更全的杂志信息网

The progress of prophylactic treatment in retinopathy of prematurity

更新时间:2016-07-05

INTRODUCTION

Retinopathy of prematurity (ROP) is one of the leading causes of visual loss in children[1]. Although the timely diagnosis and treatment of ROP have resulted in better improvement of retinal structure and visual acuity than before,however, ROP remains as a major cause of blindness in premature infants and the incidence is increased along with the elevated survival of infants born at very early gestational ages[2-6]. ROP can be divided into an early ischemic stage and a late neovascular stage, and early retinal ischemia leads to late retinal neovascularization (RNV)[7-8]. At present, most of the therapeutic treatments are focusing on depressing RNV[9-10].However, at some circumstance, such as residual anatomical changes, amblyopia and high ametropia, and injury to the vulnerable retina caused by treatments themselves, make it difficult to restore impaired vision function by inhibiting RNV[11-17]. These studies suggest that treatment for ROP should be done prior to RNV. We predict that the optimal timing for ROP treatment should focus on retinopathy during the early is chemic stage but not during the late neovascular stage.Indeed, early treatment of retinopathy of prematurity (ETROP)has been proven to be promising method for rescuing visual function in premature infants[18-25]. However, currently, ETROP still aimed at pre-threshold retinopathy, which still hurt retina,thus, to actively explore the optimal therapeutic strategy for ETROP is necessary. In this review, we summarize the potential mechanisms involved in early ischemic stage ROP,which may contribute to improve ETROP in the future.

OXYGEN AND RETINOPATHY OF PREMATURITY

Oxygen plays a critical role in ROP[26]. It was found that the relatively high levels of oxygen routinely given to premature infants were an important risk factor, and that reducing the level of oxygen given to premature babies reduced the incidence of ROP[27-28]. With advanced technology to monitor the oxygen levels applied to infants, the importance of oxygen as a risk factor has been diminished. However, for understanding the mechanisms of ROP, oxygen-induced retinopathy (OIR) is still used to generate ROP in animal models[29]. OIR can be divided into an early hyperoxic phase and a late hypoxic phase as in ROP[30]. The morphological changes of retina in the early phase of OIR are more obvious than those in the late phase[31].To explore the mechanism of hyperoxia-induced vascular loss in the early phase of OIR, which is closely related to primary retinal vascular loss in ROP, may help to develop better therapeutic strategies for treating ROP.

观念障碍:对实验课程教学本质含义认知的片面化在传统高职院校教学过程中,过度重视教师个人在理论教学过程中的作用,过度强调教学个体自主性,忽视了团队建设。其主要原因是,教学工作不是单独一个教师能够完成的工程,它需要团队合作,只有这样才能够完成教学这个巨大的工程。传统的教学方式在很大程度上限制了现阶段的教学进步,过度强调教学个体在教学活动中的作用,忽视教学团队的力量,实验课程团队教学得到的重视程度远远不够[2]。

Limited oxygen application is able to reduce the incidences of ROP, whereas it is concomitant to an increase in mortality among preterm infants. Thus, studies of how to avoid the occurrence of ROP while an infant is treated with oxygen are significant[32-33]. Clinical studies indicated that newborn resuscitation should not be conducted with 100% oxygen supplementation and the levels of SaO2 during the neonatal period in extremely low-birth-weight (LBW) infants should be maintained at between 85% and 93% or possibly between 88% and 95%, but absolutely not exceed 95%, and fluctuations should be avoided[34]. Maintain of SaO2 values between 83%and 93% in the period immediately following birth combined with the strict control of oxygen fl uctuations could prevent the early vaso-obliterative phase and the subsequent development of severe ROP in very LBW premature infants[35]. It had also been observed that the hyperoxia of 85%-93% versus 90%-99%was beneficial for the development of the immature retinal vasculature and decreased the incidence of ROP in preterm infants with body weights ≤1000 g[36].

Lower oxygen (85% to 92% SaO2) at early gestational ages(<34wk) and higher oxygen (92% to 97% SaO2) at older gestational ages (>34wk) induced normal retinal development and decreased the severity and the incidence of ROP[37-38].However, there is a debate of whether partially decreasing SaO2 increased the rate of mortality or disability in premature infants. It was reported that SaO2 of less than 90% in extremely preterm infants was associated with an increased risk of death,whereas one study found SaO2 between 85%-89% versus 91%-95% resulted in no significant effects on the mortality rate or the disability rate in extremely preterm 18mo infants, while recently, another study denied this opinion once again[32,39-40].In addition, oxygen used with 90%-99% versus 85%-93% has the similar clinical effects on the development of the early and late type 1 ROP[41]. It is also observed that hyperoxia treatment(40%-75% SaO2) initiated on P14 during the pre-proliferative phase of ischemic retinopathy was effective in accelerating the process of retinal revascularization and preventing the development of RNV[42]. Thus, hyperoxia is not always harmful to premature infants, and in some circumstance, it may be beneficial for the development of retinal vessels and reduce the incidence of ROP. Thus, to what extent that hyperoxia should be lowered which benefit to the development of retinal vasculature and simultaneously do not harm to the fate of premature infants still need be further investigated in clinical studies.

含钼花岗岩中常见的副矿物主要有磁铁矿、钛铁矿、金红石、锆石、磷灰石、榍石、锐钛矿及独居石等,有时可见褐帘石、钛金红石、辉钼矿、刚玉、磷钇矿、多钛钶矿等。

HYPOXIA INDUCIBLE FACTOR AND EARLY RETINOPATHY OF PREMATURITY

2014年春节之后,60岁的二哥为了减少自己的压力,为了避免家族企业利益与情感的冲突期,感到应该是自己退出职场的时候了,他没有考虑退休后的生活保障,没有要任何钱,无条件退出倾注自己心血与汗水的工作平台。

VEGF AND EARLY RETINOPATHY OF PREMATURITY

VEGF stimulates vasculogenesis and angiogenesis and is primarily regulated by HIF-1 at the transcriptional level under the condition of hypoxia[61]. VEGF can bind with its two membrane-bound receptors: VEGFR-1 and VEGFR-2, on endothelial cells[62]. The primary role of VEGFR-1 after binding by VEGF is to negatively regulate the bioactivity of VEGFR-2[63-64]. VEGFR-2 is essential for endothelial physiology and pathology during development, including the processes of angiogenesis and neovascularization[64-66]. In a mouse model of ischemia-induced retinal revascularization,an increased level of VEGFR-2 was noted in the vessels near the avascular area, whereas VEGFR-1 expression in the hypoxic retina was almost the same compared to that in control animals[67]. A critical role of VEGFR-1 in maintaining the vasculature of the neonatal retina has been reported and activation of VEGFR-1 by placental growth factor-1 has been proposed as an alternative strategy for preventing OIR without provoking RNV[68]. It has been noted that the levels of serum VEGF were lower at birth in infants who developed ROP than infants without ROP, and it remained low in children with ROP who required treatment[69-70], which suggested that addition of VEGF-A during the early stage might be beneficial to the development of the retinal vasculature in preterm infants. However, it is also reported that exogenous VEGF administration on P14 was not sufficient to induce RNV in hyperoxia treatment mice, whereas injection of the VEGF antagonist VEGFR1/Fc blocked both pathologic and physiological angiogenesis but did not rescue astrocytes[42]. In addition, administration of a neutralizing antibody targeting VEGF decreased the phosphorylation of VEGFR-2 within the retina and around the blood vessels, and increased levels of VEGF in the free intraretinal space during the early stage of OIR, leading to significantly and sustainably reduce RNV without interfering in the ongoing retinal vascularization[71].Thus, early administration of VEGF may be not enough to promote retinal vascular development in premature infants, and it is necessary to investigate the role of VEGFR1 and VEGFR2 in this process.

Furthermore, the effect of superoxide dismutase 1 and 2 and vitamin E on the prophylactic treatment of ROP has also been shown in the clinic, although complications of high rates of sepsis and necrotizing enterocolitis resulting from vitamin E therapy have made it difficult to use routinely for the prophylaxis of ROP[111-114]. In sum, these studies suggest that oxidative stress plays a crucial role in the formation of the avascular area during the hyperoxia phase of OIR and ROP.

NORRIN AND EARLY RETINOPATHY OF PREMATURITY

Norrin is constitutively expressed in the retina and involved in retinal angiogenesis. Together with its receptor (Frizzled-4,FZD4), Norrin activates the Wnt-signalling pathway and controls the formation of the retinal vasculature during eye development[72-73]. It was reported that Norrin could significantly reduce vascular loss in transgenic mice with OIR and could increase the anatomically accurate regrowth of vessels while suppressing RNV[74]. Abnormal Norrin production led to premature retinal vascular invasion, resulting in characteristic defects in the intraretinal vascular architecture[75]. In addition,an antagonist of FZD4 not only inhibited physiological and pathologic sprouting angiogenesis within the retina but also induced the upregulation of plasma lemma vesicle-associated protein. Thus, FZD4 is required for physiological and pathologic angiogenesis in the retina and for the regulation of retinal endothelial cell differentiation[76]. These studies suggest the role of Norrin in ROP and its therapeutic potential in the treatment of the early stage ROP.

INSULIN-LIKE GROWTH FACTORS AND EARLY RETINOPATHY OF PREMATURITY

Insulin-like growth factor (IGFs) constitute a large family of insulin-related peptides that include IGF-I and IGF-II, their cell surface receptors (IGF-IR and IGF-IIR), and IGF binding proteins (IGFBP-1 through -6) as well as their proteases and interacting molecules, which work together to regulate cell proliferation, differentiation and apoptosis[77-78]. The majority of circulating IGF-I and IGF-II binds to IGFBPs, whereas IGFBPs also regulate their biological activities and modulate cellular activity via IGF independent pathways[79-80]. It has been found that IGF-IR and insulin receptor (IR) are predominantly expressed in photoreceptors and blood vessels, in which the expression of IGF-IR is 100-fold more than that of IR[81].IGFBP-3 expression in neovascular tufts of OIR was increased more than 5-fold during hypoxia, whereas IGFBP2, IGFBP4 and IGFBP5 expression remains unchanged. In addition, it also found that neonatal mice from larger litters showed lower body weights and lower levels of circulating IGF-I than mice from smaller litters and that they were more susceptible to developing more severe OIR[82]. The early administration of IGF-I at postnatal day 4 increased body weight and resulted in more rapid maturation and less OIR[82]. The early short-term systemic administration of JB1, an IGF-I analog, increased soluble VEGFR-1 and decreased retinal OIR pathology more effectively than long-term treatment with JB1[83]. It has also been reported that the IGFI levels are deficient after premature birth and that, at birth, non-ROP neonates show higher IGF-I levels than neonates with ROP. The severity and duration of low-serum IGF-I are correlated with the development of ROP[84-86]. The serum levels of IGF-I during the third week post partum provide a suf fi cient and reliable prognostic marker for the identi fi cation of patients at high risk to develop ROP[87].IGFBP-3 regulates the elevated expression of endothelial NO synthase in human endothelial progenitor cells (EPCs) and promotes NO generation, thereby facilitating EPC migration into the ischemic retina. IGFBP-3 exposure also led to the redistribution of vasodilator-stimulated phosphoprotein, an NO-regulated protein critical for cell migration. Furthermore,IGFBP-3 increased pericyte ensheathment and reduced pericyte apoptosis, activated microglia and induced the apoptosis of neuronal cells in the developing retina with OIR, resulting in a more stable retinal vascular bed[88-89]. Fresh-frozen plasma(FFP) from adult donors contains higher concentrations of IGF-I and IGFBP-3, and two or more transfusions of FFP during the first week of life decreases the risk of developing any grade of ROP in preterm infants with a gestational age of less than 29wk[90-91]. Direct and continuous intravenous infusion of IGF-I/IGFBP-3 was effectively and safely to increase the serum concentrations of IGF-I and IGFBP-3 in preterm infants[92]. The prolonged administration of IGF-I/IGFBP-3 did not show any negative impact on blood-glucose levels and was beneficial for the total body growth of neonatal mice[93]. These studies suggest that IGF-I and IGFBP-3 may be safely used for ETROP.

OXIDATIVE STRESS AND EARLY RETINOPATHY OF PREMATURITY

Oxidative stress plays an important role in angiogenesis and neovascularization[94-95] and is also crucial for ROP.Oxidative stress is an imbalance between the production of reactive oxygen species (ROS) and reactive nitrogen species (RNS), in which cyclo-oxygenase (COX), NADPH oxidase and NO synthase are their important sources[95-97].It has been reported that the administration of indomethacin and ibuprofen, improved OIR during the hyperoxia phase in newborn C57BL/6J mice without affecting the normal retinal development. Administration of high-dose ibuprofen at birth decreased retinal VEGF levels and VEGFR-2 transcripts in a rat OIR model at postnatal day 14, whereas indomethacin only suppressed retinal VEGF164 transcripts with no effects on the expression of VEGF receptors[98-99]. Furthermore, ibuprofen is more effective than indomethacin in suppressing retinal VEGF signaling. Newborn rats treated with high-dose ibuprofen at birth showed significantly less somatic growth and higher serum and vitreous IGF-I levels than indomethacin treated rats[100]. However, early administration of indomethacin exerted more potent suppressive effects than ibuprofen on growth hormone binding protein, somatic growth, renal COX-2 and vasodilator prostanoids[101-102]. In addition, both of ibuprofen and indomethacin could increase serum IGF-I[101-102]. Although the administration of ibuprofen and indomethacin during the hyperoxia phase was able to improve OIR in the neovascular phase, their roles in ameliorating early ROP and their adverse effects on the development of newborn infants needs to be further investigated.

