Skip to main content
Cellular and Molecular Life Sciences: CMLS logoLink to Cellular and Molecular Life Sciences: CMLS
. 2012 Mar 14;69(17):2863–2878. doi: 10.1007/s00018-012-0957-x

The cerebrospinal fluid: regulator of neurogenesis, behavior, and beyond

Mauro W Zappaterra 1, Maria K Lehtinen 2,
PMCID: PMC3856656  NIHMSID: NIHMS529059  PMID: 22415326

Abstract

The cerebrospinal fluid (CSF) has attracted renewed interest as an active signaling milieu that regulates brain development, homeostasis, and disease. Advances in proteomics research have enabled an improved characterization of the CSF from development through adulthood, and key neurogenic signaling pathways that are transmitted via the CSF are now being elucidated. Due to its immediate contact with neural stem cells in the developing and adult brain, the CSF’s ability to swiftly distribute signals across vast distances in the central nervous system is opening avenues to novel and exciting therapeutic approaches. In this review, we will discuss the development of the choroid plexus-CSF system, and review the current literature on how the CSF actively regulates mammalian brain development, behavior, and responses to traumatic brain injury.

Keywords: Cerebrospinal fluid, Choroid plexus, Neurogenesis, Traumatic brain injury

Introduction

The cerebrospinal fluid (CSF) has intrigued scientists, physicians, and philosophers for close to 4,000 years. The earliest documentation of a fluid within the head dates back to the ancient Egyptians, dating back to 1700 b.c. [1]. It was the early Greeks, however, who took on the first known investigations of the CSF. Hippocrates (460–370 b.c.) described ‘hydrocephalus,’ a condition that he thought was caused by too much ‘water in the head’ [1, 2]. Subsequently, circa 300–250 b.c., Herophilus is credited for being the first to describe the choroid plexus and named it “chorioid mennix” due to the vascular similarity to the “chorioid” of the fetus [3]. Herophilus and Erasistratus, both anatomists interested in the human body, are credited for being the first to describe the ventricles of the brain and suggested they play a role in muscular contraction [2]. However, it is believed that Galen of Pergamon (circa a.d. 150), due to his fame and popularity, may have had the most historical influence on describing the function of the ventricles and the CSF by studying Ox brains [1, 2]. He described an alchemical transformation that happened at the base of the brain as a ‘vital spirit’ coming from the blood vessels that was transformed into the ‘animal spirit’ and emerged into the ventricles. This ‘animal spirit’ was then carried through the nerves to contract the muscles of the body and energize the entire physical being [2]. Galen’s prevailing theory of a fluid within the ventricles as a spirit (pneuma) lasted for over 1,500 years, until the rebirth of human anatomy at the time of the Renaissance [2]. Today, we know that the CSF is produced primarily by the choroid plexus, located in each ventricle in the brain. Elucidating the full range of CSF functions continues to be an area of research under intense investigation today [46].

The choroid plexus, the intraventricular organ

The choroid plexus is a highly vascularized secretory epithelium that begins to form shortly following anterior neural tube closure. It arises from an invagination of the dorsal roof plate along the midline of the neural tube and develops in vertebrate ventricles in a sequential manner [7]. It first can be visualized as epithelial invaginations that emanate from the single-layered roof plate of the hindbrain that create the fourth ventricle choroid plexus at approximately embryonic mouse age E11–E12 [7]. Subsequently, midline invaginations from the area chorioidea, an area of the medial wall named by His, then forms the anterior and posterior domains of the telencephalic choroid plexus, which extend into the lateral ventricles (E11–E12) [7]. Finally, the diencephalic choroid plexus forms in the third ventricle at approximately E12–E14 in the mouse [3, 8]. Subsequently, the continuity between the telencephalic and diencephalic choroid plexuses develops [9]. During mouse embryogenesis, the choroid plexus is clearly visible as an observable outgrowth already at E12.5 in the hindbrain ventricle. In the human embryo, the choroid plexus begins to develop at approximately 44 days post-ovulation in the fourth and lateral ventricles and is quite large already at 9 weeks gestation, filling a large part of the lateral ventricle and spanning the length of the fourth ventricle (Fig. 1) [10]. One of the earliest molecular markers of choroid plexus differentiation is the expression of Transthyretin [11].

Fig. 1.

Fig. 1

Coronal section of the human embryonic brain at gestational week 9. Left panel At gestational week 9 in the human embryo, the telencephalic choroid plexus occupies a large portion of the lateral ventricle. The choroid-plexus-secreted CSF in the lateral ventricles bathes the cortical neuroepithelium, thereby regulating neurogenesis and the formation of the cortical plate. Right panel a more posterior view of the developing human embryo at gestational week 9 shows that the lateral ventricle continues to be filled by the choroid plexus. The fourth ventricle choroid plexus is observed to extend along the length of the fourth ventricle. Asterisks denote third ventricle choroid plexus. ChP choroid plexus; LV lateral ventricle; NEP neuroepithelium; Tel telencephalon; Rhomb rhombencephalon; V ventricle. Reprinted with permission from the authors, Bayer SA, Altman J [10]

The choroid plexus consists of highly fenestrated capillaries, which are essentially microvessels with portions of the capillary wall sealed by thin diaphragms, mesenchymal cells, and epithelial cells. The secretory epithelial cell layer is derived from neuroepithelial cells lining the ventricles during development. The epithelial cells contain tight junctions that already function very early in development and prevent the passive diffusion of molecules into the CSF from the choroidal vasculature and interstitial space [12, 13]. As such, the choroid plexus actively secretes CSF into the ventricles and creates the blood–CSF barrier.

The localized formation of the choroid plexus is regulated by a number of growth and transcription factors. In the hindbrain, the choroid plexus develops from the lower rhombic lip that is specified by high levels of bone morphogenetic protein (BMP) signaling, and demarcated by the expression of growth factors Wnt1, and Gdf7, and the transcription factor Lmx1a [1420]. Loss-of-function and gain-of-function studies have also shown that Sonic hedgehog (Shh) originating from fourth ventricle choroid plexus epithelial cells promotes proliferation of fourth-ventricle choroid plexus progenitor cells [18] as well as the choroid plexus mesenchyme [19]. For lateral ventricle choroid plexus development, the BMPs specify choroid plexus cell fate within the dorsal midline, as demonstrated by loss of choroid plexus in mice with conditional deletion of the BMP receptor 1a (Bmpr1a) within the developing telencephalon using the Foxg1-Cre allele, a gene expressed throughout the telencephalon and along the dorsal midline [21]. In addition, the BMPs generate an activity gradient at the dorsal midline telencephalon that induces telencephalic choroid plexus differentiation [22]. The anterior and posterior domains of the telencephalic choroid plexus can be further distinguished by Gdf-7, whose expression localizes to the anterior domain [16]. During normal chick development, the telencephalic choroid plexus can be distinguished from surrounding neuroepithelial tissue as early as embryonic day 4 (E4), when the developing choroid plexus expresses Bmp7 and Otx2, lacks expression of both Emx1 and Emx2 in neighboring cells, and can be clearly differentiated by E6 with the expression of Transthyretin [11].

