzymes primarily responsible for degradation of extracellular matrix proteins, but also release ligands, inactivate cytokines, and cleave receptors(Van Lint and Libert, 2007). MMP-9 is a protease produced by vascular endothelial cells, infiltrated neutrophils, and activated microglia in ischemic brain (Kolev et al., 2003;Rosell et al., 2006). Levels of MMP-9 are elevated in HIE,with increased levels corresponding to increased severity of HIE (Sunagawa et al., 2009; Bednarek et al., 2012). Similar to SDF-1, MMP-9 levels appear to vary by species, as they can be elevated in adult mice as early as 4 hours after the hypoxic event (Gasche et al., 1999), but in a neonatal rat model,the levels did not increase until 24 hours post-hypoxic injury(Adhami et al., 2008).

BDNF

BDNF is secreted by neurons and astrocytes in the central nervous system and plays an important role in neuronal maintenance and survival (Hohn et al., 1990). Mice born without the tyrosine kinase receptor for BDNF do not feed normally and display neuronal deficiencies in their facial motor nucleus, spinal cord, and peripheral nervous system(Klein et al., 1993). BDNF levels from cord blood and serum samples taken in the fi rst few days of life are higher in full term infants versus premature infants, re fl ecting advancing maturity of the nervous and immune system (Malamitsi-Puchner et al., 2004). BDNF may also play a role in brain injury, as it is found at increased levels in the serum after HIE. Elevated BDNF levels at 72 hours of life in term newborns with HIE is a poor prognostic factor and suggests severe brain injury (Imam et al., 2009).

皮雕工艺的传播发展,需要新的设计理念,要对传统装饰艺术进行归纳总结,以便符合现代人的审美需求。工具的开发也给皮雕工艺的创新带来更多可能性。从事皮雕工艺创作,从动物、人物、场景等题材的拓展,到纹样的创新,留给皮雕工艺手作人的创意空间越来越广,皮雕工艺的发展必然是多元化的。

BNDF, as well as other neurotrophins, has been shown to increase migration of multipotent astrocytic stem cells in vitro and neuronal cells in vivo (Benraiss et al., 2001; Chmielnicki et al., 2004; Douglas-Escobar et al., 2012). It stimulates migration of stem cell progenitors in the cerebellum,and while BNDF knockout granule cells lack the ability to migrate appropriately, administration of exogenous BDNF restores their ability to migrate out of the external granule cell layer (Borghesani et al., 2002). Additionally, BNDF has been shown to be neuroprotective in a neonatal rat model of hypoxic-ischemic injury, likely via activation of the extracellular signal-related kinase (ERK) pathways (Han and Holtzman, 2000). A single intraventricular injection of BDNF decreased hypoxia-ischemia induced brain tissue loss in a 7-day old rat model by 90% when injected prior to injury and by 50% when injected after the insult. Less protection was seen after injection in 21 day old rats, indicating more robust BDNF effects in the more immature brain (Cheng et al., 1997).

花生通常是经加工后再被食用,通过加工实现蛋白低致敏或脱敏,也是食物过敏研究的重要方向之一[14]。Beyer K[15]等认为,中国花生过敏人口较少的原因主要是我国常采用水煮和油炸两种方式来加工花生,已有研究认为热加工主要是由于温度的不同对花生蛋白产生影响,除了加工引起的蛋白质结构变化可以直接影响致敏性之外,聚集性和消化吸收性等其它性质的变化也会间接影响致敏性[16]。

VEGF

VEGF plays a role in regulating both vasculogenesis and angiogenesis and is vital for brain development (Millauer et al., 1993). VEGF mediates neuronal growth and maturation in the CNS through binding the VEGFR2 receptor (Khaibullina et al., 2004; Jin et al., 2006), and is a key component in exercise-induced hippocampal neurogenesis (Fabel et al.,2003). VEGF is secreted by astrocytes and neurons (Alvarez et al., 2013), and has been found to be elevated in both the CSF and serum in human neonates with HIE (Aly et al.,2009; Vasiljevic et al., 2011). VEGF is elevated immediately following HI injury in human neonates (Aly et al., 2009) and remains elevated for up 14 days in a neonatal rat model of HI brain injury (Huang et al., 2004; Chen et al., 2012). Studies have shown that CSF levels of VEGF in human neonates correlate with severity of HIE, however correlation of serum levels with severity have demonstrated mixed results (Ergenekon et al., 2004; Aly et al., 2009).

从文化情境来看,目前有关中国文化情境下康复性景观的研究还很少。Huang和Xu(2018)分析了养生旅游目的地广西巴马瑶族自治县的游客,发现符号景观对于游客获得健康体验有决定性的作用。Wang、Cui和Xu(2018)基于关系的视角,运用康复性景观理论揭示了游客在新疆吐鲁番沙漠环境中通过对具身体验的文化解读获得治愈的体验。

Much like SDF-1, SCF also plays dual roles in both stem cell mobilization and migration. Although SCF’s role in stem cell migration has been demonstrated in neural progenitor cells (NPC) in several other types of injury, there is a paucity research assessing this factor’s effects on MSCs and after hypoxic brain injury. In a mouse model of hypothermic injury,Sun et al demonstrated upregulated gene and protein levels of SCF in injured areas of the brain, which induced NPC migration (Sun et al., 2004). Additionally, recombinant SCF induced migration of mouse embryonic stem cells after neural induction (Serfozo et al., 2006). In non-neural tissues,SCF is also activated in injury and has been shown to enhance diabetic wound healing in mice through skin stem cell migration and increases in expression of HIF-1 and VEGF(Zgheib et al., 2015).

Tumor necrosis factor α (TNFα)

TNFα is a pro-in fl ammatory cytokine with roles in cell signaling during acute in fl ammatory states (Old, 1985; Havell,1987), but also affects sleep regulation and embryonic development (Wride and Sanders, 1995; Krueger et al., 1998).In brain injury, TNFα is released from microglia, astrocytes,and neurons (Silverstein et al., 1997) and is involved in neuronal apoptosis and disruption of the blood brain barrier.Intracranial TNFα levels have been found elevated 4–24 hours post HI injury in neonatal rats (Sza fl arski et al., 1995),and CSF levels have been found to peak at 6 hours post HI injury in human neonates (Oygur et al., 1998; Silveira and Procianoy, 2003). Serum TNFα levels in humans can be significantly elevated within 24 hours of injury (Oygur et al.,1998; Silveira and Procianoy, 2003; Aly et al., 2006; Liu and Feng, 2010).

High levels of TNFα are likely neurotoxic; however, some studies suggest TNFα at lower levels may have neuroprotective effects. Despite studies demonstrating that elevated TNFα levels worsen neuropathological outcomes after nitric oxide-induced neurotoxicity (Blais and Rivest, 2004), mice completely lacking TNFα activity demonstrate increased neuronal injury after nitric oxide-induced brain damage,as well as decreased early microglial activation (Turrin and Rivest, 2006). Additionally, exposure of neonatal mouse subventricular zone stem cells to low levels of TNFα (1 ng/mL)causes neuronal differentiation and stem cell proliferation,while higher levels of TNFα (10–100 ng/mL) causes apoptosis of the stem cells (Bernardino et al., 2008). The increased migration of neural progenitor cells induced by TNFα appears to act through several mediators. One study showed that migration is inhibited in monocyte chemoattractant protein-1 (MCP-1) knockout mice (Belmadani et al., 2006),while others showed migration through upregulation of intracellular adhesion molecule (ICAM)-1 (Fu et al., 2009) and vascular cell adhesion protein (VCAM)-1 expression (Xiao et al., 2012).