It has also been shown that treatment with apocynin, a NAPDH oxidase inhibitor, reduced avascularity and apoptosis in the OIR model via pathways triggered by the generation of ROS[30,103]. In addition, glutathione peroxidase-1, Nrf2,epicatechin, the thiol donor N-acetylcysteine and vitamin E have all been shown to protect against retinal vascular cell death and to reduce the avascular OIR area in hyperoxia,and additionally, Nrf2 can protects against oxidative stressmediated damage to glia of OIR retina in hyperoxia[104-109].

RNS also plays an important role in early ROP besides ROS.It has been reported that in OIR, hyperoxia-induced vascular injury is mediated by dysfunction of endothelial NO synthase,which results in peroxynitrite formation. Treatment with arginase inhibitor 2 or deletion of the arginase 2 gene can normalize NOS activity and reduce peroxynitrite formation,thus prevents hyperoxia-induced retinal vascular injury[110].

扎实推进城乡公共文化服务一体化建设;深化文化体制改革,建立健全文化管理体系。要扎实开展精神扶贫活动,提供适合特殊群体的文化服务和产品。为广大农民群众提供丰富的文化产品。坚持文化惠民,完善基础设施,激发村民的文化自觉,提高农民的整体素质,提升农民生活品质,以文化自觉成就文化自信。此外,大力宣传乡土文化,打造具有乡土文化特色的景点,实现乡村振兴[9-10]。

NEURAL RETINA AND EARLY RETINOPATHY OF PREMATURITY

Somatostatin and Early Retinopathy of Prematurity Somatostatin inhibits the secretion of many hormones by binding to G-protein-coupled somatostatin receptors (Sstr) and then activating cellar signaling pathways, such as adenylate cyclase/cAMP and MAPK signaling[127]. It has been showed that in OIR, somatostatin and Sstr2 levels were reduced. Sstr2 was decreased in the neuroretina but increased in capillaries.Octreotide, a Sstr2 agonist, caused a notable reduction in the hypoxia-induced increase in VEGF and its receptors and inhibited apoptotic signals from retinal cells, resulting in recovery of the a- and b-waves of electroretinograms[128-130]. In the ischemic retina, VEGF was released by damaged neurons and reached the retinal capillaries, whereas the activation of Sstr2 protected neurons from ischemic damage by decrease of the VEGF release and response[131-132]. Growing evidence indicate that retinal neurodegeneration is an early event in the pathogenesis of diabetic retinopathy (DR) and administration of somatostatin has been contemplated as an appropriate therapeutic approach for DR[133]. As DR initiates with neurodegeneration as that occurs in ROP, thus, it is worth to test whether administration of somatostatin is an option for the prophylaxis of ROP.

NEUROPEPTIDES AND EARLY RETINOPATHY OF PREMATURITY

Neuropeptides and their receptors are widely distributed throughout the central and peripheral nervous systems and the peripheral organs including the retina[125-126] and their roles in ROP have been explored.

The sensitivity of photoreceptor and postreceptor cells in a rat OIR model at early ages was associated with vascular tortuosity[115]. Following the cessation of oxygen exposure, the recovery of postreceptor neural retinal-cell sensitivity and the decrease of vascular tortuosity occurred in parallel. Furthermore, mRNA expression of VEGF(164)and semaphorin IIIA (Sema3A), the neuronal guidance cue proapoptotic/repulsive factor, was elevated early and decreased with age. Low sensitivity of rod photoreceptors and postreceptor cells were significantly associated with high VEGF(164) and Sema3A expression[115-116]. Sema3A can be secreted by hypoxic neurons in the avascular retina in response to the proinflammatory cytokine interleukin-1β(IL-1β)[117]. Sema3A contributes to vascular decay and the later formation of a chemical barrier that repels neo-vessels toward the vitreous through interfering with the actions of the IL-1 receptor[117]. Sema3A can enhance normal vascular regeneration within the ischemic retina, thereby diminishing aberrant neovascularization and preserving neuroretinal function[117-118]. The reduction in astrocyte density induced by hyperoxia led to a reduced astrocytic network in hypoxia[119].Astrocytes provide important guidance for RNV. Protection of the retinal astrocytes and microglia was correlated directly with accelerated revascularization of the normal retinal plexuses and a reduction in RNV, which are normally associated with OIR[119-120]. Müller cell-derived and astrocyte-derived VEGF played a minor role in the development of the normal retinal vasculature, whereas it played an important role in hyperoxiainduced vaso-obliteration and RNV[121-123]. The ablation of the expression of neural cell adhesion molecule (N-CAM) from Müller cells and astrocytes, resulted in reduced vascular tuft formation in OIR, whereas retinal developmental angiogenesis remained unaffected[124]. These studies suggest that the neural retina appears to mediate the vascular abnormalities in OIR,and early treatment that targets the neural retina by decrease of Sema3A and N-CAM may be beneficial to retinal vasculature of ROP in hyperoxia.

Neuropeptide Y and Early Retinopathy of Prematurity Neuropeptide Y (NPY) is located primarily in the majority of sympathetic nerve fi bers and regulates hormone release from the pineal glands of mammals[134]. It has been showed[135] that retinal NPY and NPY-Y2 receptor expression was altered during the development of OIR in a mouse model and this alteration might provide a target for potential modification during the development of retinopathy. However, the change in NPY levels without the presence of NPY-Y2 receptors in the immature retina, indicate that NPY may not be involved in the physiological vascularization of the retina. A recent study further showed[136] that NPY decreased only to a slight extent during hyperoxia, and a more pronounced decrease in NPY was significantly delayed during relative hypoxia. Therefore,whether NPY is involved in early ROP is uncertain and needs to be determined.

Other Neuropeptides and Early Retinopathy of Prematurity In addition to somatostatin and NPY, vasoactive intestinal peptide, opioid peptides, angiotensin II (Ang II) and other peptides have been explored in the ischemic retina[137].However, their roles in early ROP are also uncertain. Based on neuropeptides being neuroprotective for the retina and on the intimate relationship between the retinal neurons and the retinal vasculature, it would be valuable to actively explore the role of neuropeptides in the prophylaxis of early ROP.

早在明成祖朱棣为燕王时,就贿赂太监刺探建文帝言行。称帝后更加宠信太监,“盖明世宦官出使、专征、监军、分镇、刺臣民隐事诸大权,皆自永乐间始”。[13]太监一旦大权在握,就为所欲为。因此,阮尧民及其亲信军士的不法行为是有可能的,而激变民众正是这种不法行为的必然产物。有司认为刘清仅损失粮料一罪的审讯结果,也可反证此说成立和刘清不负主要责任。此外,明廷诏命此次造船领兵者共两人,太监阮尧民居首,刘清在其后,说明阮尧民握有全权负首责。

ENDOCRINE HORMONES AND EARLY RETINOPATHY OF PREMATURITY

Erythropoietin and Early Retinopathy of Prematurity Erythropoietin (Epo) is effective in maintaining the erythrocyte mass in the circulation and shows marked neuroprotective and neotrophic effects[138-139]. Although early administration of high-dose recombinant human Epo to very preterm infants did not markedly improve brain injury or ROP [140], the low plasma levels of Epo in preterm infants provided a rationale for the use of Epo to prevent or treat anemia. It was found that local retinal Epo levels were suppressed during the vessel-loss phase and the early administration of exogenous Epo not only prevented both vessel dropout and subsequent RNV but also protected against hypoxia-induced retinal neuronal apoptosis[141]. In contrast, retinal Epo mRNA levels were highly elevated during the neovascular phase of retinopathy, resulting in that late exogenous Epo treatment did not protect the retina but instead enhanced RNV. However, it has been reported that early Epo treatment significantly increased the overall risk of ROP (any grade) compared with late Epo administration (initiated at 8 to 28 days of age)[142], and early Epo+Fe administration could induce the appearance of grade 1 ROP[143]. Therefore,understanding the change of Epo levels during ROP is critical for determining the timing for treating ROP.

Glucocorticoids and Early Retinopathy of Prematurity Early treatment with triamcinolone acetonide reduced neovascularization and subsequent endostatin presence in an OIR model, and late treatment limited pathological vascular sprouting but did not interfere with normal vascularization of the retina[144]. Ng et al[145] reported that the stage of ROP was significantly associated with the basal and peak plasma levels of adreno-cortico-tropic-hormone (ACTH) and with peak serum cortisol levels at P7. It was also reported that antenatal corticosteroids (ACS) reduced the need for exogenous surfactant endotracheal tube insertion at birth in very LBW premature infants, whereas the development of ROP did not differ between groups administered one dose or multiple doses of ACS or between a betamethasone-treated group and a dexamethasone-treated group[146]. In addition, low-dose dexamethasone therapy in 4-7 days old preterm infants with surfactant-pretreated respiratory distress syndrome facilitated weaning from mechanical ventilation and shortened the duration of oxygen supplementation, although the incidence of ROP was not different at P28[147]. Thus, whether application of glucocorticoids works in the prophylaxis of ROP cannot be concluded at present.

Early Nutrition and Weight Gain and Early Retinopathy of Prematurity Premature infants often have low weights,and early nutrition plans and weight gain have been speculated to be beneficial to the treatment of ROP. However, only limited data regarding the administration of early parenteral and enteral nutrition to very LBW infants are available[193]. It has been reported that newborn OIR mice with poor postnatal nutrition and poor weight gain exhibit a prolonged phase of proliferative retinopathy, prolonged overexpression of VEGF,low serum non-fasting levels of glucose, insulin, and IGF-I and high levels of ghrelin during the early post-OIR phase[158]. In addition, early and aggressive introduction of total parenteral nutrition and enteral feeding could benefit weight, length and head circumference measurements, reduce nutritional de fi cits in very LBW infants, increase the levels of IGF-I and IGFBP3 and reduce the risk of ROP, while other study found that feeding with human milk and vitamin, rather than parenteral nutrition, reduced the rate of severe ROP[194-198]. The risk of developing severe ROP in extremely premature infants could also be reduced by providing nutritional support via special delivery of lipids and total calories to increase weight gain,and recently, a fi sh-oil based lipid emulsion (LE) and a newer LE from alternative lipid sources with reduced polyunsaturated fatty acid (PUFA) content compared to the conventional soybean oil based LE has been preliminarily proved to be more effective in decreasing the early stages (1-2) of ROP[199-201]. It has also been suggested that monitoring postnatal longitudinal systemic factors, such as weight gain and IGF-I and IGFBP3 levels, may enhance the clinician’s ability to identify the patients who would require treatment for ROP[202]. Studies further indicated that WINROP, could accurately predict when the rate of weight gain would be decreased to a speci fi c threshold, and enabled early detection in 100% of infants who developed ROP and required treatment[203-205]. ROP typically occurs 4-6wk after birth in premature infants, which provides sufficient time to supply premature infants with adequate and reasonable nutrition to reduce the incidence and severity of ROP, and it has been proved that poor postnatal weight gain in the first two weeks is an independent risk factor for ROP requiring treatment[206], so it is meaningful to actively explore the components and methods that should be used for nutritive treatment for premature infants.

全国家用电器工业信息中心(NAIC)是受中国轻工业联合会委托、经中国轻工业信息中心批准设立的专业信息机构,主要开展行业信息收集、调查、分析研究,并组织编写国内外家用电器行业经济、科技的发展状况、水平、动向、差距和发展趋势,促进行业健康、快速发展。同时,全国家用电器工业信息中心还受中国轻工业信息中心指导,承担家用电器行业科技新成果、新技术的择优推荐工作。

Ghrelin and Early Retinopathy of Prematurity Ghrelin,a gastrointestinal endocrine peptide and predominantly generated in the gut, which also expressed in the rodent eye with the highest expression levels occurring in the retina and iris[156-157]. It was reported that OIR pups with poor weight gain showed high levels of ghrelin during the early post-OIR phase[158]. Ghrelin was produced locally in the retina and its level decreased during the vaso-obliterative phase but increased during the proliferative phase of OI [159]. Intravitreal delivery of ghrelin significantly reduced retinal vessel loss during the hyperoxic phase of OIR whereas ghrelin promoted pathologic angiogenesis during the neovascular phase[159].These fi ndings suggest that early supplementation with ghrelin might contribute to the retinal vasculature in hyperoxia, thus reducing RNV. Whether ghrelin plays a role in the prophylaxis of ROP need to be investigated.