The cerebrospinal fluid and its functions

Although the study of the CSF dates back to the early Egyptians and Greeks, it was not until about 100 years ago that Harvey Cushing observed that the choroid plexus secretes CSF into the brain’s ventricles [23], casting aside lingering views of the CSF as a possible post-mortem precipitate. It is estimated that an adult human circulates 150 ml of CSF throughout the central nervous system (CNS), and that the CSF turns over about three to four times per day, suggesting that an adult human produces approximately 500 ml of CSF daily. In the adult brain, the CSF flows from the lateral ventricles to the third ventricle, fourth ventricle, and then to the spinal cord and subarachnoid space (Fig. 2). Its flow is propelled by three main forces: (1) arterial pulsations in the choroid plexus, (2) a hydrostatic pressure gradient, and (3) ciliary motion of the ependymal cells lining the ventricles [12]. While the CSF is 99% water, provided primarily by Aquaporin-1 channels located on the apical membrane of the epithelial cells, the CSF is also a rich source of proteins, ions, lipids, hormones, cholesterol, glucose, and many other molecules and metabolites. Early work on the CSF focused primarily on the importance of pressure homeostasis in the developing brain [2426]—too little pressure decreases brain size [24], while too much pressure leads to impaired neural stem cell division and hydrocephalus [2729].

Fig. 2.

Fig. 2

Sagittal schematic of CSF flow in the mature cerebroventricular system. CSF is produced primarily by the choroid plexus, which is located in each ventricle in the brain. CSF flows from the lateral ventricles, via the foramen of Monro, to the third ventricle, and then through the cerebral aqueduct/aqueduct of Sylvius to the fourth ventricle. Upon leaving the fourth ventricle, the CSF flows down the spinal cord and fills the subarachnoid space surrounding the brain. It is ultimately resorbed into the venous circulation by arachnoid villi

More recent work has turned to investigating the biological functions of known CSF-resident factors. Many of these CSF-distributed factors have been speculated to influence a wide range of behaviors including sleep and appetite. For example, early experiments showed that CSF collected from sleep-deprived goats induces states of deep sleep for 12–24 h when infused into the cisterna magna of control laboratory cats [30]. In an attempt to isolate the molecule, Lerner and colleagues purified a brain fatty acid from the CSF of sleep-deprived cats [31], which was later identified as oleamide (cis-9,10-octadecenoamide) [32]. Further studies revealed that oleamide levels are elevated in the CSF of sleep-deprived rats [33]. When synthetic oleamide is injected into laboratory rats either intraventricularly or intraperitoneally, it decreases sleep latency and induces physiologic sleep [33, 34]. The exact mechanism by which oleamide induces sleep remains to be elucidated. However, oleamide binds to cannabinoid (CB1), GABA, and serotonin receptors [3537], and is thought to modulate its hypnotic function through these interactions.

An arousal-promoting factor has also been found in the CSF. Orexin-A (also known as hypocretin-1), a protein released by lateral hypothalamic neurons and a key regulator of wakefulness [3840], is present at lower levels in the CSF of narcoleptic patients than unaffected individuals [41]. Orexin infusion into the lateral ventricles elicits prolonged wakefulness in rats that are awake [4244] and stimulates arousal from sleep [45]. Interestingly, orexin infusion into different ventricles appears to evoke distinct behavioral responses. For example, in contrast to the pronounced waking response stimulated by lateral ventricle infusions, orexin infusion into the fourth ventricle leads to weaker effects on arousal [45], suggesting that the sites of CSF-orexin action flank, or lie close to the lateral and/or third ventricular walls, consistent with known targets of orexin projections.

In addition to sleep- and arousal-inducing factors in the CSF, several CSF-distributed factors play important roles in circadian rhythms and associated locomotor activity. For example, encapsulated suprachiasmatic nucleus (SCN) transplants to the third ventricle wall can restore circadian rhythms in hamsters whose own SCN has been ablated [46]. Although the specific SCN-secreted factors were not identified in the above-mentioned study, additional clues have since emerged, including the identification of TGF-α [47] and cardiotrophin-like cytokine [48]. When infused into the third ventricle, both of these factors disrupt circadian locomotor activity. TGF-α infusion also impairs circadian rhythmicity, an effect mediated by the EGF receptors expressed by hypothalamic subparaventricular zone neurons [47]. In addition, melatonin, a hormone that regulates circadian rhythms, is released by the pineal gland into the CSF [49, 50]. Melatonin is a potent free radical scavenger and antioxidant, and it is believed that one of the major roles of melatonin in the CNS is to behave as a neuroprotective agent that decreases oxidative stress and removes reactive oxygen species from the CNS [51]. Interestingly, patients with severe traumatic brain injury have been reported to have increased CSF melatonin levels that correlate with markers of oxidative stress [52], suggesting a possible homeostatic role for melatonin in the CSF.

CSF-borne factors have also been shown to affect appetite. When CSF collected from the lateral ventricles of fasted “donor” sheep is infused into the ventricles of satiated “recipient” sheep, increased feeding is observed. The converse experiment holds true as well, such that when satiated “recipients” receive CSF from satiated “donors,” the recipients feed less [53]. Although the exact CSF-distributed factors mediating feeding remain to be elucidated, several candidates have been suggested. Insulin injection into the lateral ventricles, for example, elicits decreased food intake in baboons [54]. In addition, low CSF-leptin:serum-leptin ratios have been reported to correlate inversely with body mass index (BMI) in some individuals [55], raising the possibility that impaired leptin transport across the blood–brain barrier could underlie increased BMI [55]. Therefore, due to its intimate contact with the thalamus, hypothalamus, and pineal gland, the CSF may convey important peptide signals regulating behavioral states and other states of consciousness as well.