MCP-1/macrophage in fl ammatory protein-1α (MIP-1α)

MCP-1, also called CCL-2, was the fi rst member of the C-C chemokine family discovered in humans, and its primary action is to induce chemotaxis of monocytes, memory T cells, and natural killer cells (Van Coillie et al., 1999; Deshmane et al., 2009). MIP-1α, also called CCL3, is also a C-C chemokine which plays a role in the chemotaxis of neutrophils, macrophages and T cells (Taub et al., 1993; Koch et al., 1994). MCP-1 and MIP-1α both appear to have similar chemotactic effects in stem cell migration, promoted via MCP-1 binding to receptor CCR2 and MIP-1α binding to CCR1 and CCR5 on the surface of stem cells (Belmadani et al., 2006; Jiang et al., 2008). MCP-1 levels are increased 2.5 to 48 hours after HI injury (Ivacko et al., 1997), while MIP-1α is released by astrocytes and neurons with levels elevated 4 hours to 5 days after HI injury (Cowell et al., 2002; Pimentel-Coelho et al., 2012).

IL-1β

IL-1β is a member of the interleukin 1 cytokine family produced by activated macrophages. It is an in fl ammatory mediator involved in cellular apoptosis, differentiation, and proliferation (No authors listed, 2018). IL-1β is also highly expressed in prenatal and postnatal brain development and guides radial migration of neural progenitors in rat neocortex development (Dziegielewska et al., 2000; Ma et al.,2014). Astrocytes release IL-1β during the acute phase of HIE (Orzylowska et al., 1999; Lau and Yu, 2001), and levels are elevated in both the CSF and serum of human neonates within 24 hours post HI-injury (Aly et al., 2006). Elevated levels predict poor prognosis in HIE, as up to 88% of patients with elevated IL-1β post HIE have abnormal neurologic outcomes at 6 months of age (Aly et al., 2006; Liu and Feng, 2010).

过程误差分析分为过程控制变量和过程变量的误差分析。多工序制造过程中关键过程控制变量作为制造系统的作用因素被称为误差源,过程变量作为被作用因素被称为零件误差。当多工序制造过程被抽象为具体数学模型后,误差源为该数学模型的自变量因子,零件误差为因变量因子。

When expression of IL-1β is inhibited, MIP-1α expression is also inhibited, and it has been hypothesized that IL-1β release at the site injured in HIE may initiate astrocytes,microglia, and neurons to secrete MCP-1 and MIP-1α (Guo et al., 2004; Jiang et al., 2008). Whether through the activation of MCP-1 and MIP-1α, or other pathways, it has been shown that pre-stimulation with IL-1β increases migration of murine MSCs in a dose-dependent manner (Sullivan et al., 2014).

Stem Cell Adhesion

Once stem cells migrate to target tissues, molecules involved with cell adhesion allow the stem cells to adhere to other cells or the extracellular matrix surrounding them. The main HIE biomarkers that have roles in stem cell adhesion are ICAM-1, VCAM-1, and MMPs.

目前的研究多着重于企业合作基础的影响因素方面,而对合作过程中影响因素的研究相对缺乏。深入挖掘对企业合作过程也有重要影响作用的潜在因素,如政策法律环境这一宏观变量,将法律法规与现有研究中提出的政府行为因子相结合,有助于加强企业在合作过程中对法律法规的重视与合理利用。因此本文对更多内外部影响因素的综合考察,在一定程度上实现了对合作影响因素理论研究的完善与丰富,可帮助企业意识到这些因素在合作中的重要作用及交互作用关系,为更好的开展协调联动工作、构建科学有效的协调联动机制提供参考与依据。

ICAM-1 & VCAM-1

ICAMs and VCAMs are expressed on vascular endothelial cells and have roles in leukocyte attachment and trans-endothelial migration at inflammatory sites through binding to integrins (Aricescu and Jones, 2007). ICAM-1 is secreted by vascular endothelial cells in HIE. Levels are signi fi cantly elevated on days 3–7 post-insult in human neonates with HIE, and increasing levels correlate with worse prognosis(Huseynova et al., 2012; Huseynova et al., 2014). ICAM-1 expression is also increased after exposure to various in fl ammatory cytokines, including IL-1β, TNFα, interferon-gamma (IFN-γ), and endotoxin (Lv et al., 2015). VCAM-1 is also elevated in the fi rst week following HIE in human neonates,versus elevations within the fi rst 48 hours in the rat model of neonatal HIE (Huseynova et al., 2012; Li et al., 2015).

VCAM-1 and ICAM-1 both act as ligands for the β-1 integrin very late activating-antigen-4 (VLA-4) and the β-2 integrin leukocyte function antigen-1 (LFA-1) expressed by MSCs and HSCs. Their roles have been elucidated in studies showing that antibodies to VCAM-1 and VLA-4 can prevent stem cell homing (Papayannopoulou and Craddock, 1997;Vermeulen et al., 1998). MSCs have been demonstrated to adhere to endothelial cells via binding and rolling mediated through P-selectin followed by VLA-4/VCAM-1 dependent trans-endothelial migration. Adhesion of MSCs to endothelial cells is significantly diminished when a VLA-4 or VCAM-1 inhibitor is used (Ruster et al., 2006; Steingen et al., 2008).

MMPs

In addition to MMP-9’s role in stem cell mobilization, it also functions in stem cell adhesion. MMPs degrade the extracellular matrix and allow stem cells to migrate through basement membrane barriers. When secretion of MMP-9 and MMP-2 is induced by SDF-1 and other growth factors,HSCs migration through basement membrane barriers along the SDF-1 gradient is enhanced (Marquez-Curtis et al., 2011). Membrane type 1 MMP (MT1-MMP) is expressed by bone marrow mononuclear cells and circulating human CD34+ stem cells and can be activated by SDF-1 and G-CSF (Janowska-Wieczorek et al., 2000; Vagima et al.,2009). MT1-MMP has been demonstrated to be important in both SDF-1-induced and SCF/c-kit-induced migration,mobilization, and homing of stem cells (Avigdor et al., 2004;Shirvaikar et al., 2004; Vagima et al., 2009).