Insulin and Early Retinopathy of Prematurity Hyperglycemia has been associated with the development of ROP in premature infants[160-161]. Early insulin therapy could decrease blood-glucose and increase IGF-I bioactivities,resulting in decrease of morbidity associated with hyperglycemia and IGF-I levels[162]. Although insulin treatment in premature infants has been suggested to increase the risk of ROP, another study showed that insulin infusions for hyperglycemia were safe and resulted in infrequent episodes of hypoglycemia with no increased risk of ROP[163-164]. In addition, the results from randomized trials also could not provide suf fi cient evidence to determine the effects of insulin administration in treating or preventing neonatal hyperglycemia in very LBW infants with ROP or other adverse effects[165-166].Thus, whether insulin treatment for hyperglycemia is beneficial for the prophylaxis of ROP needs to be further investigated.

Hypoxia inducible factor-1 (HIF-1), including an oxygenregulated HIF-1α subunit and a constitutively expressed HIF-1β subunit, regulates gene transcription by binding to the hypoxiaresponse elements in gene promoters. Some of the HIF-1 target genes are involved in adapting to insufficient oxygen or hypoxia[43-47]. HIF-1α is regulated by prolyl hydroxylases(PHD1, PHD2, and PHD3) and asparagine hydroxylase factor inhibiting HIF (FIH-1)[48-50]. During normoxia and hyperoxia, HIF-1α is unstable due to its hydroxylation and subsequent ubiquitination and proteasomal degradation[51-53],whereas during hypoxia, HIF-1α can be stabilized by PHDs via post-translational modifications[54-55]. It has been reported that increasing HIF-1α expression or decreasing HIF-1α degradation during the early stage of OIR can markedly reduce the avascular area and prevent hyperoxia-induced vessel loss, and recently, the role of liver-specific HIF-1α in promoting retinal vasculature in hyperoxia has been further proved[56-57]. However, the expression of HIF-1α in the late phase of OIR results in an inevitable increase in vascular endothelial growth factor (VEGF) expression, which accelerates neovascularization[58]. In addition, the systemic administration of dimethyloxalylglycine, a PHD inhibitor, during the early phase of OIR stabilizes HIF activity in the retina, and prevent oxygen-induced central-vessel loss and subsequent vascular tortuosity and tufting, which reduces subsequent RNV[59-60].These studies suggest that HIF-1α may be a drug target and inhibition of PHDs during the early ischemic stage may be an effective treatment for ROP.

Angiopoietin and Early Retinopathy of Prematurity Ang is an important modulator of angiogenesis. Together with Tie receptors, Ang is essential for embryonic vessel assembly and maturation and functions as a key regulator of adult vascular homeostasis[167]. Ang-1 recruits pericytes and smooth muscle cells during vascular remodeling and integrates them during neovascularization[168-170], whereas Ang-2 is a natural antagonist of Ang-1 and Tie-2[171]. There was a negative correlation between the Ang-1 and Ang-2 levels in moderately and mildly vascular-active ROP eyes[172]. Ang-1 was found to play a substantial role in the formation of the retinal vascular network during postnatal development. It was reported that Ang-1 supplementation rescued vascular retinopathies by simultaneously promoting healthy vascular network formation and inhibiting subsequent abnormal angiogenesis and neuronal dysfunction in the retinas of an OIR model[173]. The functions of Ang-1 might be related to a dual signalling pathway of Tie-2 signaling in the vascular region and integrin αvβ5 signaling in astrocytes[173]. Oliner et al[174] observed that AMG 386 (a selective Ang-1/2-neutralizing peptibody) prevented RNV in OIR when administered from P8 to P16, but it transiently impeded regression of these abnormal vessels when administered from P17 to P23. Combining AMG 386 with VEGF inhibition also led to cooperative prevention of retinal angiogenesis in this model. Whether Ang-1 supplementation is beneficial for ROP prophylaxis needs to be further investigated.

第一,对施工单位报告进行审查,核查。保证各项测量工具、操作设备、技术方案等符合设计要求、符合施工要求、满足安全性能要求,当各项准备要素达标后便可使用。施工设备的质量问题是影响施工质量的重要因素,因此必须要进行严格的质检,否则后患无穷。

教材是教师教学的帮手,也是规范教学和传承教育内容的保证。礼仪课程的教材种类繁多,但很少有教材完全符合教学需求,只是部分对教学有价值,如何汲取有价值的部分,使其固化为适合本校学生的教材,这对所有礼仪教师都是挑战。因此,性质相同、教学体系相似的院校的旅游礼仪教师可合作共同编制礼仪教材,根据本校学生的实际情况,将理论与实践内容以文字的形式展现,但必须遵循以下几个原则:结合院校的性质,教材内容符合学生的可接受性;摒弃学科中心主义的思想,建立新的教材内容体系框架;反映时代特征,体现职业适用性价值;体现教材的教育性价值,使学生多方面获益[9]。在此基础上,规范、完善理论及形体实践教学内容。

Adrenergic System and Early Retinopathy of Prematurity The adrenergic system involves the primary ligands epinephrine and norepinephrine and their adrenergic receptor(AR) families: α1, α2, α3, β1, β2 and β3 receptors[175-176]. It has been reported that in OIR, hypoxia did not influence β-AR expression beyond increasing β3-AR expression in engorged retinal tufts[177]. Propranolol dose-dependently reduced the upregulation of VEGF and decreased hypoxic levels of IGF-I mRNA and HIF-1α, thereby protecting against retinal angiogenesis and ameliorating blood-retinal barrier dysfunction in OIR[177]. It was also reported that a β2-AR antagonist, ICI 118 551, regulated Müller-cell release of VEGF in an OIR model, indicating that β2-AR activity might play a role in RO[178]. Although propranolol could retard the progression of stage 2 ROP to stages 3 and 4 ROP as well as reduce laser and intravitreal Lucentis treatment, however, it is controversial whether it can induce serious systemic adverse effects[179-180].By the way, 2% of topical propranolol could significantly reduce VEGF and IGF-1 upregulation following hypoxia and profoundly reduced HIF-1α accumulation and STAT3 phosphorylation, leading to reduce RNV in the super fi cial, but not the deep, vascular plexus in OIR[181]. Thus, it is hopeful that the agents of adrenergic system may be used in the early stage of ROP in the future.

Renin-angiotensin-aldosterone System and Retinopathy of Prematurity It has been reported[182] that renin-angiotensinaldosterone system (RAS) components are localized to blood vessels and cells in the retinal ganglion cell (RGC) layer of newborn rats, where they may stimulate the growing vasculature to extend into the peripheral retina and increase the vascular density in the periphery. It has also been reported[183-184] that Ang II via its type 1 and type 2 receptors regulates the generation of ROS by NADPH oxidase, which is crucial for the vasculature, including retinal angiogenesis.In addition, aldosterone might be through reducing glucose-6-phosphate dehydrogenase to exacerbate angiogenesis in early OIR, whereas an antagonist of the mineralocorticoid receptor(MR) reversed this effect of aldosterone[185]. Furthermore, the fact that the 32-gestational-week twin girls who were exposed to blockage of the RAS during gestation and received very little additional oxygen following birth showed severely reduced retinal vasculature and developed severe ROP later[186],suggested that RAS might play an important role in ROP development. Whether RAS is crucial for retinal vasculature and early administration Ang II is beneficial for the prophylaxis of ROP need to be further investigated.

EARLY NUTRITIONAL SUPPLEMENT AND RETINOPATHY OF PREMATURITY

Vitamin A and Early Retinopathy of Prematurity Vitamin A refers to a group of compounds, including retinol,retinaldehyde, and retinoic acid. In the retina, reversible oxidation of vitamin A produces retinaldehyde, which is an essential constituent of the visual pigment rhodopsin[187]. It has been reported that in hyperoxia, retinoic acid promoted VEGF expression in OIR retina, supported retinal vascular development and counteracted vaso-obliteration in OIR mice[188]. Premature infants were prone to subclinical vitamin A de fi ciencies during the first week of life, and these deficiencies could be treated with adequate enteral feeding and routine multivitamin supplementation, suggesting that a high dose of vitamin A was not necessary for healthy premature infants[189-190]. Mactier et al[191-192] showed that early high-dose intramuscular vitamin A supplementation in infants at risk for ROP improved retinal function at 36wk of postmenstrual age.Thus, early vitamin A supplementation may be beneficial to premature infants at risk for ROP. However, the optimal dose and most appropriate route of administration of vitamin A in preterm infants needs to be determined in the future.

Estrogen and Early Retinopathy of Prematurity The role of estrogen in ROP has been explored, particularly, with regard to angiogenesis[148]. The serum levels of estradiol were low in premature infants, which suggested that estradiol might play a role in ROP. An inhibiting effect of 17-alpha- and 17-betaestradiol on RNV in OIR was reported[149-153]. However, due to the adverse effects, it is unfeasible to treat premature infants with estradiol. In addition, since 17-beta- and 17-alphaestradiol are not highly selective for the estrogen receptor and can combine with the alpha, beta and other estrogen receptors[154-155], studies aim to explore which estrogen receptor plays the primary role in the prophylaxis of ROP are necessary.

目前,高职院校的教师到企业挂职缺乏有效的组织管理、考勤考核以及相关的保障措施。大部分教师到企业挂职均是由教师自行寻找挂职单位,并且由教师个人决定在企业挂职的工作岗位、工作内容以及学习方式等,学校与企业之间存在着缺乏沟通的机制。因缺乏有效的管理机制,学校对于教师挂职锻炼的效果及考核情况等的监管并不全面,而且企业反馈的渠道也比较单一,对于教师在挂职期间的有效管理和效果监控还是存在着一定的问题。

Iron Supplementation and Early Retinopathy of Prematurity Iron is an essential micronutrient that plays an important role in cellular function. It has been found that premature infants showed reduced iron stores, compared with full-term infants[207]. Early iron supplementation in preterm very LBW infants could improve serum ferritin and hemoglobin levels but has less effect on the incidences of ROP[208-209]. Whether iron supplementation benefits to preterm or LBW infants with regard to the development of the retinal vasculature needs to be further investigated.

Polyunsaturated Fatty Acids and Early Retinopathy of Prematurity Omega-3 and omega-6 PUFAs are essential components of cell membrane phospholipids and substrates of various enzymes. It has been reported that omega-3 PUFAs decrease the avascular area of the OIR retina by increasing vessel regrowth under hyperoxic conditions, thereby reducing the hypoxic stimulus of neovascularization[210]. Bioactive omega-3-PUFA-derived mediators also potently protected against neovascularization by suppressing tumor necrosis factor-alpha[211]. Sapieha et al[212] found that 5-lipoxygenase(LOX) played a pivotal role in the protection of dietary omega-3 PUFAs against OIR and that COX inhibitors,might be used without losing the beneficial effects of dietary omega-3 PUFAs. Recently study suggested that, in addition to anti-angiogenic metabolites of COX and LOX, cytochrome P450 epoxygenases (CYP2C8) metabolized omega-3 PUFAs and produced bioactive epoxides that were inactivated by soluble epoxide hydrolase (sEH) into transdihydrodiols[212].In an OIR model, CYP2C8 is upregulated, whereas sEH is suppressed, resulting in an increased retinal epoxide-to-diol ratio. Overexpression of CYP2C8 or sEH in mice does not affect normal retinal vascular development. The proangiogenic role of CYP2C8 in the retina on both omega-3 LCPUFAs and omega-6 LCPUFAs and the anti-angiogenic role of sEH on omega-3 LCPUFA metabolism could influence RNV[212].And recently, It has been reported that early administration parenteral omega-3 FAs in the form of fi sh-oil lipid emulsions markedly reduced the incidence of severe ROP or need for laser therapy in preterm infants[213]. Thus, early supplementation of omega-3 LCPUFAs and omega-6 LCPUFAs may contribute to retinal vasculature of premature infants and reduce the risk of ROP.

Sepsis and Early Retinopathy of Prematurity Sepsis is a potentially fatal whole-body inflammatory state that is caused by severe infection. Early-onset sepsis was associated with severe ROP, and sepsis also associated with the onset of posterior ROP[214-216]. In addition, perinatal inflammatory stress induced a significant increase in retinal vascular density,as well as a pronounced increase in activated microglial cells in RGC layer and in the outer plexiform layer immediately prior to their vascularization[217]. At maturity, perinatal in fl ammatory stress led to depleted retinal vascular beds and significantly decreased retinal function, resulting in abnormal retinal vascular development and increased vessel anastomosis and, finally, impairment in retinal function associated with microglial activation[217]. Therefore, management of sepsis may be beneficialin reducing the incidence and severity of ROP.

Hypercapnia and Early Retinopathy of Prematurity Hypercapnia is generally defined as an abnormally high level of CO2 (e.g. more than 45 mm Hg) in the arterial blood.Permissive hypercapnia has been recognized, as a method to reduce lung injury and other adverse effects and to improve survival in preterm neonates[218]. However, hypercapnia might be a risk factor for ROP in clinical setting[219]. It has been reported that increase of CO2 levels was associated with retardation of normal retinal vascular development and increase of peripheral avascular area in neonatal OIR rats, which is a critical step preceding RNV. The following findings suggest that hypercapnia may hinder efficient neovascularization and contribute to ROP[220-223]. Hypercapnia is able to induce an early increase in endothelial NO synthase and RNS. In vivo RNS is associated with retinal vaso-obliteration and leads to the nitration of arachidonic acids (AAs) into trans-AAs (TAAs).TAAs can act as mediators of nitrative stress by: 1) causing microvascular degeneration through inducing the expression of the antiangiogenic factor thrombospondin-1, which is associated with astrocyte impairment and endothelial cell death; 2) downregulation of the proangiogenic prostaglandin E2 receptor EP3. Thus, management of hypercapnia may ameliorate early ROP.