Spatial gradients of CSF proteins have also been shown to guide the behavior of distinct cell populations, including the migration of neuroblasts from the subventricular zone (SVZ) to the olfactory bulb in the adult brain [56]. In these studies, the ciliary beating of ependymal cells was shown to be crucial for providing appropriate Slit chemorepellent activity for the migrating neuroblasts [56]. Taken together, CSF-distributed factors play an active role in many aspects of normal adult brain activity, setting the stage for investigating how the CSF may dynamically regulate development as well as disease.

Brain development and the cerebrospinal fluid

Recent studies have taken advantage of advances in proteomic tools including mass spectrometry to better characterize the CSF proteome. These studies have demonstrated that the CSF proteome consists of hundreds of proteins of the extracellular matrix, regulators of osmotic pressure, ion carriers, hormone-binding proteins, regulators of lipid metabolism, and various enzymes and their regulators [5761]. The CSF shares many similarities across species [5761], facilitating a multi-tiered experimental approach across many laboratories for the study of CSF including zebrafish, chick, rodents, marsupials, and others, on brain development. While the classical model of diffusion through tissues certainly holds true [62, 63], the apical, ventricular enrichment of phosphotyrosine [64] and phospho-ERK1/2 [65] activities in cortical progenitor cells have raised the provocative idea that growth factor signaling originates from the CSF as well. Indeed, a flurry of recent studies demonstrates the dynamism of embryonic CSF during brain development. CSF alone can promote the development and growth of neural stem cells and cortical explants [66]. Although the factors responsible for the favorable effects of embryonic CSF are only beginning to be uncovered, the range of factors already known to be active in CSF during embryonic brain development includes fibroblast growth factors (FGFs) [67], insulin-like growth factors (IGFs) [66, 68], sonic hedgehog (Shh) [69], retinoic acid (RA) [66, 70], bone morphogenic proteins (BMPs) [66], Wnts [66], and others [71, 72] shown in Table 1.

Table 1.

Growth factors present in embryonic CSF

Growth factor Method of detection Species References
Amyloid beta A4 protein precursor (APP) Mass spectrometry (MS) Rat, Human [60]
BMPs Luciferase assay Rat [66]
FGF2 Western blot Chick [67]
GDF-3 MS Rat [66]
GDF-8 MS Rat [66]
IGF-1 ELISA/western blot Mouse, Rat [66, 68]
IGF-2 MS/western blot Mouse, Rat [66]
Leukemia inhibitory factor (LIF) ELISA Mouse [72]
NGF ELISA/western blot Chick [71]
Pigment epithelial derived factor (PEDF) MS Human [60]
Retinoic acid HPLC-MS, cell based assay Chick, Rat [66, 70]
Shh ELISA Mouse [69]
Wnts Cell based assay Rat [66]

FGF2, IGF2, and Shh in CSF have been demonstrated to stimulate proliferation of progenitor cells in the brain, with each factor having a distinct known cellular target to date. For example, immunodepletion of embryonic chick CSF-FGF2 reduces progenitor proliferation of chick midbrain progenitors [67, 73, 74]. Intravascularly injected FITC-conjugated FGF2 passes into the embryonic CSF [67], suggesting that somatic sources with access to CSF may regulate neurogenesis as well. The following sections will summarize findings for IGFs, Shh, RA, as well as other factors.

Insulin-like growth factor 1 and 2 signaling in the cerebrospinal fluid

The IGFs (IGF1 and IGF2) are a class of growth factors present in the embryonic CSF [66, 68, 75, 76]. Well established in the regulation of prenatal growth and body size [7779], the IGFs bind with strongest affinity to the IGF1 receptor (IGF1R) to stimulate proliferation [80]. Conditional deletion of IGF1R in neural precursors leads to microcephaly [66, 81, 82]. In contrast, IGF1 overexpression promotes S-phase commitment, accelerated cell cycle kinetics, and cell survival, leading to hyperplasia [8285]. Depending on cellular context, IGF1 can also regulate neuronal differentiation, glial development, and cell size [86]. Downstream of IGF1R signaling, several mouse mutants can be leveraged for the study of brain size, including Irs2 [87], Pdk1 [88], and Pten [89].

IGF2 endows the CSF with robust growth and survival-promoting effects during neurogenesis [66]. A transient spike in CSF-IGF2 levels at middle-to late-stages of neurogenesis (E16–19 in rat; E16–18 in mice) stimulates the proliferation of neural precursor cells in explant cultures of the developing cortex [66], as well as the growth and maintenance of neurospheres, an in vitro model of neural stem cells [90, 91]. CSF-IGF2 binds to the apical membrane and primary cilia of cortical progenitors, and the proliferation-inducing effects of CSF appear IGF2-dependent, as gain-of-function and loss-of-function experiments in vitro in explants and neurospheres produce opposing effects on proliferation. Igf2-deficient mice have a defect in neurogenesis affecting the uppermost layers of the cortex [66]. The choroid plexus expresses IGF2 [92, 93], though other sources of IGF ligands exist, including the developing vasculature [94], neighboring cells, and perhaps even somatic sources. Studies involving intracerebroventricular injections of IGF1, IGF1-neutralizing antibodies, and IGF1R inhibitors further demonstrate that IGF signals delivered by the embryonic CSF trigger proliferative events in the cortical ventricular zone (VZ) [84].

The effects of CSF-IGF2 are also age-dependent [66]. CSF-IGF2 levels in rodents are modest at the earliest stages of neurogenesis, peak near the end of neurogenesis, and decrease postnatally, suggesting that IGF2 may regulate specific aspects of neurogenesis. Intriguingly, in Drosophila, insulin/IGF-like peptides secreted by glia stimulate quiescent neuroblasts to reenter the cell cycle in Drosophila [95, 96]. An analogous role of IGF signaling in the mammalian brain is an interesting idea that has not yet been explored.

Sonic hedgehog signaling in the cerebrospinal fluid

Sonic hedgehog (Shh), a morphogen well established in ventral brain and cerebellar development, is also found in the CSF. The hindbrain choroid plexus epithelial cells secrete Shh into the CSF [18, 69], and this CSF-Shh has been suggested to stimulate the expansion of a progenitor domain in the hindbrain choroid plexus [18]. Interestingly, Shh also signals directly to choroid plexus pericytes to direct vascular growth, an essential component of normal choroid plexus development and expansion [19]. Pericyte signals may subsequently influence both progenitor proliferation as well as angiogenesis [19]. Thus, more detailed mouse genetic approaches will elucidate the exact impact of Shh on distinct cell lineages and on choroid plexus development. In addition to this tissue-autonomous role for choroid-plexus-secreted-Shh, CSF-Shh appears to be a key mitogen for proliferating cerebellar granule precursors. In this context, Shh was previously thought to be produced primarily by Purkinje cells [9799]. However, Wnt1-Cre-mediated deletion of Shh in the hindbrain choroid plexus also impairs proliferation of cerebellar granule neuron precursors [69]. These findings support a model in which choroid plexus borne Shh may also act in a paracrine manner to instruct cerebellar development.