Stem Cell Proliferation

EPO is best known for its role in the regulation of erythropoiesis (Erslev, 1953); however, it also has protective and reparative abilities in the heart (Teixeira et al., 2012), spinal cord (Xiong et al., 2011), bone (Chen et al., 2014), kidneys(Liu et al., 2013), and brain (Khairallah et al., 2016). EPO has been found to upregulate VEGF and angiogenesis, serve as a direct angiogenic factor, inhibit apoptosis, increase differentiation of MSCs, and mobilize endothelial progenitor cells (Ribatti et al., 1999; Heeschen et al., 2003; Liu et al.,2013; Chen et al., 2014). EPO is primarily secreted by the kidney, but can also be released by astrocytes, and is elevated in neonatal HIE. The highest plasma and CSF levels of EPO are found right after hypoxic injury (Marti et al., 1996; Juul et al., 1999), and its release is in part due to HIF-1-induced upregulation (Prass et al., 2003). EPO may also induce MSC migration and facilitate angiogenesis via the EPO receptor expressed on human MSCs (Zwezdaryk et al., 2007) (Figure 4). In a rat model of spinal cord injury, EPO was demonstrated to increase migration of transplanted bone marrow MSCs to site of injury, increase brain-derived neurotrophic factor (BDNF) and VEGF expression, decrease neuronal apoptosis at the injury site, and hasten clinical neurologic recovery at 28 days post injury compared to both sham treatment and treatment with stem cells alone (Li et al.,2017).

IL-6

IL-6 is another pro-inflammatory mediator that at high levels can also cause inflammation, enhance vascular permeability, and increase secondary cerebral edema (Lv et al.,2015). IL-6 is released from microglia, astrocytes, and neurons (Saliba and Henrot, 2001) in response to brain injury,and levels are signi fi cantly higher in the cord blood and CSF of neonates with HIE, with increasing plasma IL-6 levels associated with increasing HIE severity (Chiesa et al., 2003;Aly et al., 2006; Orrock et al., 2016). Serum samples of IL-6 were elevated at 24 and 72 hours in infants with HIE and were correlated with adverse outcome (Orrock et al., 2016).IL-6 levels are also elevated in human neonates with HIE in serum and CSF samples drawn within 24 hours of birth.,and increased CSF IL-6 levels correlated with worse neurologic outcomes at 6 months of age and increased mortality rate (Aly et al., 2006).

根据上述反应方程式得到铝作还原剂,NaOH和H2O全作氧化剂,所以氧化剂和还原剂的物质的量之比是2∶(2+2)=1∶2。

例2:公司贯彻“以信息、安防、节能环保等三大科技产业为主业,以金融投资和科技园建设为两翼,主业突出,两翼齐飞”的“一主两翼”发展方针。

Like many other pro-inflammatory cytokines, IL-6 also functions paradoxically as a neuroprotective mediator by inhibiting the production of TNFα and IL-1β and through increasing neural growth factor release (Lv et al., 2015)and promoting proliferation of neural stem progenitor cells (Covey et al., 2011; Pimentel-Coelho et al., 2012). In a neonatal rat model of HI-injury, addition of IL-6 enhanced neuronal growth and self-renewal NPCs, and NPC numbers were reduced with addition of IL-6 inhibitor (Covey et al.,2011).

VEGF

In addition to VEGF’s role in stem cell migration, it also induces stem cell proliferation. VEGF functions to protect the cerebral cortex from hypoxic injury and promote survival, repair, and regeneration of neurons in the cerebral cortex when hypoxic injury does occur. VEGF also causes proliferation and angiogenesis of vascular endothelial cells and enhances the proliferation of NPCs (Jin et al., 2002b;Sun et al., 2010; Dzietko et al., 2013). Intracerebral injection of VEGF led to increased endothelial proliferation, increase in total vessel volume, and reduced brain injury following neonatal ischemic stroke in a neonatal rat model (Dzietko et al., 2013). In a similar animal model, rats transplanted with VEGF-transfected NPCs post HI-injury had improved learning and memory abilities at 30 days (Yao et al., 2016).

Conclusion

Stem cell transplant has shown promise as a therapy for neonatal HIE, but further research is needed to prove its safety and efficacy. One of the ways that efficacy of stem cell transplantation may be improved in the future is through the alteration of the stem cells and upregulation of certain factors to optimize migration, adhesion, and differentiation. It will be important for future investigations of MSC manipulation to assess not only the effects on the injured tissue, but also the effects on the upregulated factors on healthy tissue (trapping of cells in the microcirculation of the lung, effects on the immune system, etc.). By examining the factors that are naturally elevated after neonatal HI brain injury, we hope to better understand the interactions between the injured tissue and the endogenous stem cells. Understanding the interactions of these biomarkers with stem cells opens the door to future research into manipulating these biomarkers to advance the success of stem cell treatments in neonates with HIE.

Author contributions: Drs. Parry and Peeples both conceptualized the review, drafted the initial manuscript, and provided critical revision of the fi nal manuscript.

Conflicts of interest: We have no financial ties to any products described in the study and have no con fl icts of interest to declare.

Copyright license agreement: The Copyright License Agreement has been signed by all authors before publication.

Financial support: None.

Plagiarism check: Checked twice by iThenticate.

Peer review: Externally peer reviewed.

Open access statement: This is an open access journal, and articles are distributed under the terms of the Creative Commons Attribution-Non-Commercial-ShareAlike 4.0 License, which allows others to remix, tweak,and build upon the work non-commercially, as long as appropriate credit is given and the new creations are licensed under the identical terms.

Open peer reviewer: Yuan Wang, Sichuan University, China.

Additional fi le: Open peer review report 1.

实行“1+3”专业人才培养模式改革,即第一学年全面学习各个舞种的基本功,按照专业发展要求实行拉丁舞专业理论核心课程,以加强对专业教育的引导和衔接。第二学年至第四学年全面实行专业技能提升教育。培养专业能力拔尖的学生进入校队,并聘请专业能力较强的教师在课余时间授课,给学生参与竞赛和表演创造条件,以培养学生实践能力。

References

Adhami F, Yu D, Yin W, Schloemer A, Burns KA, Liao G, Degen JL,Chen J, Kuan CY (2008) Deleterious effects of plasminogen activators in neonatal cerebral hypoxia-ischemia. Am J Pathol 172:1704-1716.

Alvarez JI, Katayama T, Prat A (2013) Glial influence on the blood brain barrier. Glia 61:1939-1958.

Aly H, Khashaba MT, El-Ayouty M, El-Sayed O, Hasanein BM (2006)IL-1beta, IL-6 and TNF-alpha and outcomes of neonatal hypoxic ischemic encephalopathy. Brain Dev 28:178-182.

Aly H, Hassanein S, Nada A, Mohamed MH, Atef SH, Atiea W (2009)Vascular endothelial growth factor in neonates with perinatal asphyxia. Brain Dev 31:600-604.

Aricescu AR, Jones EY (2007) Immunoglobulin superfamily cell adhesion molecules: zippers and signals. Curr Opin Cell Biol 19:543-550.

Avigdor A, Schwartz S, Goichberg P, Petit I, Hardan I, Resnick I, Slavin S,Nagler A, Lapidot T (2004) Membrane type 1-matrix metalloproteinase is directly involved in G-CSF induced human hematopoietic stem and progenitor cell mobilization. Blood 104:2675-2675.

Ballen K (2017) Umbilical cord blood transplantation: challenges and future directions. Stem Cells Transl Med 6:1312-1315.

Bednarek N, Svedin P, Garnotel R, Favrais G, Loron G, Schwendiman L, Hagberg H, Morville P, Mallard C, Gressens P (2012) Increased MMP-9 and TIMP-1 in mouse neonatal brain and plasma and in human neonatal plasma after hypoxia-ischemia: a potential marker of neonatal encephalopathy. Pediatr Res 71:63-70.