Early Light Reduction and Early Retinopathy of Prematurity Recent study suggested that continuous light radiation caused a time-dependent decrease in RGC-5 response and resulted in photo-damage within 10h due to the depletion of adenosine 5’-triphosphate and an increase in ROS levels,similar to photo-damage in vivo[224]. Thus, it is reasonable to postulate that an early reduction in light exposure in premature infants might decrease the incidence of acute ROP. However,this hypothesis was not supported by studies that decreased exposure of the retina to light in premature infants did not reduce the incidence of ROP[225]. Whether light contributes to ROP needs to be further investigated.

北京兰台律师事务所税法律师武嘉认为,逃避缴纳税款的纳税人需要承担行政责任,补交税款、滞纳金和罚款。如果在刑事立案前如数补交税款、滞纳金、罚款的,可能不予追究刑事责任,结果或许没有想象那么坏。

Stem Cells and Early Retinopathy of Prematurity Stem cells (SCs) are pluripotent cells with self-renewing capability.Recent studies have shown that SCs play important roles in RNV[226-227], which suggests a role of SCs in ROP. It was found that a deviation in the functional bioactivities of bone marrowderived EPCs (BM-derived EPCs) enabled intact vascular development under abnormal oxygen dynamics[228]. EPCs were increased significantly in the peripheral blood and bone marrow of mice with OIR and a decrease in circulating EPCs might arrest vessel growth during normal retinal development in OIR rats[229-230]. Early EPCs and very small embryonic-like SCs were also significantly increased in preterm infants with ROP, which suggested that EPCs and circulating SCs may play a role in ROP[231]. In addition, BM-derived monocyte lineage cells (BM-MLCs) could differentiate into endothelial cell(EC)-like cells and function as EC progenitors that acquire the ability to adhere to injured endothelium in a MCP-1-dependent manner[232]. The reduction of BM-MLCs infiltrating into the OIR retina is associated with an increase in the avascular area and preretinal neovascular tufts, which suggests that recruitment of BM-MLCs to the hypoxic retina may be used to promote intraretinal revascularization, thereby preventing RNV[233]. Furthermore, it has been reported that a number of adult BM-derived myeloid progenitor cells could migrate to avascular regions of the OIR retina and then differentiated into microglia to facilitate normalization of the vasculature[234].These studies suggest a role of SCs in regulating vascular regeneration in ROP, which also provides a rationale for stem cell therapy in ROP in the future.

Gene Expression Profiles and Retinal Proteome Changes During Early OIR In the past years, studies are also focusing on analysing gene expression profiles and retinal proteomic alterations during OIR. It has been shown that the expression of 83 genes, which are associated with development, metabolism,transport, stress response, cell adhesion, in fl ammation or vision,are significantly altered in hyperoxic P12 retinas. In particular,genes associated with retinal growth and vascular development,such as Pdgfb and Robo4, were downregulated[235]. In a mouse OIR model, enriched genes associated with cytoskeletal formation were identified at P8, whereas the enriched genes associated with various pathological processes, including the modulation of RNV were identified at P13[236]. Furthermore,in the iTRAQ study, upregulation of 25 proteins and downregulation of 14 proteins were identified in OIR retinas at P12 compared with the control retina[237]. These genes and proteins identified in OIR may be potential novel therapeutic targets for treating ROP.

CONCLUSION

Current treatments of ROP are focusing on RNV, which have caused new problems and debates over the current management protocols[238]. Since it takes approximately 4 to 6wk from birth to the RNV of ROP, the optimal preventive treatments for ROP should also be applied during these periods[239].In Figure 1, we summarized the molecular regulators and signalling pathways that may contribute to the pathogenesis prior to RNV in OIR and in premature infants with ROP, which includes signalling regulators of HIF-1α, VEGF, IGF and Norrin, oxidative stress, neuroretina and its communication with the retinal vasculature, neuropeptides, endocrine hormones, early nutrition supplementation, sepsis and SCs as well as blood SaO2 and hypercapnia. We also discussed the preclinical and clinical studies on preventive treatment of early OIR and ROP, which includes: 1) supplementation of HIF-1α,Norrin, IGF-I or IGFBP-3, somatostatin, NPY or Epo, vitamin A, iron, PUFAs or other nutrients; 2) administration of VEGF,VEGFR-1 agonist or VEGFR-2 antagonist, glucocorticoids,highly selective estrogen receptor agonist, ghrelin, insulin,Ang-1, Ang-2 inhibitor, β-AR antagonist or Ang-II; 3)suppression of oxidative stress; 4) protection of neural cells in retina and decrease of Sema3A and N-CAM; 5) mitigation and even elimination of sepsis; 6) management of hypercapnia;7) decrease of light exposure; 8) transfusion of stem cells; 9)increase of the expression of Pdgfb, Robo4 and cytoskeletal formation genes. We wish to not only highlight the importance of the mechanisms or signalling pathways in regulating early stage of ROP but also provide guidance for exploring novel treatments for early phase OIR and ROP prior to RNV in the future.

Figure 1 The factors related to early ROP We summarize the molecular regulators and signalling pathways that may contribute to the pathogenesis in OIR and ROP, which includes HIF-1α, VEGF,IGF and Norrin, oxidative stress, neuroretina and its communication with the retinal vasculature, neuropeptides, endocrine hormones, early nutrition supplementation, sepsis and stem cells as well as blood SaO2 and hypercapnia. Targeting these molecular regulators and pathways may reduce the risk and severity of ROP.

ACKNOWLEDGEMENTS

Foundations: Supported by the National Natural Science Foundation of China (No.81570873); the Science and Technology Department of Shaanxi Province (No.2015JM8481).

Conflicts of Interest: Zhang HB, None; Wang XD, None;Xu K, None; Li XG, None.

REFERENCES

1 Zin A, Gole GA. Retinopathy of prematurity-incidence today. Clin Perinatol 2013;40(2):185-200.

2 C hang JY, Lee KS, Hahn WH, Chung SH, Choi YS, Shim KS, Bae CW. Decreasing trends of neonatal and infant mortality rates in Korea:compared with Japan, USA, and OECD nations. J Korean Med Sci 2011;26(9):1115-1123.

3 C ourtright P, Hutchinson AK, Lewallen S. Visual impairment in children in middle- and lower-income countries. Arch Dis Child 2011;96(12):1129-1134.

4 Z epeda-Romero LC, Barrera-de-Leon JC, Camacho-Choza C, Gonzalez Bernal C, Camarena-Garcia E, Diaz-Alatorre C, Gutierrez-Padilla JA, Gilbert C. Retinopathy of prematurity as a major cause of severe visual impairment and blindness in children in schools for the blind in Guadalajara city, Mexico. Br J Ophthalmol 2011;95(11):1502-1505.

5 A ugestad LB, Klingenberg O, Fosse P. Braille use among Norwegian children from 1967 to 2007: trends in the underlying causes. Acta Ophthalmol 2012;90(5):428-434.

6 L au C, Ambalavanan N, Chakraborty H, Wingate MS, Carlo WA.Extremely low birth weight and infant mortality rates in the United States.Pediatrics 2013;131(5):855-860.

7 Chen J, Smith LE. Retinopathy of prematurity. Angiogenesis 2007;10(2):133-140.

8 Lutty GA, McLeod DS. Retinal vascular development and oxygeninduced retinopathy: a role for adenosine. Prog Retin Eye Res 2003;22(1):95-111.

9 Multicenter trial of cryotherapy for retinopathy of prematurity:ophthalmological outcomes at 10 years. Arch Ophthalmol 2001;119(8):1110-1118.

10 Mintz-Hittner HA, Kennedy KA, Chuang AZ. efficacy of intravitreal bevacizumab for stage 3+ retinopathy of prematurity. N Engl J Med 2011;364(7):603-615.

11 Quinn GE, Dobson V, Davitt BV, Hardy RJ, Tung B, Pedroza C, Good WV. Progression of myopia and high myopia in the early treatment for retinopathy of prematurity study: fi ndings to 3 years of age. Ophthalmology 2008;115(6):1058-1064.e1.

12 Davitt BV, Dobson V, Quinn GE, Hardy RJ, Tung B, Good WV.Astigmatism in the early treatment for retinopathy of prematurity study:fi ndings to 3 years of age. Ophthalmology 2009;116(2):332-339.

13 Yang CS, Wang AG, Sung CS, Hsu WM, Lee FL, Lee SM. Long-term visual outcomes of laser-treated threshold retinopathy of prematurity: a study of refractive status at 7 years. Eye (Lond) 2010;24(1):14-20.

14 Davitt BV, Quinn GE, Wallace DK, Dobson V, Hardy RJ, Tung B,Lai D, Good WV. Astigmatism progression in the early treatment for retinopathy of prematurity study to 6 years of age. Ophthalmology 2011;118(12):2326-2329.

15 Repka MX. Outcome of eyes developing retinal detachment during the early treatment for retinopathy of prematurity study. Arch Ophthalmol 2011;129(9):1175.

16 Bremer DL, Rogers DL, Good WV, Tung B, Hardy RJ, Fellows R. Glaucoma in the Early Treatment for Retinopathy of Prematurity(ETROP) study. J AAPOS 2012;16(5):449-452.

17 Houston SK, Wykoff CC, Berrocal AM, Hess DJ, Murray TG. Laser treatment for retinopathy of prematurity. Lasers Med Sci 2013;28(2):683-692.

18 Clark D, Mandal K. Treatment of retinopathy of prematurity. Early Hum Dev 2008;84(2):95-99.

19 Early Treatment For Retinopathy Of Prematurity Cooperative Group.Revised indications for the treatment of retinopathy of prematurity: results of the early treatment for retinopathy of prematurity randomized trial.Arch Ophthalmol 2003;121(12):1684-1694.

20 Good WV. Final results of the early treatment for retinopathy of prematurity (ETROP) randomized trial. Trans Am Ophthalmol Soc 2004;102:233-248.

21 Good WV. The early treatment for retinopathy of prematurity study:structural fi ndings at age 2 years. Br J Ophthalmol 2006;90(11):1378-1382.

22 Axer-Siegel R, Maharshak I, Snir M, Friling R, Ehrlich R, Sherf I,Shalev B, Sirota L, Weinberger D. Diode laser treatment of retinopathy of prematurity: anatomical and refractive outcomes. Retina 2008;28(6):839-846.

23 Soh Y, Fujino T, Hatsukawa Y. Progression and timing of treatment of zone I retinopathy of prematurity. Am J Ophthalmol 2008;146(3):369-374.

24 Good WV, Hardy RJ, Dobson V, Palmer EA, Phelps DL, Tung B, Redford M. Final visual acuity results in the early treatment for retinopathy of prematurity study. Arch Ophthalmol 2010;128(6):663-671.

25 Dobson V, Quinn GE, Summers CG, Hardy RJ, Tung B, Good WV.Grating visual acuity results in the early treatment for retinopathy of prematurity study. Arch Ophthalmol 2011;129(7):840-846.

26 Owen LA, Hartnett ME. Current concepts of oxygen management in retinopathy of prematurity. J Ophthalmic Vis Res 2014;9(1):94-100.

27 Ryan H. Retrolental fibroplasia; a clinicopathologic study. Am J Ophthalmol 1952;35(3):329-342.

28 Locke JC. Retrolental fibroplasia definitive role of oxygen administration in its etiology. AMA Arch Ophthalmol 1954;51(1):73-79.

29 Smith LE, Wesolowski E, McLellan A, Kostyk SK, D’Amato R,Sullivan R, D’Amore PA. Oxygen-induced retinopathy in the mouse.Invest Ophthalmol Vis Sci 1994;35(1):101-111.

30 Hartnett ME. The effects of oxygen stresses on the development of features of severe retinopathy of prematurity: knowledge from the 50/10 OIR model. Doc Ophthalmol 2010;120(1):25-39.

31 Lange C, Ehlken C, Stahl A, Martin G, Hansen L, Agostini HT.Kinetics of retinal vaso-obliteration and neovascularisation in the oxygen-induced retinopathy (OIR) mouse model. Graefes Arch Clin Exp Ophthalmol 2009;247(9):1205-1211.

32 Stenson BJ, Tarnow-Mordi WO, Darlow BA, et al. Oxygen saturation and outcomes in preterm infants. N Engl J Med 2013;368(22):2094-2104.

33 Stenson BJ. Oxygen targets for preterm infants. Neonatology 2013;103(4):341-345.

34 Saugstad OD. Optimal oxygenation at birth and in the neonatal period.Neonatology 2007;91(4):319-322.

35 Wright KW, Sami D, Thompson L, Ramanathan R, Joseph R,Farzavandi S. A physiologic reduced oxygen protocol decreases the incidence of threshold retinopathy of prematurity. Trans Am Ophthalmol Soc 2006;104:78-84.