Retinoic acid in the cerebrospinal fluid

Retinoic acid (RA), a hormone signal derived from vitamin A, has been identified in CSF [6, 66, 70]. RA provides key long-range signaling activity for the developing brain [100102]. RA synthetic and catabolic enzymes are expressed in the choroid plexus [66] as well as the meninges [103]. Thus, the meninges are an important source of RA as well. Foxc1 mutant mice, which have reduced meningeal RA secretion, have diminished expansion of the neuroepithelium and decreased numbers of intermediate progenitor cells [103]. While choroid-plexus-derived-RA is secreted directly into ventricular CSF, meningeal sources of RA may reach the neuroepithelial cells via the lateral ventricular CSF as well as by crossing the cerebral mantle. The roles of meningeal-RA suggest a wide range of mechanisms by which signals at both the basal process [104, 105] and apical process may play key roles in the maintenance of the apical progenitors [5].

Other potential signaling activities in the cerebrospinal fluid

The CSF is home to hundreds of proteins and signaling activities, providing an elaborate range of biological functions for this complex fluid. For example, Wnt signaling, which plays a fundamental role in regulating early development of the CNS [106108], has been identified in embryonic CSF [66]. Similarly, dynamic BMP activity [21, 109] is also found in the CSF [66]. Consistent with the coordination of BMP signaling by the choroid plexus-CSF system, growth and differentiation factors 3 and 8 (Gdf3 and Gdf8), both members of the TGF-β superfamily of proteins that can modulate BMP signaling [110], are found in the CSF [66]. Our knowledge of CSF-distributed signals likely represents the tip of the iceberg with regard to active signaling in this highly dynamic fluid (Fig. 3).

Fig. 3.

Fig. 3

Coronal schematic of choroid plexus-secreted factors distributed in the CSF during embryonic cortical development. IGF2 (represented by purple spheres) and other factors secreted by the choroid plexus are delivered to target cells on the apical, ventricular surface of the developing cerebral cortex

The cerebrospinal fluid and brain injury

The CSF has also attracted interest in the field of brain injury. The CSF utilizes volume transmission, or bulk flow, of neurogenic and neuroprotective factors believed to have therapeutic effects following brain injury. The continuous exchange of CSF also serves as a “sink” that flushes the ventricular system of debris and injury products. The choroid-plexus-CSF system can be disrupted in a wide range of CNS injuries, from hydrocephalus to age-associated neurologic diseases (e.g., Alzheimer’s disease) [12, 111]. For the purposes of this review, we will focus primarily on brain injury via traumatic or ischemic events.

There are a number of mechanisms that may occur during brain injury that disrupt the choroid plexus-CSF (CP-CSF) homeostasis [111]. Some of the experimental models for investigating traumatic brain injury (TBI) typically include: lateral head movement acceleration, exposure to a blast, penetrating brain injury, and impact acceleration. These injury approaches induce damage to the choroid plexus, alterations in CSF secretion, increased leukocytes within the brain and CSF, decreased clearance of toxins, catabolites and proteins, and therefore diminished supply of nutrients and growth factors to the brain [111]. It is now believed that recovery from brain injury involves the gradual repair of these mechanisms in order to return the CP-CSF system to homeostasis to promote neuroregeneration and neuroprotection [111].

Growth factors in CNS injury

A number of growth factors are upregulated in the brain and CSF following injury. These include, but are not limited to, IGF1 [112114], IGF2 [115], FGF [116, 117], nerve growth factor (NGF) [118, 119], transforming growth factor-beta (TGF-β) [120122], glial-derived neurotrophic factor (GDNF) [123], brain-derived neurotrophic factor (BDNF) [112, 124, 125], Neurotrophin 3 (NT-3) [112, 126, 127], Neurotrophin 4/5 [128], vascular endothelial growth factor (VEGF) [129], erythropoietin [130132], tumor necrosis factor-alpha (TNF-alpha) [133135], and soluble amyloid precursor protein (sAPP) [136, 137] (Fig. 4; Table 2). Recently, NGF was found to be elevated in the CSF of children post TBI at both 2 and 48 h post injury. In addition, increased NGF levels, together with lower IL-1β levels, were reported to correlate with improved neurological outcomes [118, 138, 139].

Fig. 4.

Fig. 4

Schematic illustrating brain states and associated factors distributed in the CSF. Different brain/body states including sleep/wake, appetite, and brain injury and repair have been linked with the presence of distinct factors in the CSF. In the case of brain injury and repair, most of the factors listed have been introduced intraventricularly into injury models, where they were reported to have beneficial effects on recovery from injury. The primary source and mode of action for many CSF factors remain to be elucidated

Table 2.

Growth factors present in CSF in brain injury

Growth factor Proposed function in brain injury Species; injury condition Reference
BDNF Neuroprotection Human; Ischemia [125]
Erythropoietin Neuroprotection Human; Ischemia [130132]
IGF-2 Assists with wound healing via stimulation of neurogenesis and tissue homeostasis; possibly involved in promoting phagocytic activity Rat; Penetrating injury [115]
NGF Supports survival and growth of neurons Human; TBI [118, 119, 138, 139]
sAPP Neuroprotection, stimulates proliferation of neural stem cells Human; TBI [136, 137]
TGF-β Supports neuronal survival, suppresses inflammation, regulates glial scar formation, fibrosis, and microglial activation Human; TBI [120, 122]
TNF-alpha Dual role exhibiting both neurotoxic properties in striatum and neuroprotection in hippocampus Human, Rat; TBI [133135]
VEGF Neuroprotection Human; TBI [129]