Belmadani A, Tran PB, Ren D, Miller RJ (2006) Chemokines regulate the migration of neural progenitors to sites of neuroin fl ammation. J Neurosci 26:3182-3191.

Benraiss A, Chmielnicki E, Lerner K, Roh D, Goldman SA (2001) Adenoviral brain-derived neurotrophic factor induces both neostriatal and olfactory neuronal recruitment from endogenous progenitor cells in the adult forebrain. J Neurosci 21:6718-6731.

Bernardino L, Agasse F, Silva B, Ferreira R, Grade S, Malva JO (2008)Tumor necrosis factor-alpha modulates survival, proliferation, and neuronal differentiation in neonatal subventricular zone cell cultures. Stem Cells 26:2361-2371.

Blais V, Rivest S (2004) Effects of TNF-alpha and IFN-gamma on nitric oxide-induced neurotoxicity in the mouse brain. J Immunol 172:7043-7052.

Borghesani PR, Peyrin JM, Klein R, Rubin J, Carter AR, Schwartz PM,Luster A, Corfas G, Segal RA (2002) BDNF stimulates migration of cerebellar granule cells. Development 129:1435-1442.

Cashen AF, Nervi B, DiPersio J (2007) AMD3100: CXCR4 antagonist and rapid stem cell-mobilizing agent. Future Oncol 3:19-27.

Ceradini DJ, Kulkarni AR, Callaghan MJ, Tepper OM, Bastidas N,Kleinman ME, Capla JM, Galiano RD, Levine JP, Gurtner GC (2004)Progenitor cell trafficking is regulated by hypoxic gradients through HIF-1 induction of SDF-1. Nat Med 10:858-864.

Chen H, Xiong T, Qu Y, Zhao F, Ferriero D, Mu D (2012) mTOR activates hypoxia-inducible factor-1alpha and inhibits neuronal apoptosis in the developing rat brain during the early phase after hypoxia-ischemia. Neurosci Lett 507:118-123.

Chen S, Li J, Peng H, Zhou J, Fang H (2014) Administration of erythropoietin exerts protective effects against glucocorticoid-induced osteonecrosis of the femoral head in rats. Int J Mol Med 33:840-848.

Cheng Y, Gidday JM, Yan Q, Shah AR, Holtzman DM (1997) Marked age-dependent neuroprotection by brain-derived neurotrophic factor against neonatal hypoxic-ischemic brain injury. Ann Neurol 41:521-529.

Chiesa C, Pellegrini G, Panero A, De Luca T, Assumma M, Signore F,Pacifico L (2003) Umbilical cord interleukin-6 levels are elevated in term neonates with perinatal asphyxia. Eur J Clin Invest 33:352-358.

Chmielnicki E, Benraiss A, Economides AN, Goldman SA (2004) Adenovirally expressed noggin and brain-derived neurotrophic factor cooperate to induce new medium spiny neurons from resident progenitor cells in the adult striatal ventricular zone. J Neurosci 24:2133-2142.

Cotten CM, Murtha AP, Goldberg RN, Grotegut CA, Smith PB, Goldstein RF, Fisher KA, Gustafson KE, Waters-Pick B, Swamy GK,Rattray B, Tan S, Kurtzberg J (2014) Feasibility of autologous cord blood cells for infants with hypoxic-ischemic encephalopathy. J Pediatr 164:973-979.e1.

Covey MV, Loporchio D, Buono KD, Levison SW (2011) Opposite effect of in fl ammation on subventricular zone versus hippocampal precursors in brain injury. Ann Neurol 70:616-626.

Cowell RM, Xu H, Galasso JM, Silverstein FS (2002) Hypoxic-ischemic injury induces macrophage inflammatory protein-1alpha expression in immature rat brain. Stroke 33:795-801.

Cui LL, Nitzsche F, Pryazhnikov E, Tibeykina M, Tolppanen L, Rytkönen J, Huhtala T, Mu JW, Khiroug L, Boltze J, Jolkkonen J (2017)Integrin α4 overexpression on rat mesenchymal stem cells enhances transmigration and reduces cerebral embolism after intracarotid injection. Stroke 48:2895-2900.

Cunningham LA, Wetzel M, Rosenberg GA (2005) Multiple roles for MMPs and TIMPs in cerebral ischemia. Glia 50:329-339.

Decimo I, Bifari F, Krampera M, Fumagalli G (2012) Neural stem cell niches in health and diseases. Curr Pharm Des 18:1755-1783.

Deng J, Zou ZM, Zhou TL, Su YP, Ai GP, Wang JP, Xu H, Dong SW(2011) Bone marrow mesenchymal stem cells can be mobilized into peripheral blood by G-CSF in vivo and integrate into traumatically injured cerebral tissue. Neurol Sci 32:641-651.

Deshmane SL, Kremlev S, Amini S, Sawaya BE (2009) Monocyte chemoattractant protein-1 (MCP-1): an overview. J Interferon Cytokine Res 29:313-326.

Douglas-Escobar M, Weiss MD (2015) Hypoxic-ischemic encephalopathy: a review for the clinician. JAMA Pediatr 169:397-403.

Douglas-Escobar M, Rossignol C, Steindler D, Zheng T, Weiss MD(2012) Neurotrophin-induced migration and neuronal differentiation of multipotent astrocytic stem cells in vitro. PLoS One 7:e51706.

Dziegielewska KM, Moller JE, Potter AM, Ek J, Lane MA, Saunders NR (2000) Acute-phase cytokines IL-1beta and TNF-alpha in brain development. Cell Tissue Res 299:335-345.

Dzietko M, Derugin N, Wendland MF, Vexler ZS, Ferriero DM (2013)Delayed VEGF treatment enhances angiogenesis and recovery after neonatal focal rodent stroke. Transl Stroke Res 4:189-200.

Elmahdy H, El-Mashad AR, El-Bahrawy H, El-Gohary T, El-Barbary A, Aly H (2010) Human recombinant erythropoietin in asphyxia neonatorum: pilot trial. Pediatrics 125:e1135-1142.

Ergenekon E, Gucuyener K, Erbas D, Aral S, Koc E, Atalay Y (2004)Cerebrospinal fl uid and serum vascular endothelial growth factor and nitric oxide levels in newborns with hypoxic ischemic encephalopathy. Brain Dev 26:283-286.

Erslev A (1953) Humoral regulation of red cell production. Blood 8:349-357.

Fabel K, Fabel K, Tam B, Kaufer D, Baiker A, Simmons N, Kuo CJ,Palmer TD (2003) VEGF is necessary for exercise-induced adult hippocampal neurogenesis. Eur J Neurosci 18:2803-2812.

Fu X, Han B, Cai S, Lei Y, Sun T, Sheng Z (2009) Migration of bone marrow-derived mesenchymal stem cells induced by tumor necrosis factor-alpha and its possible role in wound healing. Wound Repair Regen 17:185-191.

Gasche Y, Fujimura M, Morita-Fujimura Y, Copin JC, Kawase M,Massengale J, Chan PH (1999) Early appearance of activated matrix metalloproteinase-9 after focal cerebral ischemia in mice: a possible role in blood-brain barrier dysfunction. J Cereb Blood Flow Metab 19:1020-1028.