36 Tlucek PS, Corff KE, Bright BC, Bedwell SM, Sekar KC, Siatkowski RM. Effect of decreasing target oxygen saturation on retinopathy of prematurity. J AAPOS 2010;14(5):406-411.

37 Sears JE, Pietz J, Sonnie C, Dolcini D, Hoppe G. A change in oxygen supplementation can decrease the incidence of retinopathy of prematurity.Ophthalmology 2009;116(3):513-518.

38 Chen ML, Guo L, Smith LE, Dammann CE, Dammann O. High or low oxygen saturation and severe retinopathy of prematurity: a meta-analysis.Pediatrics 2010;125(6):e1483-e1492.

39 Schmidt B, Whyte RK, Asztalos EV, Moddemann D, Poets C, Rabi Y, Solimano A, Roberts RS. Effects of targeting higher vs lower arterial oxygen saturations on death or disability in extremely preterm infants: a randomized clinical trial. JAMA 2013;309(20):2111-2120.

40 Tarnow-Mordi W, Stenson B, Kirby A, et al. Outcomes of two trials of oxygen-saturation targets in preterm infants. N Engl J Med 2016;374(8):749-760.

41 Tlucek PS, Grace SF, Anderson MP, Siatkowski RM. Effect of the oxygen saturation target on clinical characteristics of early- versus lateonset retinopathy of prematurity. J AAPOS 2 012;16(1):70-74.

42 Zhang W, Yokota H, Xu Z, Narayanan SP, Yancey L, Yoshida A,Marcus DM, Caldwell RW, Caldwell RB, Brooks SE. Hyperoxia therapy of pre-proliferative is chemic retinopathy in a mouse model. Invest Ophthalmol Vis Sci 2011;52(9):6384-6395.

43 Wang GL, Jiang BH, Rue EA, Semenza GL. Hypoxia-inducible factor 1 is a basic-helix-loop-helix-PAS heterodimer regulated by cellular O2 tension. Proc Natl Acad Sci U S A 1995;92(12):5510-5514.

44 Jiang BH, Rue E, Wang GL, Roe R, Semenza GL. Dimerization, DNA binding, and transactivation properties of hypoxia-inducible factor 1. J Biol Chem 1996;271(30):17771-17778.

45 Semenza GL, Jiang BH, Leung SW, Passantino R, Concordet JP, Maire P, Giallongo A. Hypoxia response elements in the aldolase A, enolase 1,and lactate dehydrogenase A gene promoters contain essential binding sites for hypoxia-inducible factor 1. J Biol Chem 1996;271(51):32529-32537.

46 Goda N, Dozier SJ, Johnson RS. HIF-1 in cell cycle regulation,apoptosis, and tumor progression. Antioxid Redox Signal 2003;5(4):467-473.

47 Semenza GL. Hypoxia-inducible factors in physiology and medicine.Cell 2012;148(3):399-408.

48 Metzen E, Berchner-Pfannschmidt U, Stengel P, Marxsen JH, Stolze I, Klinger M, Huang WQ, Wotzlaw C, Hellwig-Bürgel T, Jelkmann W,Acker H, Fandrey J. Intracellular localisation of human HIF-1 alpha hydroxylases: implications for oxygen sensing. J Cell Sci 2003;116(Pt 7):1319-1326.

49 K aelin WG Jr, Ratcliffe PJ. Oxygen sensing by metazoans: the central role of the HIF hydroxylase pathway. Mol Cell 2008;30(4):393-402.

50 C howdhury R, McDonough MA, Mecinović J, Loenarz C, Flashman E, Hewitson KS, Domene C, Scho fi eld CJ. Structural basis for binding of hypoxia-inducible factor to the oxygen-sensing prolyl hydroxylases.Structure 2009;17(7):981-989.

51 Hua ng LE, Gu J, Schau M, Bunn HF. Regulation of hypoxiainducible factor 1alpha is mediated by an O2-dependent degradation domain via the ubiquitin- proteasome pathway. Proc Natl Acad Sci U S A 1998;95(14):7987-7992.

52 Iva n M, Kondo K, Yang H, Kim W, Valiando J, Ohh M, Salic A,Asara JM, Lane WS, Kaelin WG Jr. HIFalpha targeted for VHL-mediated destruction by proline hydroxylation: implications for O2 sensing. Science 2001;292(5516):464-468.

53 Jaa kkola P, Mole DR, Tian YM, Wilson MI, Gielbert J, Gaskell SJ, von Kriegsheim A, Hebestreit HF, Mukherji M, Scho fi eld CJ, Maxwell PH,Pugh CW, Ratcliffe PJ. Targeting of HIF-alpha to the von Hippel-Lindau ubiquitylation complex by O2-regulated prolyl hydroxylation. Science 2001;292(5516):468-472.

54 Bru ick RK, McKnight SL. A conserved family of prolyl-4-hydroxylases that modify HIF. Science 2001;294(5545):1337-1340.

55 Eps tein AC, Gleadle JM, McNeill LA, et al. C elegans EGL-9 and mammalian homologs de fi ne a family of dioxygenases that regulate HIF by prolyl hydroxylation. Cell 2001;107(1):43-54.

56 Hua ng H, Van de Veire S, Dalal M, Parlier R, Semba RD, Carmeliet P,Vinores SA. Reduced retinal neovascularization, vascular permeability,and apoptosis in ischemic retinopathy in the absence of prolyl hydroxylase-1 due to the prevention of hyperoxia-induced vascular obliteration. Invest Ophthalmol Vis Sci 2011;52(10):7565-7573.

57 Hoppe G, Lee TJ, Yoon S, Yu M, Peachey NS, Rayborn M, Zutel MJ, Trichonas G, Au J, Sears JE. Inducing a visceral organ to protect a peripheral capillary bed: stabilizing hepatic HIF-1α prevents oxygeninduced retinopathy. Am J Pathol 2014;184(6):1890-1899.

58 Wang H, Chen X. Effects of a Rho kinase inhibitor on the sequential expression of ICAM-1, HIF-1α, Bcl-2 and caspase-3 in the retina of rats with oxygen-induced retinopathy. Int J Mol Med 2013;32(2):457-463.

59 Sears JE, Hoppe G, Ebrahem Q, Anand-Apte B. Prolyl hydroxylase inhibition during hyperoxia prevents oxygen-induced retinopathy. Proc Natl Acad Sci U S A 2008;105(50):19898-19903.

60 Tricho nas G, Lee TJ, Hoppe G, Au J, Sears JE. Prolyl hydroxylase inhibition during hyperoxia prevents oxygen-induced retinopathy in the rat 50/10 model. Invest Ophthalmol Vis Sci 2013;54(7):4919-4926.

61 Forsyt he JA, Jiang BH, Iyer NV, Agani F, Leung SW, Koos RD,Semenza GL. Activation of vascular endothelial growth factor gene transcription by hypoxia-inducible factor 1. Mol Cell Biol 1996;16(9):4604-4613.

62 Ferrar a N, Gerber HP, LeCouter J. The biology of VEGF and its receptors. Nat Med 2003;9(6):669-676.

63 Fong G H, Rossant J, Gertsenstein M, Breitman ML. Role of the Flt-1 receptor tyrosine kinase in regulating the assembly of vascular endothelium. Nature 1995;376(6535):66-70.

64 Koch S, Claesson-Welsh L. Signal transduction by vascular endothelial growth factor receptors. Cold Spring Harb Perspect Med 2012;2(7):a006502.

65 Millau er B, Wizigmann-Voos S, Schnürch H, Martinez R, Møller NP, Risau W, Ullrich A. High af fi nity VEGF binding and developmental expression suggest Flk-1 as a major regulator of vasculogenesis and angiogenesis. Cell 1993;72(6):835-846.

66 Millauer B, Shawver LK, Plate KH, Risau W, Ullrich A. Glioblastoma growth inhibited in vivo by a dominant-negative Flk-1 mutant. Nature 1994,367(6463):576-579.

67 Suzuma K, Takagi H, Otani A, Suzuma I, Honda Y. Increased expression of KDR/Flk-1 (VEGFR-2) in murine model of is chemiainduced retinal neovascularization. Microvasc Res 1998;56(3):183-191.68 Shih SC, Ju M, Liu N; Smith LE. Selective stimulation of VEGFR-1 prevents oxygen-induced retinal vascular degeneration in retinopathy of prematurity. J Clin Invest 2003;112(1):50-57.

69 Yenice O, Cerman E, Ashour A, Firat R, Haklar G, Sirikci O, Akman I, Kazokoglu H. Serum erythropoietin, insulin-like growth factor 1, and vascular endothelial growth factor in etiopathogenesis of retinopathy of prematurity. Ophthalmic Surg Lasers Imaging Retina 2013;44(6):549-554.

70 Kwinta P, Bik-Multanowski M, Mitkowska Z, Tomasik T, Pietrzyk JJ.The clinical role of vascular endothelial growth factor (VEGF) system in the pathogenesis of retinopathy of prematurity. Graefes Arch Clin Exp Ophthalmol 2008;246(10):1467-1475.

71 Geisen P, Peterson LJ, Martiniuk D, Uppal A, Saito Y, Hartnett ME.Neutralizing antibody to VEGF reduces intravitreous neovascularization and may not interfere with ongoing intraretinal vascularization in a rat model of retinopathy of prematurity. Mol Vis 2008;14:345-357.

72 Ohlmann A, Tamm ER. Norrin: molecular and functional properties of an angiogenic and neuroprotective growth factor. Prog Retin Eye Res 2012;31(3):243-257.

73 Lee H, J o DH, Kim JH, Kim JH. Norrin expression in endothelial cells in the developing mouse retina. Acta Histochem 2013;115(5):447-451.

74 Ohlmann A, Seitz R, Braunger B, Seitz D, Bösl MR, Tamm ER. Norrin promotes vascular regrowth after oxygen-induced retinal vessel loss and suppresses retinopathy in mice. J Neurosci 2010;30(1):183-193.

75 Wang Y, R attner A, Zhou Y, Williams J, Smallwood PM, Nathans J.Norrin/Frizzled4 signaling in retinal vascular development and blood brain barrier plasticity. Cell 2012;151(6):1332-1344.

76 Paes KT, Wang E, Henze K, Vogel P, Read R, Suwanichkul A,Kirkpatrick LL, Potter D, Newhouse MM, Rice DS. Frizzled 4 is required for retinal angiogenesis and maintenance of the blood-retina barrier. Invest Ophthalmol Vis Sci 2011;52(9):6452-6461.

77 Jones JI, Clemmons DR. Insulin-like growth factors and their binding proteins: biological actions. Endocr Rev 1995;16(1):3-34.

78 Khandwala HM, McCutcheon IE, Flyvbjerg A, Friend KE. The effects of insulin-like growth factors on tumorigenesis and neoplastic growth.Endocr Rev 2000;21(3):215-244.

79 Firth SM, Baxter RC. Cellular actions of the insulin-like growth factor binding proteins. Endocr Rev 2002;23(6):824-854.

80 Duan C, Xu Q. Roles of insulin-like growth factor (IGF) binding proteins in regulating IGF actions. Gen Comp Endocrinol 2005;142(1-2):44-52.

81 Lofqvist C, Willett KL, Aspegren O, Smith AC, Aderman CM, Connor KM, Chen J, Hellstrom A, Smith LE. Quanti fi cation and localization of the IGF/insulin system expression in retinal blood vessels and neurons during oxygen-induced retinopathy in mice. Invest Ophthalmol Vis Sci 2009;50(4):1831-1837.

82 Vanhaesebrouck S, Daniëls H, Moons L, Vanhole C, Carmeliet P, De Zegher F. Oxygen-induced retinopathy in mice: ampli fi cation by neonatal IGF-I deficit and attenuation by IGF-I administration. Pediatr Res 2009;65(3):307-310.

8 3 Brock RS, Gebrekristos BH, Kuniyoshi KM, Modanlou HD, Falcao MC, Beharry KD. Biomolecular effects of JB1 (an IGF-I peptide analog) in a rat model of oxygen-induced retinopathy. Pediatr Res 2011;69(2):135-141.8 4 Hellström A, Engström E, Hård AL, Albertsson-Wikland K, Carlsson B, Niklasson A, Löfqvist C, Svensson E, Holm S, Ewald U, Holmström G, Smith LE. Postnatal serum insulin-like growth factor I de fi ciency is associated with retinopathy of prematurity and other complications of premature birth. Pediatrics 2003;112(5):1016-1020.

85 Smi th LE. IGF-1 and retinopathy of prematurity in the preterm infant.Biol Neonate 2005;88(3):237-244.

86 Cek mez F, Pirgon O, Aydemir G, Dundar B, Cekmez Y, Karaoglu A,Fidanc K, Tunc T, Aydinoz S, Karademir F, Süleymanoglu S. Correlation between cord blood apelin and IGF-1 levels in retinopathy of prematurity.Biomark Med 2012;6(6):821-825.