The IGF signaling pathway plays an important role in models of brain injury. IGF1 was shown to be upregulated and to localize to the site of cortical contusion up to 48 h post-injury in a mouse model of cortical impact brain injury [113], suggesting a possible role in neuroprotection and neurogenesis by IGF1. Previous cortical contusion studies in rats also showed similar results with Igf1 mRNA increasing at the site of injury and peaking by day 3 [114]. Although these studies did not assess changes in IGF1 within the CSF, they indicate that the IGF signaling pathway may be an important modulator in TBI models. This observation is further supported by studies with IGF2. IGF2 is highly expressed in the choroid plexus, meninges, and CSF throughout development and into adulthood [140142]. As mentioned above, increased IGF2 levels in vitro increase proliferation of neural stem cells both in cortical explant cultures and isolated neural stem cells [66]. In addition, the choroid plexus is also known to secrete IGF binding proteins (IGFBPs), which assist in the transportation of IGFs and also are believed to modulate their activity [143]. In adulthood, it is thought that IGF2 levels in the CNS remain relatively stable [140, 141]. However, evidence suggests that IGF2 may be an important regulatory molecule released in a penetrating model of TBI [115]. Walter and colleagues exposed adult rats to a cortical penetrating incision and reported that IGF2 levels increase within the CSF, peaking at day 7 post-penetrating TBI. Importantly, between 1–7 days post injury, IGF2 protein localizes to the cells within the damaged tissue. Walter et al. also determined that IGFBPs increase in the CSF during that time period, with a predominant increase in IGFBP-2, which is thought to be one of the primary transporters of IGF2 to its target receptor and regulator of its activity [144]. After day 7 post-injury, during the chronic phase of wound recovery, IGF2 levels in the CSF decline with a relative increase in IGFBP-5, thought to be responsible for sequestering and downregulating IGF2 activity, suggesting a possible biphasic response by IGF2 in CNS injury. Throughout adulthood, IGF2 secretion into the CSF may be stimulated by TBI or any injury to the CNS. IGF2 may be an important acute phase protein in TBI for assisting with wound healing through its stimulation of neurogenesis and neuroprotection, whose activity appears to be modulated by concomitant secretion of distinct IGFBPs.

A biphasic response to CNS injury has also been shown for the hormone-like peptide augurin, encoded by the esophageal cancer related gene-4 (Ecrg4) gene [145, 146]. The Ecrg4 gene has been shown to be a novel tumor suppressor gene [147, 148]. Its protein product, augurin, inhibits proliferation and has been implicated in cell senescence in aged oligodendrocyte precursor cells [149]. In CNS injury, augurin is believed to act as an acute-phase respondent [145, 146]. In studies by Podvin et al. and Gonzalez et al., augurin localizes to the apical membrane of the choroid plexus, is secreted or cleaved from the membrane, and is released into the CSF. Upon penetrating cortical injury in adult rats, Ecrg4 gene expression and augurin immunoreactivity decrease in the choroid plexus 1–3 days post-injury, returning to normal levels by day 7. Importantly, overexpression of the Ecrg4 gene using an adenovirus vector injected into the lateral ventricle leads to decreased bromodeoxyuridine (BrdU) staining along the SVZ of injured rats. Although changes in augurin immunoreactivity in the CSF were not assayed, the authors speculate that augurin undergoes proteolytic cleavage and is released by choroid plexus epithelial cells in response to brain injury. This release depletes augurin from the choroid plexus and CSF, leading to increased cell proliferation for an acute period of time after CNS injury before stores of augurin are repleted by day 7. In this manner, augurin is speculated to participate in the acute phase of wound recovery by dis-inhibiting neural stems cells and promoting their proliferation, a function mediated by CSF-based signaling [145, 146].

Taken together, it appears that one key mechanism for promoting neural repair following TBI is through the endogenous upregulation of neurotrophic factors important in neurogenesis and neuronal survival. An increasing number of growth factors are being discovered in the CSF in response to brain injury and the mechanism of action is slowly being elucidated (Table 2). The CSF may act as a vehicle for volume transmission and transportation of important nutrients and growth factors to sites of brain injury and globally to the CNS to help maintain homeostasis, promote neuroprotection and neurogenesis, and for the removal of toxic by-products generated by the injury.

The potential therapeutics of modulating the CSF in brain injury

Several studies have directly tested the possibility of inducing neurogenesis or promoting neuroprotection by directly infusing known neurogenic growth factors into the ventricles of animal brain injury models. For instance, Sun et al. [150] infused FGF intraventricularly for 1-week post-TBI, which resulted in increased cell proliferation in neurogenic zones of the SVZ and dentate gyrus (DG) and increased survival of newly generated neurons. These favorable effects of FGF correlate with improved water maze testing at 2–25 days post-TBI, suggesting a possible link between increased neurogenesis and memory [150]. Intraventricular administration of FGF2 also has neuroprotective effects in rat models of focal cerebral ischemia. FGF infusion again increases the numbers of proliferating cells in the SVZ and DG [151], and in some models has actually been reported to decrease infarct size [152]. These observations underscore an interesting connection between FGF2 and CSF since CSF–FGF regulates neurogenesis in the mesencephalon of the developing chick brain [67]. In addition to FGF, intraventricular EGF administration in rats post-TBI (via a fluid percussion injury) also appears to display increased proliferation in SVZ and DG at 1 week post-TBI with improved cognitive functions at 4 weeks post-TBI [153]. Interestingly, NGF infused into the lateral ventricle decreases spatial memory deficits post cortical impact injury as measured by the Morris water maze, an effect suggested to be due to the prevention of cholinergic deficits often seen after TBI [154]. The exact molecular mechanisms mediating these behavioral improvements remain to be elucidated.

Numerous other neurotrophic factors including VEGF, GDNF, and BDNF, have been infused into the CSF to assess their ability to promote neurogenesis and neuroprotection, and to determine if cognitive functioning can be improved following injury [155161]. In synergistic studies, EGF and erythropoietin infused intraventricularly promote regeneration of damaged cortex and improve motor tasks following focal cerebral ischemia [162]. By a cell-tracking experimental approach using β-galactosidase-labeled SVZ cells, on day 11 post-stroke, 10% of the labeled cells were within the lesioned area, and 55% of the labeled cells were seen migrating from the SVZ. In the same study, on day 18 post injury, 11% of the BrdU-labeled cells in the newly regenerated tissue were also NeuN positive, a marker for neurons, suggesting that a proportion of the newly differentiated neuronal cells in the regenerated cortex appeared to originate from neural stem cells in the forebrain SVZ [162]. This capacity of endogenous neural stem cells to migrate to the site of CNS damage has been demonstrated by other groups as well, and is thought to be mediated in part by the Stem Cell Factor SCF/c-kit pathway [163]. A growing body of evidence in animal models of TBI and stroke suggests that modulating the CSF environment, and therefore creating an enhanced neurogenic and neuroprotective niche, stimulates endogenous neural stem cell proliferation as well as migration, contributing to improved cognitive functions. However, it remains to be seen how effectively these newly generated neurons establish long-term connections in existing brain circuitry.