Gong Y, Fan Y, Hoover-Plow J (2011) Plasminogen regulates stromal cell-derived factor-1/CXCR4-mediated hematopoietic stem cell mobilization by activation of matrix metalloproteinase-9. ArteriosclerThromb Vasc Biol 31:2035-2043.

Guo CJ, Douglas SD, Gao Z, Wolf BA, Grinspan J, Lai JP, Riedel E, Ho WZ (2004) Interleukin-1beta upregulates functional expression of neurokinin-1 receptor (NK-1R) via NF-kappaB in astrocytes. Glia 48:259-266.

Han BH, Holtzman DM (2000) BDNF protects the neonatal brain from hypoxic-ischemic injury in vivo via the ERK pathway. J Neurosci 20:5775-5781.

Havell EA (1987) Production of tumor necrosis factor during murine listeriosis. J Immunol 139:4225-4231.

Heeschen C, Aicher A, Lehmann R, Fichtlscherer S, Vasa M, Urbich C,Mildner-Rihm C, Martin H, Zeiher AM, Dimmeler S (2003) Erythropoietin is a potent physiologic stimulus for endothelial progenitor cell mobilization. Blood 102:1340-1346.

Heissig B, Hattori K, Dias S, Friedrich M, Ferris B, Hackett NR, Crystal RG, Besmer P, Lyden D, Moore MA, Werb Z, Ra fi i S (2002) Recruitment of stem and progenitor cells from the bone marrow niche requires MMP-9 mediated release of kit-ligand. Cell 109:625-637.

Heissig B, Lund LR, Akiyama H, Ohki M, Morita Y, Romer J, Nakauchi H, Okumura K, Ogawa H, Werb Z, Dano K, Hattori K (2007)The plasminogen fi brinolytic pathway is required for hematopoietic regeneration. Cell Stem Cell 1:658-670.

Hohn A, Leibrock J, Bailey K, Barde YA (1990) Identification and characterization of a novel member of the nerve growth factor/brain-derived neurotrophic factor family. Nature 344:339-341.

Huang YF, Zhuang SQ, Chen DP, Liang YJ, Li XY (2004) Angiogenesis and its regulatory factors in brain tissue of neonatal rat hypoxic-ischemic encephalopathy. Zhonghua Er Ke Za Zhi 42:210-214.

Huseynova S, Panakhova N, Orujova P (2012) Endothelial dysfunction and cellular adhesion molecule activation in preterm infants with hypoxic ischemic encephalopathy. Am Int J Contemp Res 2:47-52.

Huseynova S, Panakhova N, Orujova P, Hasanov S, Guliyev M, Orujov A (2014) Elevated levels of serum sICAM-1 in asphyxiated low birth weight newborns. Sci Rep 4:6850.

Ida JA, Jr., Dubois-Dalcq M, McKinnon RD (1993) Expression of the receptor tyrosine kinase c-kit in oligodendrocyte progenitor cells. J Neurosci Res 36:596-606.

Imam SS, Gad GI, Atef SH, Shawky MA (2009) Cord blood brain derived neurotrophic factor: diagnostic and prognostic marker in fullterm newborns with perinatal asphyxia. Pak J Biol Sci 12:1498-1504.

Ip JE, Wu Y, Huang J, Zhang L, Pratt RE, Dzau VJ (2007) Mesenchymal stem cells use integrin beta1 not CXC chemokine receptor 4 for myocardial migration and engraftment. Mol Biol Cell 18:2873-2882.

Ivacko J, Szaflarski J, Malinak C, Flory C, Warren JS, Silverstein FS(1997) Hypoxic-ischemic injury induces monocyte chemoattractant protein-1 expression in neonatal rat brain. J Cereb Blood Flow Metab 17:759-770.

Janowska-Wieczorek A, Matsuzaki A, L AM (2000) The hematopoietic microenvironment: matrix metalloproteinases in the hematopoietic microenvironment. Hematology 4:515-527.

Jiang L, Newman M, Saporta S, Chen N, Sanberg C, Sanberg PR, Willing AE (2008) MIP-1alpha and MCP-1 induce migration of human umbilical cord blood cells in models of stroke. Curr Neurovasc Res 5:118-124.

Jimenez J, Lesage F, Richter J, Nagatomo T, Salaets T, Zia S, Mori Da Cunha MG, Vanoirbeek J, Deprest JA, Toelen J (2018) Upregulation of vascular endothelial growth factor in amniotic fluid stem cells enhances their potential to attenuate lung injury in a preterm rabbit model of bronchopulmonary dysplasia. Neonatology 113:275-285.

Jin K, Mao XO, Greenberg DA (2006) Vascular endothelial growth factor stimulates neurite outgrowth from cerebral cortical neurons via Rho kinase signaling. J Neurobiol 66:236-242.

Jin K, Mao XO, Sun Y, Xie L, Greenberg DA (2002a) Stem cell factor stimulates neurogenesis in vitro and in vivo. J Clin Invest 110:311-319.

Jin K, Zhu Y, Sun Y, Mao XO, Xie L, Greenberg DA (2002b) Vascular endothelial growth factor (VEGF) stimulates neurogenesis in vitro and in vivo. Proc Natl Acad Sci U S A 99:11946-11950.

Junge CE, Sugawara T, Mannaioni G, Alagarsamy S, Conn PJ, Brat DJ,Chan PH, Traynelis SF (2003) The contribution of protease-activated receptor 1 to neuronal damage caused by transient focal cerebral ischemia. Proc Natl Acad Sci U S A 100:13019-13024.

Juul SE, Stallings SA, Christensen RD (1999) Erythropoietin in the cerebrospinal fl uid of neonates who sustained CNS injury. Pediatr Res 46:543-547.

Kawahara N, Croll SD, Wiegand SJ, Klatzo I (1997) Cortical spreading depression induces long-term alterations of BDNF levels in cortex and hippocampus distinct from lesion effects: implications for ischemic tolerance. Neurosci Res 29:37-47.

Khaibullina AA, Rosenstein JM, Krum JM (2004) Vascular endothelial growth factor promotes neurite maturation in primary CNS neuronal cultures. Brain Res Dev Brain Res 148:59-68.

Khairallah MI, Kassem LA, Yassin NA, Gamal el Din MA, Zekri M,Attia M (2016) Activation of migration of endogenous stem cells by erythropoietin as potential rescue for neurodegenerative diseases.Brain Res Bull 121:148-157.

Klein R, Smeyne RJ, Wurst W, Long LK, Auerbach BA, Joyner AL,Barbacid M (1993) Targeted disruption of the trkB neurotrophin receptor gene results in nervous system lesions and neonatal death.Cell 75:113-122.

Koch AE, Kunkel SL, Harlow LA, Mazarakis DD, Haines GK, Burdick MD, Pope RM, Strieter RM (1994) Macrophage in fl ammatory protein-1 alpha. A novel chemotactic cytokine for macrophages in rheumatoid arthritis. J Clin Invest 93:921-928.

Kolev K, Skopal J, Simon L, Csonka E, Machovich R, Nagy Z (2003)Matrix metalloproteinase-9 expression in post-hypoxic human brain capillary endothelial cells: H2O2 as a trigger and NF-kappaB as a signal transducer. Thromb Haemost 90:528-537.