87 Pér ez-Muñuzuri A, Fernández-Lorenzo JR, Couce-Pico ML, Blanco-Teijeiro MJ, Fraga-Bermúdez JM. Serum levels of IGF1 are a useful predictor of retinopathy of prematurity. Acta Paediatr 2010;99(4):519-525.

88 Kielczew ski JL, Jarajapu YP, McFarland EL, Cai J, Afzal A, Li Calzi S, Chang KH, Lydic T, Shaw LC, Busik J, Hughes J, Cardounel AJ,Wilson K, Lyons TJ, Boulton ME, Mames RN, Chan-Ling T, Grant MB.Insulin-like growth factor binding protein-3 mediates vascular repair by enhancing nitric oxide generation. Circ Res 2009;105(9):897-905.

89 Kielczew ski JL, Hu P, Shaw LC, Li Calzi S, Mames RN, Gardiner TA,McFarland E, Chan-Ling T, Grant MB. Novel protective properties of IGFBP-3 result in enhanced pericyte ensheathment, reduced microglial activation, increased microglial apoptosis, and neuronal protection after ischemic retinal injury. Am J Pathol 2011;178(4):1517-1528.

90 Hansen-Pupp I, Engström E, Niklasson A, Berg AC, Fellman V,Löfqvist C, Hellström A, Ley D. Fresh-frozen plasma as a source of exogenous insulin-like growth factor-I in the extremely preterm infant. J Clin Endocrinol Metab 2009;94(2):477-482.

91 Dani C, Po g gi C, Bresci C, Corsini I, Frosini S, Pratesi S. Early freshfrozen plasma transfusion decreases the risk of retinopathy of prematurity.Transfusion 2014;54(4):1002-1007.

92 Ley D, Hans en-Pupp I, Niklasson A, Domellöf M, Friberg LE, Borg J,Löfqvist C, Hellgren G, Smith LE, Hård AL, Hellström A. Longitudinal infusion of a complex of insulin-like growth factor-I and IGF-binding protein-3 in fi ve preterm infants: pharmacokinetics and short-term safety.Pediatr Res 2013;73(1):68-74.

93 Hellgren G, Ha n W, Wang X, Löfqvist C, Hagberg H, Mallard C,Hellström A. Safety aspects of longitudinal administration of IGF-I/IGFBP-3 complex in neonatal mice. Growth Horm IGF Res 2011;21(4):205-211.

94 Zhou Y, Yan H, Gu o M, Zhu J, Xiao Q, Zhang L. Reactive oxygen species in vascular formation and development. Oxid Med Cell Longev 2013;2013:374963.

95 Kim YW, Byzova TV. Oxidative stress in angiogenesis and vascular disease. Blood 2014;123(5):625-631.

96 Griendling KK, So rescu D, Ushio-Fukai M. NAD(P)H oxidase: role in cardiovascular biology and disease. Circ Res 2000;86(5):494-501.

97 Hernanz R, Brione s AM, Salaices M, Alonso MJ. New roles for old pathways? A circuitous relationship between reactive oxygen species and cyclo-oxygenase in hypertension. Clin Sci 2014;126(2):111-121.

98 Nandgaonkar BN, Rotschild T, Yu K, Higgins RD. Indomethacin improves oxygen-induced retinopathy in the mouse. Pediatr Res 1999;46(2):184-188.

99 Sharma J, Barr SM, Geng Y, Yun Y, Higgins RD. Ibuprofen improves oxygen-induced retinopathy in a mouse model. Curr Eye Res 2003;27(5):309-314.

100 Beharry KD, Moda nlou HD, Hasan J, Gharraee Z, Abad-Santos P,Sills JH, Jan A, Nageotte S, Aranda JV. Comparative effects of early postnatal ibuprofen and indomethacin on VEGF, IGF-I, and GH during rat ocular development. Invest Ophthalmol Vis Sci 2006;47(7):3036-3043.

101 Hasan J, Beharry KD, Gharraee Z, Stavitsky Y, Abad-Santos P, Abad-Santos M, Aranda JV, Modanlou HD. Early postnatal ibuprofen and indomethacin effects in suckling and weanling rat kidneys. Prostaglandins Other Lipid Mediat 2008;85(3):81-88.

102 Gebrekristos B, Beharry KD, Brock RS, Kuniyoshi K, Abad-Santos P,Abad-Santos M, Aranda JV, Modanlou HD. Hormonal in fl uences of early postnatal indomethacin and ibuprofen in neonatal rats. Growth Horm IGF Res 2010;20(1):31-38.

103 Saito Y, Geisen P, Uppal A, Hartnett ME. Inhibition of NAD(P)H oxidase reduces apoptosis and avascular retina in an animal model of retinopathy of prematurity. Mol Vis 2007;13:840-853.

104 Penn JS. Oxygen- induced retinopathy in the rat: possible contribution of peroxidation reactions. Doc Ophthalmol 1990;74(3):179-186.

105 Penn JS, Thum LA, Naash MI. Oxygen-induced retinopathy in the rat. Vitamins C and E as potential therapies. Invest Ophthalmol Vis Sci 1992;33(6):1836-1845.

106 Abdelsaid MA, Pi llai BA, Matragoon S, Prakash R, Al-Shabrawey M, El-Remessy AB. Early intervention of tyrosine nitration prevents vasoobliteration and neovascularization in ischemic retinopathy. J Pharmacol Exp Ther 2010;332(1):125-134.

107 Uno K, Prow TW, Bhutto IA, Yerrapureddy A, McLeod DS, Yamamoto M, Reddy SP, Lutty GA. Role of Nrf2 in retinal vascular development and the vaso-obliterative phase of oxygen-induced retinopathy. Exp Eye Res 2010;90(4):493-500.

108 Tan SM, Stefanov ic N, Tan G, Wilkinson-Berka JL, de Haan JB.Lack of the antioxidant glutathione peroxidase-1 (GPx1) exacerbates retinopathy of prematurity in mice. Invest Ophthalmol Vis Sci 2013;54(1):555-562.

109 Deliyanti D, Lee JY, Petratos S, Meyer CJ, Ward KW, Wilkinson-Berka JL, de Haan JB. A potent Nrf2 activator, dh404, bolsters antioxidant capacity in glial cells and attenuates ischaemic retinopathy. Clin Sci 2016;130(15):1375-1387.

110 Suwanpradid J, Rojas M, Behzadian MA, Caldwell RW, Caldwell RB. Arginase 2 de fi ciency prevents oxidative stress and limits hyperoxiainduced retinal vascular degeneration. PLoS One 2014;9(11):e110604.

111 Kretzer FL, Meh ta RS, Johnson AT, Hunter DG, Brown ES, Hittner HM. Vitamin E protects against retinopathy of prematurity through action on spindle cells. Nature 1984;309(5971):793-795.

112 Giusti B, Vestri ni A, Poggi C, Magi A, Pasquini E, Abbate R, Dani C. Genetic polymorphisms of antioxidant enzymes as risk factors for oxidative stress-associated complications in preterm infants. Free Radic Res 2012;46(9):1130-1139.

113 Johnson L, Bowen FW Jr, Abbasi S, Herrmann N, Weston M, Sacks L, Porat R, Stahl G, Peckham G, Delivoria-Papadopoulos M. Relationship of prolonged pharmacologic serum levels of vitamin E to incidence of sepsis and necrotizing enterocolitis in infants with birth weight 1,500 grams or less. Pediatrics 1985;75(4):619-638.

114 Brion LP, Bell E F, Raghuveer TS. Vitamin E supplementation for prevention of morbidity and mortality in preterm infants. Cochrane Database Syst Rev 2003;3:CD003665.

115 Fulton AB, Akula JD, Mocko JA, Hansen RM, Benador IY, Beck SC,Fahl E, Seeliger MW, Moskowitz A, Harris ME. Retinal degenerative and hypoxic ischemic disease. Doc Ophthalmol 2009;118(1):55-61.

116 Akula JD, Mocko JA, Benador IY, Hansen RM, Favazza TL,Vyhovsky TC, Fulton AB. The neurovascular relation in oxygen-induced retinopathy. Mol Vis 2008;14:2499-2508.

117 Rivera JC, Sitar as N, Noueihed B, Hamel D, Madaan A, Zhou T, Honoré JC, Quiniou C, Joyal JS, Hardy P, Sennlaub F, Lubell W,Chemtob S. Microglia and interleukin-1β in ischemic retinopathy elicit microvascular degeneration through neuronal semaphorin-3A.Arterioscler Thromb Vasc Biol 2013;33(8):1881-1891.

118 Joyal JS, Sitaras N, Binet F, et al. Ischemic neurons prevent vascular regeneration of neural tissue by secreting semaphorin 3A. Blood 2011;117(22):6024-6035.

119 Bucher F, Stahl A, A gostini HT, Martin G. Hyperoxia causes reduced density of retinal astrocytes in the central avascular zone in the mouse model of oxygen-induced retinopathy. Mol Cell Neurosci 2013,56:225-233.

120 Dorrell MI, Aguilar E, Jacobson R, Trauger SA, Friedlander J,Siuzdak G, Friedlander M. Maintaining retinal astrocytes normalizes revascularization and prevents vascular pathology associated with oxygen-induced retinopathy. Glia 2010;58(1):43-54.

121 Bai Y, Ma JX, Guo J, Wang J, Zhu M, Chen Y, Le YZ. Müller cellderived VEGF is a significant contributor to retinal neovascularization. J Pathol 2009;219:446-454.

122 Scott A, Powner MB, G andhi P, Clarkin C, Gutmann DH, Johnson RS, Ferrara N, Fruttiger M. Astrocyte-derived vascular endothelial growth factor stabilizes vessels in the developing retinal vasculature. PLoS ONE 2010;5(7):e11863.

123 Weidemann A, Krohne T U, Aguilar E, Kurihara T, Takeda N, Dorrell MI, Simon MC, Haase VH, Friedlander M, Johnson RS. Astrocyte hypoxic response is essential for pathological but not developmental angiogenesis of the retina. Glia 2010;58(10):1177-1185.

124 Håkansson J, Ståhlber g A, Wolfhagen Sand F, Gerhardt H, Semb H.N-CAM exhibits a regulatory function in pathological angiogenesis in oxygen induced retinopathy. PLoS One 2011;6(10):e26026.

125 Bagnoli P, Dal Monte M, Casini G. Expression of neuropeptides and their receptors in the developing retina of mammals. Histol Histopathol 2003;18(4):1219-1242.

126 Strand FL. Neuropeptide s: general characteristics and neuropharmaceutical potential in treating CNS disorders. Prog Drug Res 2003;61:1-37.

127 Eigler T, Ben-Shlomo A. Somatostatin system: molecular mechanisms regulating anterior pituitary hormones. J Mol Endocrinol 2014;53(1):R1-R19.

128 Monte MD, Ristori C, Ca mmalleri M, Bagnoli P. Effects of somatostatin analogues on retinal angiogenesis in a mouse model of oxygen-induced retinopathy: involvement of the somatostatin receptor subtype 2. Invest Ophthalmol Vis Sci 2009,50(8):3596-3606.

129 Dal Monte M, Ristori C, Videau C, Loudes C, Martini D, Casini G,Epelbaum J, Bagnoli P. Expression, localization, and functional coupling of the somatostatin receptor subtype 2 in a mouse model of oxygeninduced retinopathy. Invest Ophthalmol Vis Sci 2010;51(4):1848-1856.

130 Dal Monte M, Latina V, Cupisti E, Bagnoli P. Protective role of somatostatin receptor 2 against retinal degeneration in response to hypoxia. Naunyn Schmiedebergs Arch Pharmacol 2012;385(5):481-494.

131 Mastrodimou N, Kiagiada ki F, Thermos K. The role of nitric oxide and cGMP in somatostatin’s protection against retinal ischemia. Invest Ophthalmol Vis Sci 2008;49(1):342-349.

132 Cervia D, Catalani E, Dal Monte M, Casini G. Vascular endothelial growth factor in the ischemic retina and its regulation by somatostatin. J Neurochem 2012;120(5):818-829.

133 Hernández C, Simó R. So matostatin replacement: a new strategy for treating diabetic retinopathy. Curr Med Chem 2013;20(26):3251-3257.

134 Møller M, Phansuwan-Pujito P, Badiu C. Neuropeptide y in the adult and fetal human pineal gland. Biomed Res Int 2014;2014:868567.

135 Yoon HZ, Yan Y, Geng Y, Hig gins RD. Neuropeptide Y expression in a mouse model of oxygen-induced retinopathy. Clin Exp Ophthalmol 2002;30(6):424-429.

136 Schmid E, Nogalo M, Bechrak is NE, Fischer-Colbrie R, Tasan R, Sperk G, Theurl M, Beer AG, Kirchmair R, Herzog H, Troger J.Secretoneurin, substance P and neuropeptide Y in the oxygen-induced retinopathy in C57Bl/6N mice. Peptides 2012;37(2):252-257.

137 Cervia D, Casini G. The neu ropeptide systems and their potential role in the treatment of mammalian retinal ischemia: a developing story. Curr Neuropharmacol 2013;11(1):95-101.