Although the majority of studies in animal models have been performed with intraventricularly infused growth factors, recent reports suggest that neurotrophic agents can be intranasally administered, with positive effects on the CNS [164171]. The route for the delivery of growth factors from the nasal epithelium to the CNS is poorly understood. In one model, olfactory and trigeminal nerves transport factors to the olfactory bulb and brainstem, from where they distribute throughout the CNS, including into the CSF [172]. This mode of delivery has been suggested for IGF1 [171]. Intranasal IGF1 has also been suggested to decrease infarct size and improve neurological functioning in animal models of focal ischemic brain injury [173, 174]. Intranasal FGF administered following focal cerebral ischemia in rats has been reported to increase neurogenesis in the SVZ and DG, as well as improve behavioral recovery [165]. Recently in human subjects, it was shown that intranasal insulin penetrates the blood–brain barrier, leading to CSF access [175]. Intranasal insulin was also reported to improve memory and cognitive functioning in patients with mild Alzheimer’s disease [166, 169, 170]. Although not all studies assayed for CSF levels of proteins following intranasal administration, many have been shown to be delivered to the CSF, including insulin, exendin, orexin-A, calcitonin gene-related peptide, galanin-like peptide, leptin, melanocortin, and arginine-vasopressin [167].

The possibility of simultaneously administering a cocktail of neurogenic and neuroprotective factors to the CNS provides an intriguing, non-invasive method that is particularly exciting from the therapeutic point of view. The CSF contains a plethora of proteins in addition to growth factors, including proteases, antioxidants, small molecules, and hormones whose function in the CSF is not fully understood [4, 6, 59, 60, 176]. A recent study also showed that media conditioned with choroid plexus supports neural stem cell proliferation compared to media conditioned with cortical tissue [66]. Interestingly, in newborns with hypoxic brain damage, increased levels of antioxidants within the CSF provide neuronal protection [177]. Together, these observations raise the question of whether manipulating endogenous choroid plexus, or injecting or implanting choroid plexus intraventricularly or at the site of brain damage, may provide essential neurogenic growth factors, as well as other proteins necessary to enhance CSF homeostasis. Studies with transplanted choroid plexus in models of focal CNS ischemia have reported reduced infarct size and improved neurological functioning compared to controls [178], with ELISA studies confirming the secretion of GDNF, BDNF, and NGF from the choroid plexus [179]. In these studies, implanted choroid plexus provides essentially an entire cocktail of factors to the injured CNS, enhancing recovery, neuroprotection, and neurogenesis. Although still in its infancy, genetically modifying one’s own choroid plexus for secreting distinct peptides or proteins into the CSF offers an intriguing future possibility for CNS therapy [180].

Finally, the CSF is a potential vehicle for volume transmission of implanted neural stem cells, which to date have been used in animal models of spinal cord and brain injury [111, 181183]. Surprisingly, in an animal model of Alzheimer’s disease in which kainic acid was used to trigger neuronal damage to the hippocampal CA3 region, neural stem cells injected within the ventricles were reported to migrate to the site of injury, differentiate into neurons and astrocytes, and improve cognitive functioning [184]. A similar effect has been observed in an animal model of spinal cord injury, in which neural stem cells injected into the 4th ventricle CSF migrated to the site of injury [181, 182]. A significant limitation to the therapeutic potential is the necessity of intraventricular, intracerebral, or intraspinal injections. It has been suggested that neural stem cells may also be delivered to the CNS intranasally, however the mechanisms underlying these effects remain unclear.

Conclusions

From the early Egyptians to today, the CSF has piqued the curiosity of many. Once thought to control muscle contractions, to carry a certain pneuma or spirit, or to simply be a post-mortem precipitate, the diverse and active roles of the CSF in the developing and adult CNS are finally unfolding. CSF components, which can potentially be dispersed over large areas, may be more significant and pervasive regulators of cortical development, neural stem cell renewal, disease, neurodegeneration, behavior, sleep/wake cycles and other states of consciousness than previously thought. Cells lining the neural tube during development are responsive to CSF signals governing neurogenesis. In the mature brain, cells with access to the ventricles may similarly respond to factors released into the CSF following brain injury, neurodegenerative diseases, aging, as well as daily changes in behavior and activity. The CSF may thus serve as a vehicle for immediate signaling to major control centers of the brain. As such, using the CSF as a therapeutic vehicle to promote CNS homeostasis has many potential benefits. Although the field is still in its infancy, developing strategies to selectively target compounds to the CSF and other areas of the CNS, should open avenues to major therapeutic breakthroughs for TBI, stroke, neurodegeneration, aging, and beyond.

Acknowledgments

We thank V. Ho and A. Malesz for critical reading of this manuscript. We are grateful for support from the Eleanor and Miles Shore Fellowship Program for Scholars in Medicine/Children’s Hospital Boston Career Development Award, and the NIH (Award number R00 NS072192). M.K.L. is an Alfred P. Sloan Research Fellow.