Krueger JM, Fang J, Taishi P, Chen Z, Kushikata T, Gardi J (1998)Sleep. A physiologic role for IL-1 beta and TNF-alpha. Ann N Y Acad Sci 856:148-159.

Kumar S, Ponnazhagan S (2012) Mobilization of bone marrow mesenchymal stem cells in vivo augments bone healing in a mouse model of segmental bone defect. Bone 50:1012-1018.

Kurinczuk JJ, White-Koning M, Badawi N (2010) Epidemiology of neonatal encephalopathy and hypoxic-ischaemic encephalopathy.Early Hum Dev 86:329-338.

Lau LT, Yu AC (2001) Astrocytes produce and release interleukin-1,interleukin-6, tumor necrosis factor alpha and interferon-gamma following traumatic and metabolic injury. J Neurotrauma 18:351-359.

Li J, Guo W, Xiong M, Zhang S, Han H, Chen J, Mao D, Yu H, Zeng Y (2017) Erythropoietin facilitates the recruitment of bone marrow mesenchymal stem cells to sites of spinal cord injury. Exp Ther Med 13:1806-1812.

Li L, McBride DW, Doycheva D, Dixon BJ, Krafft PR, Zhang JH, Tang J (2015) G-CSF attenuates neuroinflammation and stabilizes the blood-brain barrier via the PI3K/Akt/GSK-3beta signaling pathway following neonatal hypoxia-ischemia in rats. Exp Neurol 272:135-144.

Liu J, Feng ZC (2010) Increased umbilical cord plasma interleukin-1 beta levels was correlated with adverse outcomes of neonatal hypoxic-ischemic encephalopathy. J Trop Pediatr 56:178-182.

Liu NM, Tian J, Wang WW, Han GF, Cheng J, Huang J, Zhang JY(2013) Effect of erythropoietin on mesenchymal stem cell differentiation and secretion in vitro in an acute kidney injury microenvironment. Genet Mol Res 12:6477-6487.

Lv H, Wang Q, Wu S, Yang L, Ren P, Yang Y, Gao J, Li L (2015) Neonatal hypoxic ischemic encephalopathy-related biomarkers in serum and cerebrospinal fl uid. Clin Chim Acta 450:282-297.

Ma L, Li XW, Zhang SJ, Yang F, Zhu GM, Yuan XB, Jiang W (2014)Interleukin-1 beta guides the migration of cortical neurons. J Neuroin fl ammation 11:114.

Malamitsi-Puchner A, Economou E, Rigopoulou O, Boutsikou T(2004) Perinatal changes of brain-derived neurotrophic factor in pre- and fullterm neonates. Early Hum Dev 76:17-22.

Manova K, Bachvarova RF, Huang EJ, Sanchez S, Pronovost SM,Velazquez E, McGuire B, Besmer P (1992) c-kit receptor and ligand expression in postnatal development of the mouse cerebellum suggests a function for c-kit in inhibitory interneurons. J Neurosci 12:4663-4676.

Marquez-Curtis LA, Turner AR, Sridharan S, Ratajczak MZ, Janowska-Wieczorek A (2011) The ins and outs of hematopoietic stem cells: studies to improve transplantation outcomes. Stem Cell Rev 7:590-607.

Marti HH, Wenger RH, Rivas LA, Straumann U, Digicaylioglu M,Henn V, Yonekawa Y, Bauer C, Gassmann M (1996) Erythropoietin gene expression in human, monkey and murine brain. Eur J Neurosci 8:666-676.

Matsui Y, Zsebo KM, Hogan BL (1990) Embryonic expression of a haematopoietic growth factor encoded by the Sl locus and the ligand for c-kit. Nature 347:667-669.

Matthys P, Hatse S, Vermeire K, Wuyts A, Bridger G, Henson GW, De Clercq E, Billiau A, Schols D (2001) AMD3100, a potent and speci fi c antagonist of the stromal cell-derived factor-1 chemokine receptor CXCR4, inhibits autoimmune joint in fl ammation in IFN-gamma receptor-de fi cient mice. J Immunol 167:4686-4692.

Millauer B, Wizigmann-Voos S, Schnurch H, Martinez R, Moller NP,Risau W, Ullrich A (1993) High affinity VEGF binding and developmental expression suggest Flk-1 as a major regulator of vasculogenesis and angiogenesis. Cell 72:835-846.

Miller JT, Bartley JH, Wimborne HJ, Walker AL, Hess DC, Hill WD,Carroll JE (2005) The neuroblast and angioblast chemotaxic factor SDF-1 (CXCL12) expression is briefly up regulated by reactive astrocytes in brain following neonatal hypoxic-ischemic injury. BMC Neurosci 6:63.

Mori M, Matsubara K, Matsubara Y, Uchikura Y, Hashimoto H, Fujioka T, Matsumoto T (2015) Stromal cell-derived factor-1alpha plays a crucial role based on neuroprotective role in neonatal brain injury in rats. Int J Mol Sci 16:18018-18032.

Motro B, van der Kooy D, Rossant J, Reith A, Bernstein A (1991) Contiguous patterns of c-kit and steel expression: analysis of mutations at the W and Sl loci. Development (Cambridge, England) 113:1207-1221.

No authors listed (2018) IL1B interleukin 1 beta [Homo sapiens (human)]. In: Bethesda (MD, USA): National Library of Medicine (US),National Center for Biotechnology Information.

Old LJ (1985) Tumor necrosis factor (TNF). Science 230:630-632.

Oranskii IE, Rudakov AA, Mironova IB (1977) Comparative study of the action exerted by sodium chloride iodine-bromine baths, corrected in their composition, on various hemodynamic and hemostasic characteristics in ischemic heart disease. Vopr Kurortol Fizioter Lech Fiz Kult:20-23.

Orrock JE, Panchapakesan K, Vezina G, Chang T, Harris K, Wang Y,Knoblach S, Massaro AN (2016) Association of brain injury and neonatal cytokine response during therapeutic hypothermia in newborns with hypoxic-ischemic encephalopathy. Pediatr Res 79:742-747.

Orzylowska O, Oderfeld-Nowak B, Zaremba M, Januszewski S, Mossakowski M (1999) Prolonged and concomitant induction of astroglial immunoreactivity of interleukin-1beta and interleukin-6 in the rat hippocampus after transient global ischemia. Neurosci Lett 263:72-76.

Oygur N, Sonmez O, Saka O, Yegin O (1998) Predictive value of plasma and cerebrospinal fluid tumour necrosis factor-alpha and interleukin-1 beta concentrations on outcome of full term infants with hypoxic-ischaemic encephalopathy. Arch Dis Child Fetal Neonatal Ed 79:F190-193.

Papayannopoulou T, Craddock C (1997) Homing and trafficking of hemopoietic progenitor cells. Acta Haematologica 97:97-104.

Parikh P, Juul SE (2018) Neuroprotective strategies in neonatal brain injury. J Pediatr 192:22-32.