138 Maiese K, Li F, Chong ZZ. N ew avenues of exploration for erythropoietin. JAMA 2005;293(1):90-95.

139 Sirén AL, Fasshauer T, Bart els C, Ehrenreich H. Therapeutic potential of erythropoietin and its structural or functional variants in the nervous system. Neurotherapeutics 2009;6(1):108-127.

140 Fauchère JC, Dame C, Vonthe in R, Koller B, Arri S, Wolf M, Bucher HU. An approach to using recombinant erythropoietin for neuroprotection in very preterm infants. Pediatrics 2008;122(2):375-382.

141 Chen J, Connor KM, Aderman C M, Smith LE. Erythropoietin de fi ciency decreases vascular stability in mice. J Clin Invest 2008;118(2):526-533.

142 Aher SM, Ohlsson A. Late e rythropoietin for preventing red blood cell transfusion in preterm and/or low birth weight infants. Cochrane Database Syst Rev 2014(4):CD004868.

143 Figueras-Aloy J, Alvarez-Domí nguez E, Morales-Ballus M, Salvia-Roiges MD, Moretones-Suñol G. Early administration of erythropoietin in the extreme premature, a risk factor for retinopathy of prematurity? An Pediatr (Barc) 2010;73(6):327-333.

144 May CA. The in fl uence of triamc inolone on endostatin-like proteins in oxygen-induced retinopathy of prematurity. Exp Eye Res 2012;100:86-87.

145 Ng PC, Kwok AK, Lee CH, Tam BS, Lam CW, Ma KC, Chan IH, Wong E, Lam DS, Fok TF. Early pituitary-adrenal responses and retinopathy of prematurity in very low birth weight infants. Pediatr Res 2004;55(1):114-119.

146 Wang YC, Tseng HI, Yang SN, Lu CC, Wu JR, Dai ZK, Hsu JH,Chen HL. Effects of antenatal corticosteroids on neonatal outcomes in very-low-birth-weight preterm newborns: a 10-year retrospective study in a medical center. Pediatr Neonatol 2012;53(3):178-183.

147 Tsukahara H, Watanabe Y, Yasuto mi M, Kobata R, Tamura S, Kimura K, Hiraoka M, Mayumi M. Early (4-7 days of age) dexamethasone therapy for prevention of chronic lung disease in preterm infants. Biol Neonate 1999;76(5):283-290.

148 Miller VM, Duckles SP. Vascular actions of estrogens: functional implications. Pharmacol Rev 2008;60(2):210-241.

149 Tulchinsky D, Osathanondh R, Be lisle S, Ryan KJ. Plasma estrone,estradiol, estriol and their precursors in pregnancies with anencephalic fetuses. J Clin Endocrinol Metab 1977;45(5):1100-1103.

150 Miyamoto N, Mandai M, Takagi H, Suzuma I, Suzuma K, Koyama S, Otani A, Oh H, Honda Y. Contrasting effect of estrogen on VEGF induction under different oxygen status and its role in murine ROP. Invest Ophthalmol Vis Sci 2002;43(6):2007-2014.

151 Zhang H, Sun N, Liang H, Xiao X, Liu X, Wang Y. The protective effect of 17 beta-estradiol on oxygen-induced retinopathy and its relation with the changes of malondialdehyde. J Biomed Res 2010;24(2):138-144.

152 Zhang HB, Sun NX, Liang HC, Xia o XH, Liu XN, Wang YN.17-Alpha-estradiol ameliorating oxygen-induced retinopathy in a murine model. Jpn J Ophthalmol 2012;56(4):407-415.

153 Zhang H, Wang X, Xu K, Wang Y, Wang Y, Liu X, Zhang X, Wang L,Li X. 17β-estradiol ameliorates oxygen-induced retinopathy in the early hyperoxic phase. Biochem Biophys Rese Commun 2015;457(4):700-705.

154 Katzenellenbogen BS, Choi I, De lage-Mourroux R, Ediger TR, Martini PG, Montano M, Sun J, Weis K, Katzenellenbogen JA.Molecular mechanisms of estrogen action: selective ligands and receptor pharmacology. J Steroid Biochem Mol Biol 2000;74(5):279-285.

155 Soltysik K, Czekaj P. Membrane e strogen receptors - is it an alternative way of estrogen action? J Physiol Pharmacol 2013;64(2):129-142.

156 Azevedo-Pinto S, Pereira-Silva P, Rocha-Sousa A. Ghrelin in ocular pathophysiology: from the anterior to the posterior segment. Peptides 2013;47:12-19.

157 Hosoda H, Kojima M, Kangawa K. B iological, physiological, and pharmacological aspects of ghrelin. J Pharmacol Sci 2006;100(5):398-410.

158 Stahl A, Chen J, Sapieha P, Seaw ard MR, Krah NM, Dennison RJ,Favazza T, Bucher F, Löfqvist C, Ong H, Hellström A, Chemtob S, Akula JD, Smith LE. Postnatal weight gain modifies severity and functional outcome of oxygen-induced proliferative retinopathy. Am J Pathol 2010;177(6):2715-2723.

159 Zaniolo K, Sapieha P, Shao Z, Stah l A, Zhu T, Tremblay S, Picard E,Madaan A, Blais M, Lachapelle P, Mancini J, Hardy P, Smith LE, Ong H, Chemtob S. Ghrelin modulates physiologic and pathologic retinal angiogenesis through GHSR-1a. Invest Ophthalmol Vis Sci 2011;52(8):5376-5386.

160 Chavez-Valdez R, McGowan J, Cannon E, Lehmann CU.Contribution of early glycemic status in the development of severe retinopathy of prematurity in a cohort of ELBW infants. J Perinatol 2011;31(12):749-756.

161 Mohamed S, Murray JC, Dagle JM, C olaizy T. Hyperglycemia as a risk factor for the development of retinopathy of prematurity. BMC Pediatr 2013;13:78.

162 Beardsall K, Ogilvy-Stuart AL, Fry styk J, Chen JW, Thompson M,Ahluwalia J, Ong KK, Dunger DB. Early elective insulin therapy can reduce hyperglycemia and increase insulin-like growth factor-I levels in very low birth weight infants. Pediatr 2007;151(6):611-617.

163 Kaempf JW, Kaempf AJ, Wu Y, Stawar z M, Niemeyer J, Grunkemeier G. Hyperglycemia, insulin and slower growth velocity may increase the risk of retinopathy of prematurity. J Perinatol 2011;31(4):251-257.

164 Heald A, Abdel-Latif ME, Kent AL. Insulin infusion for hyperglycaemia in very preterm infants appears safe with no effect on morbidity, mortality and long-term neurodevelopmental outcome. J Matern Fetal Neonatal Med 2012;25(11):2415-2418.

165 Bottino M, Cowett RM, Sinclair JC. Interventions for treatment of neonatal hyperglycemia in very low birth weight infants. Cochrane Database Syst Rev 2011(10):CD007453.

166 Sinclair JC, Bottino M, Cowett RM. Interventions for prevention of neonatal hyperglycemia in very low birth weight infants. Cochrane Database Syst Rev 2011(10):CD007615.

167 Augustin HG, Koh GY, Thurston G, A litalo K. Control of vascular morphogenesis and homeostasis through the angiopoietin-Tie system. Nat Rev Mol Cell Biol 2009;10(3):165-177.

168 Suri C, Jones PF, Patan S, Bartunk ova S, Maisonpierre PC, Davis S,Sato TN, Yancopoulos GD. Requisite role of angiopoietin-1, a ligand for the TIE2 receptor, during embryonic angiogenesis. Cell 1996;87(7):1171-1180.

169 Yancopoulos GD, Davis S, Gale NW, Rudge JS, Wiegand SJ, Holash J. Vascular-specific growth factors and blood vessel formation. Nature 2000;407(6801):242-248.

170 Jones N, Iljin K, Dumont DJ, Alita lo K. Tie receptors: new modulators of angiogenic and lymphangiogenic responses. Nat Rev Mol Cell Biol 2001;2(4):257-267.

171 Maisonpierre PC, Suri C, Jones PF, Bartunkova S, Wiegand SJ,Radziejewski C, Compton D, McClain J, Aldrich TH, Papadopoulos N, Daly TJ, Davis S, Sato TN, Yancopoulos GD. Angiopoietin-2, a natural antagonist for Tie2 that disrupts in vivo angiogenesis. Science 1997;277(5322):55-60.

172 Sato T, Shima C, Kusaka S. Vitreou s levels of angiopoietin-1 and angiopoietin-2 in eyes with retinopathy of prematurity. Am J Ophthalmol 2011;151(2):353-357.

173 Lee J, Kim KE, Choi DK, Jang JY, J ung JJ, Kiyonari H, Shioi G,Chang W, Suda T, Mochizuki N, Nakaoka Y, Komuro I, Yoo OJ, Koh GY. Angiopoietin-1 guides directional angiogenesis through integrin αvβ5 signaling for recovery of ischemic retinopathy. Sci Transl Med 2013;5(203):203ra127.

174 Oliner JD, Bready J, Nguyen L, Estra da J, Hurh E, Ma H, Pretorius J, Fanslow W, Nork TM, Leedle RA, Kaufman S, Coxon A. AMG 386,a selective angiopoietin 1/2-neutralizing peptibody, inhibits angiogenesis in models of ocular neovascular diseases. Invest Ophthalmol Vis Sci 2012;53(4):2170-2180.

175 Baker JG, Hill SJ, Summers RJ. Evolution of β-blockers: from anti-anginal drugs to ligand-directed signalling. Trends Pharmacol Sci 2011;32(4):227-234.

1 76 Daly CJ, McGrath JC. Previously unsuspected widespread cellular and tissue distribution of β-adrenoceptors and its relevance to drug action.Trends Pharmacol Sci 2011;32(4):219-226.

17 7 Ristori C, Filippi L, Dal Monte M, Martini D, Cammalleri M,Fortunato P, la Marca G, Fiorini P, Bagnoli P. Role of the adrenergic system in a mouse model of oxygen-induced retinopathy: antiangiogenic effects of beta-adrenoreceptor blockade. Invest Ophthalmol Vis Sci 2011;52(1):155-170.

17 8 Martini D, Monte MD, Ristori C, Cupisti E, Mei S, Fiorini P,Filippi L, Bagnoli P. Antiangiogenic effects of β2 -adrenergic receptor blockade in a mouse model of oxygen-induced retinopathy. J Neurochem 2011;119(6):1317-1329.

179 Filippi L, Cavallaro G, Bagnoli P, Dal Monte M, Fiorini P, Donzelli G, Tinelli F, Araimo G, Cristofori G, la Marca G, Della Bona ML, La Torre A, Fortunato P, Furlanetto S, Osnaghi S, Mosca F. Oral propranolol for retinopathy of prematurity: risks, safety concerns, and perspectives. J Pediatr 2013;163(6):1570-1577.e6.

180 Bancalari A, Schade R, Muñoz T, Lazcano C, Parada R, Peña R. Oral propranolol in early stages of retinopathy of prematurity. J Perinat Med 2016;44(5):499-503.

181 D al Monte M, Casini G, la Marca G, Isacchi B, Filippi L, Bagnoli P.Eye drop propranolol administration promotes the recovery of oxygeninduced retinopathy in mice. Exp Eye Res 2013;111:27-35.

182 Sarlos S, Wilkinson-Berka JL. The renin-angiotensin system and the developing retinal vasculature. Invest Ophthalmol Vis Sci 2005;46(3):1069-1077.

183 N guyen Dinh Cat A, Montezano AC, Burger D, Touyz RM.Angiotensin II, NADPH oxidase, and redox signaling in the vasculature.Antioxid Redox Signal 2013;19(10):1110-1120.

184 W ilkinson-Berka JL, Rana I, Armani R, Agrotis A. Reactive oxygen species, Nox and angiotensin II in angiogenesis: implications for retinopathy. Clin Sci 2013;124(10):597-615.

185 W ilkinson-Berka JL, Tan G, Jaworski K, Miller AG. Identi fi cation of a retinal aldosterone system and the protective effects of mineralocorticoid receptor antagonism on retinal vascular pathology. Circ Res 2009;104(1):124-133.

186 H ård AL, Wennerholm UB, Niklasson A, Hellström A. Severe ROP in twins after blockage of the renin-angiotensin system during gestation.Acta Paediatr 2008;97(8):1142-1144.

187 Per rotta S, Nobili B, Rossi F, Di Pinto D, Cucciolla V, Borriello A,Oliva A, Della Ragione F. Vitamin A and infancy. Biochemical, functional,and clinical aspects. Vitam Horm 2003;66:457-591.

188 Wang L, Shi P, Xu Z, Li J, Xie Y, Mitton K, Drenser K, Yan Q.Up-regulation of VEGF by retinoic acid during hyperoxia prevents retinal neovascularization and retinopathy. Invest Ophthalmol Vis Sci 2014;55(7):4276-4287.

189 She nai JP, Chytil F, Jhaveri A, Stahlman MT. Plasma vitamin A and retinol-binding protein in premature and term neonates. J Pediatr 1981;99(2):302-305.