Abbreviations

BDNF

Brain-derived neurotrophic factor

BMI

Body mass index

BMP

Bone morphogenetic protein

Bmpr1a

BMP receptor 1a

BrdU

Bromodeoxyuridine

CNS

Central nervous system

ChP

Choroid plexus

CSF

Cerebrospinal fluid

DG

Dentate gyrus

ELISA

Enzyme-linked immunosorbent assay

ERK1/2

Extracellular signal-regulated kinase 1/2

Ecrg4

Esophageal cancer-related gene-4

FGF

Fibroblast growth factors

GDF

Growth differentiation factors

GDNF

Glial-derived neurotrophic factor

IGF

Insulin-like growth factors

IGF1R

IGF1 receptor

IGFBPs

IGF-binding proteins

Irs2

Insulin receptor substrate 2

LIF

Leukemia inhibiting factor

Lmx1a

LIM homeobox transcription factor 1, alpha

LV

Lateral ventricle

MS

Mass spectrometry

NEP

Neuroepithelium

NGF

Nerve growth factor

NT-3

Neurotrophin 3

Pdk1

Phosphoinositide-dependent kinase-1

PEDF

Pigment epithelial derived factor

Pten

Phosphatase and tensin homolog

RA

Retinoic acid

Rhomb

Rhombencephalon

sAPP

Soluble amyloid precursor protein

Shh

Sonic hedgehog

SCN

Suprachiasmatic nucleus

SVZ

Subventricular zone

Tel

Telencephalon

TGF

Transforming growth factor

TNF

Tumor necrosis factor

TBI

Traumatic brain injury

V

Ventricle

VEGF

Vascular endothelial growth factor

VZ

Ventricular zone

Wnt1

Wingless-type MMTV integration site family, member 1

References

  • 1.Liddelow SA. Fluids and barriers of the CNS: a historical viewpoint. Fluids Barriers CNS. 2011;8(1):2. doi: 10.1186/2045-8118-8-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Woollam DH. The historical significance of the cerebrospinal fluid. Med Hist. 1957;1(2):91–114. doi: 10.1017/s0025727300021025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Dohrmann GJ. The choroid plexus: a historical review. Brain Res. 1970;18(2):197–218. doi: 10.1016/0006-8993(70)90324-0. [DOI] [PubMed] [Google Scholar]
  • 4.Chodobski A, Szmydynger-Chodobska J. Choroid plexus: target for polypeptides and site of their synthesis. Microsc Res Tech. 2001;52(1):65–82. doi: 10.1002/1097-0029(20010101)52:1<65::AID-JEMT9>3.0.CO;2-4. [DOI] [PubMed] [Google Scholar]
  • 5.Lehtinen MK, Walsh CA. Neurogenesis at the brain-cerebrospinal fluid interface. Annu Rev Cell Dev Biol. 2011;27:653–679. doi: 10.1146/annurev-cellbio-092910-154026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Redzic ZB, Preston JE, Duncan JA, Chodobski A, Szmydynger-Chodobska J. The choroid plexus-cerebrospinal fluid system: from development to aging. Curr Top Dev Biol. 2005;71:1–52. doi: 10.1016/S0070-2153(05)71001-2. [DOI] [PubMed] [Google Scholar]
  • 7.Bailey P. Morphology of the roofplate of the forebrain and the lateral choroid plexuses in the human embryo. J. Comp. Neurol. 1916;26:79–120. doi: 10.1002/cne.900260104. [DOI] [Google Scholar]
  • 8.Sturrock RR. A morphological study of the development of the mouse choroid plexus. J Anat. 1979;129(Pt 4):777–793. [PMC free article] [PubMed] [Google Scholar]
  • 9.Kappers JA. The development of the paraphysis cerebri in man with comments on its relationship to the intercolumnar tubercle and its significance for the origin of cystic tumors in the third ventricle. J Comp Neurol. 1955;102(2):425–509. doi: 10.1002/cne.901020205. [DOI] [PubMed] [Google Scholar]
  • 10.Bayer SA, Altman J (2007) Atlas of human central nervous system development. vol 5, CRC Press, Boca Raton
  • 11.von Frowein J, Wizenmann A, Gotz M. The transcription factors Emx1 and Emx2 suppress choroid plexus development and promote neuroepithelial cell fate. Dev Biol. 2006;296(1):239–252. doi: 10.1016/j.ydbio.2006.04.461. [DOI] [PubMed] [Google Scholar]
  • 12.Johanson CE, Stopa EG, McMillan PN. The blood-cerebrospinal fluid barrier: structure and functional significance. Methods Mol Biol. 2011;686:101–131. doi: 10.1007/978-1-60761-938-3_4. [DOI] [PubMed] [Google Scholar]
  • 13.Johansson PA, Dziegielewska KM, Liddelow SA, Saunders NR. The blood-CSF barrier explained: when development is not immaturity. BioEssays. 2008;30(3):237–248. doi: 10.1002/bies.20718. [DOI] [PubMed] [Google Scholar]
  • 14.Awatramani R, Soriano P, Rodriguez C, Mai JJ, Dymecki SM. Cryptic boundaries in roof plate and choroid plexus identified by intersectional gene activation. Nat Genet. 2003;35(1):70–75. doi: 10.1038/ng1228. [DOI] [PubMed] [Google Scholar]
  • 15.Chizhikov VV, Lindgren AG, Mishima Y, Roberts RW, Aldinger KA, Miesegaes GR, Currle DS, Monuki ES, Millen KJ. Lmx1a regulates fates and location of cells originating from the cerebellar rhombic lip and telencephalic cortical hem. Proc Natl Acad Sci U S A. 2010;107(23):10725–10730. doi: 10.1073/pnas.0910786107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Currle DS, Cheng X, Hsu CM, Monuki ES. Direct and indirect roles of CNS dorsal midline cells in choroid plexus epithelia formation. Development. 2005;132(15):3549–3559. doi: 10.1242/dev.01915. [DOI] [PubMed] [Google Scholar]
  • 17.Landsberg RL, Awatramani RB, Hunter NL, Farago AF, DiPietrantonio HJ, Rodriguez CI, Dymecki SM. Hindbrain rhombic lip is comprised of discrete progenitor cell populations allocated by Pax6. Neuron. 2005;48(6):933–947. doi: 10.1016/j.neuron.2005.11.031. [DOI] [PubMed] [Google Scholar]
  • 18.Huang X, Ketova T, Fleming JT, Wang H, Dey SK, Litingtung Y, Chiang C. Sonic hedgehog signaling regulates a novel epithelial progenitor domain of the hindbrain choroid plexus. Development. 2009;136(15):2535–2543. doi: 10.1242/dev.033795. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Nielsen CM, Dymecki SM. Sonic hedgehog is required for vascular outgrowth in the hindbrain choroid plexus. Dev Biol. 2010;340(2):430–437. doi: 10.1016/j.ydbio.2010.01.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Hunter NL, Dymecki SM. Molecularly and temporally separable lineages form the hindbrain roof plate and contribute differentially to the choroid plexus. Development. 2007;134(19):3449–3460. doi: 10.1242/dev.003095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Hebert JM, Mishina Y, McConnell SK. BMP signaling is required locally to pattern the dorsal telencephalic midline. Neuron. 2002;35(6):1029–1041. doi: 10.1016/S0896-6273(02)00900-5. [DOI] [PubMed] [Google Scholar]