Peled A, Grabovsky V, Habler L, Sandbank J, Arenzana-Seisdedos F,Petit I, Ben-Hur H, Lapidot T, Alon R (1999) The chemokine SDF-1 stimulates integrin-mediated arrest of CD34(+) cells on vascular endothelium under shear fl ow. J Clin Invest 104:1199-1211.

Petit I, Szyper-Kravitz M, Nagler A, Lahav M, Peled A, Habler L,Ponomaryov T, Taichman RS, Arenzana-Seisdedos F, Fujii N, Sandbank J, Zipori D, Lapidot T (2002) G-CSF induces stem cell mobilization by decreasing bone marrow SDF-1 and up-regulating CXCR4.Nat Immunol 3:687-694.

Pimentel-Coelho PM, Rosado-de-Castro PH, da Fonseca LM, Mendez-Otero R (2012) Umbilical cord blood mononuclear cell transplantation for neonatal hypoxic-ischemic encephalopathy. Pediatr Res 71:464-473.

Plumier JC, David JC, Robertson HA, Currie RW (1997) Cortical application of potassium chloride induces the low-molecular weight heat shock protein (Hsp27) in astrocytes. J Cereb Blood Flow Metab 17:781-790.

Prass K, Scharff A, Ruscher K, Lowl D, Muselmann C, Victorov I, Kapinya K, Dirnagl U, Meisel A (2003) Hypoxia-induced stroke tolerance in the mouse is mediated by erythropoietin. Stroke 34:1981-1986.

Pruijt JF, Fibbe WE, Laterveer L, Pieters RA, Lindley IJ, Paemen L,Masure S, Willemze R, Opdenakker G (1999) Prevention of interleukin-8-induced mobilization of hematopoietic progenitor cells in rhesus monkeys by inhibitory antibodies against the metalloproteinase gelatinase B (MMP-9). Proc Natl Acad Sci U S A 96:10863-10868.

Qiu FH, Ray P, Brown K, Barker PE, Jhanwar S, Ruddle FH, Besmer P(1988) Primary structure of c-kit: relationship with the CSF-1/PDGF receptor kinase family--oncogenic activation of v-kit involves deletion of extracellular domain and C terminus. EMBO J 7:1003-1011.

Ramirez-Rodriguez GB, Perera-Murcia GR, Ortiz-Lopez L, Vega-Rivera NM, Babu H, Garcia-Anaya M, Gonzalez-Olvera JJ (2017)Vascular endothelial growth factor in fl uences migration and focal adhesions, but not proliferation or viability, of human neural stem/progenitor cells derived from olfactory epithelium. Neurochem Int 108:417-425.

Rennert RC, Sorkin M, Garg RK, Gurtner GC (2012) Stem cell recruitment after injury: lessons for regenerative medicine. Regen Med 7:833-850.

Ribatti D, Presta M, Vacca A, Ria R, Giuliani R, Dell’Era P, Nico B,Roncali L, Dammacco F (1999) Human erythropoietin induces a pro-angiogenic phenotype in cultured endothelial cells and stimulates neovascularization in vivo. Blood 93:2627-2636.

Rosell A, Ortega-Aznar A, Alvarez-Sabin J, Fernandez-Cadenas I, Ribo M, Molina CA, Lo EH, Montaner J (2006) Increased brain expression of matrix metalloproteinase-9 after ischemic and hemorrhagic human stroke. Stroke 37:1399-1406.

Rosenkranz K, Kumbruch S, Lebermann K, Marschner K, Jensen A, Dermietzel R, Meier C (2010) The chemokine SDF-1/CXCL12 contributes to the ‘homing’ of umbilical cord blood cells to a hypoxic-ischemic lesion in the rat brain. J Neurosci Res 88:1223-1233.

Ruddy RM, Morshead CM (2018) Home sweet home: the neural stem cell niche throughout development and after injury. Cell Tissue Res 371:125-141.

Ruster B, Gottig S, Ludwig RJ, Bistrian R, Muller S, Seifried E, Gille J,Henschler R (2006) Mesenchymal stem cells display coordinated rolling and adhesion behavior on endothelial cells. Blood 108:3938-3944.

Saliba E, Henrot A (2001) In fl ammatory mediators and neonatal brain damage. Biology of the neonate 79:224-227.

Schmidt NO, Przylecki W, Yang W, Ziu M, Teng Y, Kim SU, Black PM, Aboody KS, Carroll RS (2005) Brain tumor tropism of transplanted human neural stem cells is induced by vascular endothelial growth factor. Neoplasia 7:623-629.

Serfozo P, Schlarman MS, Pierret C, Maria BL, Kirk MD (2006) Selective migration of neuralized embryonic stem cells to stem cell factor and media conditioned by glioma cell lines. Cancer Cell Int 6:1.

Shirvaikar N, Montano J, Turner AR, Ratajczak MZ, Janowska-Wieczorek A (2004) Upregulation of MT1-MMP expression by hyaluronic acid enhances homing-related responses of hematopoietic CD34+cells to an SDF-1 gradient. Blood 104:2889.

Silveira RC, Procianoy RS (2003) Interleukin-6 and tumor necrosis factor-alpha levels in plasma and cerebrospinal fluid of term newborn infants with hypoxic-ischemic encephalopathy. J Pediatr 143:625-629.

Silverstein FS, Barks JD, Hagan P, Liu XH, Ivacko J, Sza fl arski J (1997)Cytokines and perinatal brain injury. Neurochem Int 30:375-383.

Steingen C, Brenig F, Baumgartner L, Schmidt J, Schmidt A, Bloch W(2008) Characterization of key mechanisms in transmigration and invasion of mesenchymal stem cells. J Mol Cell Cardiol 44:1072-1084.

Sullivan CB, Porter RM, Evans CH, Ritter T, Shaw G, Barry F, Murphy JM (2014) TNFalpha and IL-1beta in fl uence the differentiation and migration of murine MSCs independently of the NF-kappaB pathway. Stem Cell Res Ther 5:104.

Sun J, Zhou W, Ma D, Yang Y (2010) Endothelial cells promote neural stem cell proliferation and differentiation associated with VEGF activated Notch and Pten signaling. Dev Dyn 239:2345-2353.

Sun L, Lee J, Fine HA (2004) Neuronally expressed stem cell factor induces neural stem cell migration to areas of brain injury. J Clin Invest 113:1364-1374.

Sun Y, Jin K, Xie L, Childs J, Mao XO, Logvinova A, Greenberg DA(2003) VEGF-induced neuroprotection, neurogenesis, and angiogenesis after focal cerebral ischemia. J Clin Invest 111:1843-1851.

Sunagawa S, Ichiyama T, Honda R, Fukunaga S, Maeba S, Furukawa S(2009) Matrix metalloproteinase-9 and tissue inhibitor of metalloproteinase-1 in perinatal asphyxia. Brain Dev 31:588-593.

Sza fl arski J, Burtrum D, Silverstein FS (1995) Cerebral hypoxia-ischemia stimulates cytokine gene expression in perinatal rats. Stroke 26:1093-1100.

Taub DD, Conlon K, Lloyd AR, Oppenheim JJ, Kelvin DJ (1993) Preferential migration of activated CD4+ and CD8+ T cells in response to MIP-1 alpha and MIP-1 beta. Science 260:355-358.