190 Feu ngpean B, Suthutvoravut U, Supapannachart S, Rakthai S,Chatvuttinun S. Vitamin A status in premature infants. Chotmaihet thangphaet 2002;85(Suppl 4):S1219-S1223.

191 Mactier H, McCulloch DL, Hamilton R, Galloway P, Bradnam MS,Young D, Lavy T, Farrell L, Weaver LT. Vitamin A supplementation improves retinal function in infants at risk of retinopathy of prematurity. J Pediatr 2012;160(6):954-959.

192 Mac tier H. Vitamin A for preterm infants; where are we now? Semin Fetal Neonatal Med 2013;pii: S1744-165X(13)0005-X.

193 Ehr enkranz RA. Early, aggressive nutritional management for very low birth weight infants: what is the evidence? Semin Perinatol 2007;31(2):48-55.

194 Din erstein A, Nieto RM, Solana CL, Perez GP, Otheguy LE, Larguia AM. Early and aggressive nutritional strategy (parenteral and enteral)decreases postnatal growth failure in very low birth weight infants. J Perinatol 2006;26(7):436-442.

195 Can E, Bülbül A, Uslu S, Bolat F, Cömert S, Nuhoğlu A. Early aggressive parenteral nutrition induced high insulin-like growth factor 1 (IGF-1) and insulin-like growth factor binding protein 3 (IGFBP3)levels can prevent risk of retinopathy of prematurity. Iran J Pediatr 2013;23(4):403-410.

196 Porcell i PJ, Weaver RG Jr. The in fl uence of early postnatal nutrition on retinopathy of prematurity in extremely low birth weight infants. Early Hum Dev 2010;86(6):391-396.

197 Maayan- Metzger A, Avivi S, Schushan-Eisen I, Kuint J. Human milk versus formula feeding among preterm infants: short-term outcomes. Am J Perinatol 2012;29(2):121-126.

198 Manzon i P, Stol fi I, Pedicino R, et al. Human milk feeding prevents retinopathy of prematurity (ROP) in preterm VLBW neonates. Early Hum Dev 2013,89(Suppl 1):S64-S68.

199 VanderV een DK, Martin CR, Mehendale R, Allred EN, Dammann O, Leviton A, Investigators FTES. Early nutrition and weight gain in preterm newborns and the risk of retinopathy of prematurity. PLoS One 2013(5):e64325.

200 Beken S, Dilli D, Fettah ND, Kabataş EU, Zenciroğlu A, Okumuş N.The in fl uence of fi sh-oil lipid emulsions on retinopathy of prematurity in very low birth weight infants: a randomized controlled trial. Early Hum Dev 2014;90(1):27-31.

201 Kapoor V, Glover R, Malviya MN. Alternative lipid emulsions versus pure soy oil based lipid emulsions for parenterally fed preterm infants.Cochrane Database Syst Rev 2015(12):CD009172.

202 Löfqvist C, Andersson E, Sigurdsson J, Engström E, Hård AL, Niklasson A, Smith LE, Hellström A. Longitudinal postnatal weight and insulin-like growth factor I measurements in the prediction of retinopathy of prematurity. Arch Ophthalmol 2006;124(12):1711-1718.

203 Hellström A, Hår d AL, Engström E, Niklasson A, Andersson E, Smith L, Löfqvist C. Early weight gain predicts retinopathy in preterm infants: new,simple, ef fi cient approach to screening. Pediatrics 2009;123(4):e638-e645.

204 Wu C, Löfqvist C, Sm ith LE, VanderVeen DK, Hellström A.Importance of early postnatal weight gain for normal retinal angiogenesis in very preterm infants: a multicenter study analyzing weight velocity deviations for the prediction of retinopathy of prematurity. Arch Ophthalmol 2012;130(8):992-999.

205 Binenbaum G, Tomlinson LA. Postnatal Growth and retinopathy of prematurity study: rationale, design, and subject characteristics.Ophthalmic Epidemiol 2017;24(1):36-47.

206 Kim J, Jin JY, Kim SS. Postnatal weight gain in the first two weeks as a predicting factor of severe retinopathy of prematurity requiring treatment. Korean J Pediatr 2015;58(2):52-59.

207 Mukhopadhyay K, Yadav RK, Kishore SS, Garewal G, Jain V,Narang A. Iron status at birth and at 4 weeks in preterm-SGA infants in comparison with preterm and term-AGA infants. J Matern Fetal Neonatal Med 2012;25(8):1474-1478.

208 Joy R, Krishnamurthy S, Bethou A, Rajappa M, Ananthanarayanan PH, Bhat BV. Early versus late enteral prophylactic iron supplementation in preterm very low birth weight infants: a randomised controlled trial.Arch Dis Child Fetal Neonatal Ed 2014;99(2):F105-F109.

209 Mills RJ, Davies MW. E nteral iron supplementation in preterm and low birth weight infants. Cochrane Database Syst Rev 2012(3): CD005095.210 Vayalthrikkovil S, Bashir RA, Rabi Y, Amin H, Spence JM, Robertson HL, Lodha A. Parenteral fish-oil lipid emulsions in the prevention of severe retinopathy of prematurity: a systematic review and meta-analysis.Am J Perinatol 2017;34(7):705-715.

211 Connor KM, SanGiovanni JP, Lofqvist C, Aderman CM, Chen J,Higuchi A, Hong S, Pravda EA, Majchrzak S, Carper D, Hellstrom A,Kang JX, CheWeY, Salem N Jr, Serhan CN, Smith LEH. Increased dietary intake of omega-3-polyunsaturated fatty acids reduces pathological retinal angiogenesis. Nat Med 2007;13(7):868-873.

212 Sapieha P, Stahl A, Ch en J, Seaward MR, Willett KL, Krah NM,Dennison RJ, Connor KM, Aderman CM, Liclican E, Hellstrom A, Kang JX, CheWeY, Salem N Jr, Serhan CN, Smith LEH. 5-Lipoxygenase metabolite 4-HDHA is a mediator of the antiangiogenic effect of ω-3 polyunsaturated fatty acids. Sci Transl Med 2011;3(69):69ra12.

213 Shao Z, Fu Z, Stahl A, et al. Cytochrome P450 2C8 ω3-long-chain polyunsaturated fatty acid metabolites increase mouse retinal pathologic neovascularization-brief report. Arterioscler Thromb Vasc Biol 2014;34(3):581-586.

214 Tolsma KW, Allred EN, C hen ML, Duker J, Leviton A, Dammann O.Neonatal bacteremia and retinopathy of prematurity: the ELGAN study.Arch Ophthalmol 2011;129(12):1555-1563.

215 Mularoni A, Madrid M, Az peitia A, Valls i Soler A. The role of coagulase-negative staphylococci in early-onset sepsis in a large European cohort of very low birth weight infants. Pediatr Infect Dis J 2014;33(5):e121-e125.

216 Borroni C, Carlevaro C, Morzenti S, De Ponti E, Bozzetti V, Console V, Capobianco S, Tagliabue PE. Survey on retinopathy of prematurity(ROP) in Italy. Ital J Pediatr 2013;39:43.

217 Tremblay S, Miloudi K, C haychi S, Favret S, Binet F, Polosa A,Lachapelle P, Chemtob S, Sapieha P. Systemic inflammation perturbs developmental retinal angiogenesis and neuroretinal function. Invest Ophthalmol Vis Sci 2013;54(13):8125-8139.

218 Ryu J, Haddad G, Carlo W A. Clinical effectiveness and safety of permissive hypercapnia. Clin Perinatol 2012;39(3):603-612.

219 Martínez-Cruz CF, Salgad o-Valladares M, Poblano A, Trinidad-Pérez MC. Risk factors associated with retinopathy of prematurity and visual alterations in infants with extremely low birth weight. Rev Invest Clin 2012;64(2):136-143.

220 Holmes JM, Leske DA, Zhang S. The effect of raised inspired carbon dioxide on normal retinal vascular development in the neonatal rat. Curr Eye Res 1997;16(1):78-81.

221 Holmes JM, Zhang S, Leske DA, Lanier WL. Carbon dioxide-induced retinopathy in the neonatal rat. Curr Eye Res 1998;17(6):608-616.

222 Checchin D, Sennlaub F, Si rinyan M, Brault S, Zhu T, Kermorvant-Duchemin E, Hardy P, Balazy M, Chemtob S. Hypercapnia prevents neovascularization via nitrative stress. Free Radic Biol Med 2006;40(3):543-553.

223 Leduc M, Kermorvant-Duchem in E, Checchin D, Sennlaub F,Sirinyan M, Kooli A, Lachapelle P, Chemtob S. Hypercapnia- and trans-arachidonic acid-induced retinal microvascular degeneration:implications in the genesis of retinopathy of prematurity. Semin Perinatol 2006;30(3):129-138.

224 Bennet D, Kim S. Impedance-based cell culture platform to assess light-induced stress changes with antagonist drugs using retinal cells. Anal Chem 2013;85(10):4902-4911.

225 Jorge EC, Jorge EN, El Dib RP. Early light reduction for preventing retinopathy of prematurity in very low birth weight infants. Cochrane Database Syst Rev 2013(8):CD000122.

226 Asahara T, Masuda H, Takah ashi T, Kalka C, Pastore C, Silver M,Kearne M, Magner M, Isner JM. Bone marrow origin of endothelial progenitor cells responsible for postnatal vasculogenesis in physiological and pathological neovascularization. Circ Res 1999;85(3):221-228.

227 Grant MB, May WS, Caballer o S, Brown GA, Guthrie SM, Mames RN, Byrne BJ, Vaught T, Spoerri PE, Peck AB, Scott EW. Adult hematopoietic stem cells provide functional hemangioblast activity during retinal neovascularization. Nat Med 2002;8(6):607-612.

228 Nakagawa Y, Masuda H, Ito R, Kobori M, Wada M, Shizuno T, Sato A, Suzuki T, Kawai K, Asahara T. Aberrant kinetics of bone marrowderived endothelial progenitor cells in the murine oxygen-induced retinopathy model. Invest Ophthalmol Vis Sci 2011;52(11):7835-7841.

229 Villalvilla A, Moro M, Arr uza L, Redondo S, Fernández-Cruz A,Fernández-Durango R. Circulating endothelial progenitor cells are reduced in rat oxygen-induced retinopathy despite a retinal SDF-1/CXCR4 and VEGF proangiogenic response. Life Sci 2012;91(7-8):264-270.

230 Liu X, McBride J, Zhou Y, Li u Z, Ma JX. Regulation of endothelial progenitor cell release by Wnt signaling in bone marrow. Invest Ophthalmol Vis Sci 2013;54(12):7386-7394.

231 Machalinska A, Modrzejewska M, Kotowski M, Dziedziejko V,Kucia M, Kawa M, Safranow K, Baskiewicz-Masiuk M, Modrzejewska A, Karczewicz D, Rudnicki J, Machalinski B. Circulating stem cell populations in preterm infants: implications for the development of retinopathy of prematurity. Arch Ophthalmol 2010;128(10):1311-1319.232 Fujiyama S, Amano K, Uehira K, Yoshida M, Nishiwaki Y, Nozawa Y, Jin D, Takai S, Miyazaki M, Egashira K, Imada T, Iwasaka T,Matsubara H. Bone marrow monocyte lineage cells adhere on injured endothelium in a monocyte chemoattractant protein-1- dependent manner and accelerate reendothelialization as endothelial progenitor cells. Circ Res 2003;93(10):980-989.

233 Ishikawa K, Yoshida S, Nakao S, Sassa Y, Asato R, Kohno R, Arima M, Kita T, Yoshida A, Ohuchida K, Ishibashi T. Bone marrow-derived monocyte lineage cells recruited by MIP-1β promote physiological revascularization in mouse model of oxygen-induced retinopathy. Lab Invest 2012;92(1):91-101.

234 Ritter MR, Banin E, Moreno SK, Aguilar E, Dorrell MI, Friedlander M. Myeloid progenitors differentiate into microglia and promote vascular repair in a model of ischemic retinopathy. J Clin Invest 2006;116(12):3266-3276.

235 Ishikawa K, Yoshida S, Kadota K, Nakamura T, Niiro H, Arakawa S,Yoshida A, Akashi K, Ishibashi T. Gene expression pro fi le of hyperoxic and hypoxic retinas in a mouse model of oxygen-induced retinopathy.Invest Ophthalmol Vis Sci 2010;51(8):4307-4319.

236 Yang X, Dong X, Jia C, Wang Y. Pro fi ling of genes associated with the murine model of oxygen-induced retinopathy. Mol Vis 2013;19:775-788.

237 Kim SJ, Jin J, Kim YJ, Kim Y, Yu HG. Retinal proteome analysis in a mouse model of oxygen-induced retinopathy. J Proteome Res 2012;11(11):5186-5203.

238 Fleck BW. Management of retinopathy of prematurity. Arch Dis Child Fetal Neonatal Ed 2013;98:F454-F456.

239 Romagnoli C. Risk factors and growth factors in ROP. Early Hum Dev 2009;85(10 Suppl):S79-S82.

Hong-Bing Zhang,Xiao-Dong Wang,Kun Xu,Xiao-Gang Li
《International Journal of Ophthalmology》2018年第5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