Teixeira M, Rodrigues-Santos P, Garrido P, Costa E, Parada B, Sereno J, Alves R, Belo L, Teixeira F, Santos-Silva A, Reis F (2012) Cardiac antiapoptotic and proproliferative effect of recombinant human erythropoietin in a moderate stage of chronic renal failure in the rat.J Pharm Bioallied Sci 4:76-83.

Turrin NP, Rivest S (2006) Tumor necrosis factor alpha but not interleukin 1 beta mediates neuroprotection in response to acute nitric oxide excitotoxicity. J Neurosci 26:143-151.

Vagima Y, Avigdor A, Goichberg P, Shivtiel S, Tesio M, Kalinkovich A, Golan K, Dar A, Kollet O, Petit I, Perl O, Rosenthal E, Resnick I,Hardan I, Gellman YN, Naor D, Nagler A, Lapidot T (2009) MT1-MMP and RECK are involved in human CD34+ progenitor cell retention, egress, and mobilization. J Clin Invest 119:492-503.

Van Coillie E, Van Damme J, Opdenakker G (1999) The MCP/eotaxin subfamily of CC chemokines. Cytokine Growth Factor Rev 10:61-86.

Van Lint P, Libert C (2007) Chemokine and cytokine processing by matrix metalloproteinases and its effect on leukocyte migration and in fl ammation. J Leukoc Biol 82:1375-1381.

Vasiljevic B, Maglajlic-Djukic S, Gojnic M, Stankovic S, Ignjatovic S,Lutovac D (2011) New insights into the pathogenesis of perinatal hypoxic-ischemic brain injury. Pediatr Int 53:454-462.

Vermeulen M, Le Pesteur F, Gagnerault MC, Mary JY, Sainteny F,Lepault F (1998) Role of adhesion molecules in the homing and mobilization of murine hematopoietic stem and progenitor cells. Blood 92:894-900.

Wang H, Wang X, Qu J, Yue Q, Hu Y, Zhang H (2015) VEGF enhances the migration of MSCs in neural differentiation by regulating focal adhesion turnover. J Cell Physiol 230:2728-2742.

Wang K, Li Y, Zhu T, Zhang Y, Li W, Lin W, Li J, Zhu C (2017) Overexpression of c-Met in bone marrow mesenchymal stem cells improves their effectiveness in homing and repair of acute liver failure.Stem Cell Res Ther 8:162.

Wang X, Jung J, Asahi M, Chwang W, Russo L, Moskowitz MA, Dixon CE, Fini ME, Lo EH (2000) Effects of matrix metalloproteinase-9 gene knock-out on morphological and motor outcomes after traumatic brain injury. J Neurosci 20:7037-7042.

Wang Y, Deng Y, Zhou GQ (2008) SDF-1alpha/CXCR4-mediated migration of systemically transplanted bone marrow stromal cells towards ischemic brain lesion in a rat model. Brain Res 1195:104-112.

Wride MA, Sanders EJ (1995) Potential roles for tumour necrosis factor alpha during embryonic development. Anat Embryol (Berl)191:1-10.

Wu YW, Mathur AM, Chang T, McKinstry RC, Mulkey SB, Mayock DE, Van Meurs KP, Rogers EE, Gonzalez FF, Comstock BA, Juul SE, Msall ME, Bonifacio SL, Glass HC, Massaro AN, Dong L, Tan KW, Heagerty PJ, Ballard RA (2016) High-dose erythropoietin and hypothermia for hypoxic-ischemic encephalopathy: a phase II trial.Pediatrics 137.

Xiang Q, Hong D, Liao Y, Cao Y, Liu M, Pang J, Zhou J, Wang G,Yang R, Wang M, Xiang AP (2017) Overexpression of gremlin1 in mesenchymal stem cells improves hindlimb ischemia in mice by enhancing cell survival. J Cell Physiol 232:996-1007.

Xiao Q, Wang SK, Tian H, Xin L, Zou ZG, Hu YL, Chang CM, Wang XY, Yin QS, Zhang XH, Wang LY (2012) TNF-alpha increases bone marrow mesenchymal stem cell migration to ischemic tissues. Cell Biochem Biophys 62:409-414.

Xiong M, Chen S, Yu H, Liu Z, Zeng Y, Li F (2011) Neuroprotection of erythropoietin and methylprednisolone against spinal cord ischemia-reperfusion injury. J Huazhong Univ Sci Technolog Med Sci 31:652-656.

Yang Y, Rosenberg GA (2015) Matrix metalloproteinases as therapeutic targets for stroke. Brain Res 1623:30-38.

Yao Y, Zheng XR, Zhang SS, Wang X, Yu XH, Tan JL, Yang YJ (2016)Transplantation of vascular endothelial growth factor-modified neural stem/progenitor cells promotes the recovery of neurological function following hypoxic-ischemic brain damage. Neural Regen Res 11:1456-1463.

Yu Q, Liu L, Lin J, Wang Y, Xuan X, Guo Y, Hu S (2015) SDF-1α/CXCR4 axis mediates the migration of mesenchymal stem cells to the hypoxic-ischemic brain lesion in a rat model. Cell J 16:440-447.

Zgheib C, Xu J, Mallette AC, Caskey RC, Zhang L, Hu J, Liechty KW(2015) SCF increases in utero-labeled stem cells migration and improves wound healing. Wound Repair Regen 23:583-590.

Zhang SC, Fedoroff S (1997) Cellular localization of stem cell factor and c-kit receptor in the mouse nervous system. J Neurosci Res 47:1-15.

Zhang SC, Fedoroff S (1999) Expression of stem cell factor and c-kit receptor in neural cells after brain injury. Acta Neuropathol 97:393-398.

Zhao BQ, Wang S, Kim HY, Storrie H, Rosen BR, Mooney DJ, Wang X,Lo EH (2006) Role of matrix metalloproteinases in delayed cortical responses after stroke. Nat Med 12:441-445.

Zhu C, Kang W, Xu F, Cheng X, Zhang Z, Jia L, Ji L, Guo X, Xiong H, Simbruner G, Blomgren K, Wang X (2009) Erythropoietin improved neurologic outcomes in newborns with hypoxic-ischemic encephalopathy. Pediatrics 124:e218-226.

Zwezdaryk KJ, Coffelt SB, Figueroa YG, Liu J, Phinney DG, LaMarca HL, Florez L, Morris CB, Hoyle GW, Scandurro AB (2007) Erythropoietin, a hypoxia-regulated factor, elicits a pro-angiogenic program in human mesenchymal stem cells. Exp Hematol 35:640-652.

StephanieM.Parry,EricS.Peeples
《Neural Regeneration Research》2018年第7期文献

服务严谨可靠 7×14小时在线支持 支持宝特邀商家 不满意退款

本站非杂志社官网,上千家国家级期刊、省级期刊、北大核心、南大核心、专业的职称论文发表网站。
职称论文发表、杂志论文发表、期刊征稿、期刊投稿,论文发表指导正规机构。是您首选最可靠,最快速的期刊论文发表网站。
免责声明:本网站部分资源、信息来源于网络,完全免费共享,仅供学习和研究使用,版权和著作权归原作者所有
如有不愿意被转载的情况,请通知我们删除已转载的信息 粤ICP备2023046998号