  • 22.Cheng X, Hsu CM, Currle DS, Hu JS, Barkovich AJ, Monuki ES. Central roles of the roof plate in telencephalic development and holoprosencephaly. J Neurosci. 2006;26(29):7640–7649. doi: 10.1523/JNEUROSCI.0714-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Cushing H. Studies on the Cerebro-Spinal Fluid : I. Introduction. J Med Res. 1914;31(1):1–19. [PMC free article] [PubMed] [Google Scholar]
  • 24.Desmond ME, Jacobson AG. Embryonic brain enlargement requires cerebrospinal fluid pressure. Dev Biol. 1977;57(1):188–198. doi: 10.1016/0012-1606(77)90364-5. [DOI] [PubMed] [Google Scholar]
  • 25.Lowery LA, Sive H. Totally tubular: the mystery behind function and origin of the brain ventricular system. BioEssays. 2009;31(4):446–458. doi: 10.1002/bies.200800207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Pexieder T, Jelinek R. Pressure of the CSF and the morphogenesis of the CNS. II. Pressure necessary for normal development of brain vesicles. Folia Morphol (Praha) 1970;18(2):181–192. [PubMed] [Google Scholar]
  • 27.Desmond ME, Levitan ML, Haas AR. Internal luminal pressure during early chick embryonic brain growth: descriptive and empirical observations. Anat Rec A Discov Mol Cell Evol Biol. 2005;285(2):737–747. doi: 10.1002/ar.a.20211. [DOI] [PubMed] [Google Scholar]
  • 28.Mashayekhi F, Draper CE, Bannister CM, Pourghasem M, Owen-Lynch PJ, Miyan JA. Deficient cortical development in the hydrocephalic Texas (H-Tx) rat: a role for CSF. Brain. 2002;125(Pt 8):1859–1874. doi: 10.1093/brain/awf182. [DOI] [PubMed] [Google Scholar]
  • 29.Owen-Lynch PJ, Draper CE, Mashayekhi F, Bannister CM, Miyan JA. Defective cell cycle control underlies abnormal cortical development in the hydrocephalic Texas rat. Brain. 2003;126(Pt 3):623–631. doi: 10.1093/brain/awg058. [DOI] [PubMed] [Google Scholar]
  • 30.Pappenheimer JR, Miller TB, Goodrich CA. Sleep-promoting effects of cerebrospinal fluid from sleep-deprived goats. Proc Natl Acad Sci U S A. 1967;58(2):513–517. doi: 10.1073/pnas.58.2.513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Lerner RA, Siuzdak G, Prospero-Garcia O, Henriksen SJ, Boger DL, Cravatt BF. Cerebrodiene: a brain lipid isolated from sleep-deprived cats. Proc Natl Acad Sci U S A. 1994;91(20):9505–9508. doi: 10.1073/pnas.91.20.9505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Cravatt BF, Prospero-Garcia O, Siuzdak G, Gilula NB, Henriksen SJ, Boger DL, Lerner RA. Chemical characterization of a family of brain lipids that induce sleep. Science. 1995;268(5216):1506–1509. doi: 10.1126/science.7770779. [DOI] [PubMed] [Google Scholar]
  • 33.Basile AS, Hanus L, Mendelson WB. Characterization of the hypnotic properties of oleamide. NeuroReport. 1999;10(5):947–951. doi: 10.1097/00001756-199904060-00010. [DOI] [PubMed] [Google Scholar]
  • 34.Mendelson WB, Basile AS. The hypnotic actions of oleamide are blocked by a cannabinoid receptor antagonist. NeuroReport. 1999;10(15):3237–3239. doi: 10.1097/00001756-199910190-00021. [DOI] [PubMed] [Google Scholar]
  • 35.Laposky AD, Homanics GE, Basile A, Mendelson WB. Deletion of the GABA(A) receptor beta 3 subunit eliminates the hypnotic actions of oleamide in mice. NeuroReport. 2001;12(18):4143–4147. doi: 10.1097/00001756-200112210-00056. [DOI] [PubMed] [Google Scholar]
  • 36.Leggett JD, Aspley S, Beckett SR, D’Antona AM, Kendall DA. Oleamide is a selective endogenous agonist of rat and human CB1 cannabinoid receptors. Br J Pharmacol. 2004;141(2):253–262. doi: 10.1038/sj.bjp.0705607. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Mendelson WB, Basile AS. The hypnotic actions of the fatty acid amide, oleamide. Neuropsychopharmacology. 2001;25(5 Suppl):S36–S39. doi: 10.1016/S0893-133X(01)00341-4. [DOI] [PubMed] [Google Scholar]
  • 38.Berridge CW, Espana RA, Vittoz NM. Hypocretin/orexin in arousal and stress. Brain Res. 2010;1314:91–102. doi: 10.1016/j.brainres.2009.09.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Chemelli RM, Willie JT, Sinton CM, Elmquist JK, Scammell T, Lee C, Richardson JA, Williams SC, Xiong Y, Kisanuki Y, Fitch TE, Nakazato M, Hammer RE, Saper CB, Yanagisawa M. Narcolepsy in orexin knockout mice: molecular genetics of sleep regulation. Cell. 1999;98(4):437–451. doi: 10.1016/S0092-8674(00)81973-X. [DOI] [PubMed] [Google Scholar]
  • 40.Lin L, Faraco J, Li R, Kadotani H, Rogers W, Lin X, Qiu X, de Jong PJ, Nishino S, Mignot E. The sleep disorder canine narcolepsy is caused by a mutation in the hypocretin (orexin) receptor 2 gene. Cell. 1999;98(3):365–376. doi: 10.1016/S0092-8674(00)81965-0. [DOI] [PubMed] [Google Scholar]
  • 41.Dauvilliers Y, Baumann CR, Carlander B, Bischof M, Blatter T, Lecendreux M, Maly F, Besset A, Touchon J, Billiard M, Tafti M, Bassetti CL. CSF hypocretin-1 levels in narcolepsy, Kleine-Levin syndrome, and other hypersomnias and neurological conditions. J Neurol Neurosurg Psychiatry. 2003;74(12):1667–1673. doi: 10.1136/jnnp.74.12.1667. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Bourgin P, Huitron-Resendiz S, Spier AD, Fabre V, Morte B, Criado JR, Sutcliffe JG, Henriksen SJ, de Lecea L. Hypocretin-1 modulates rapid eye movement sleep through activation of locus coeruleus neurons. J Neurosci. 2000;20(20):7760–7765. doi: 10.1523/JNEUROSCI.20-20-07760.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Hagan JJ, Leslie RA, Patel S, Evans ML, Wattam TA, Holmes S, Benham CD, Taylor SG, Routledge C, Hemmati P, Munton RP, Ashmeade TE, Shah AS, Hatcher JP, Hatcher PD, Jones DN, Smith MI, Piper DC, Hunter AJ, Porter RA, Upton N. Orexin A activates locus coeruleus cell firing and increases arousal in the rat. Proc Natl Acad Sci U S A. 1999;96(19):10911–10916. doi: 10.1073/pnas.96.19.10911. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Piper DC, Upton N, Smith MI, Hunter AJ. The novel brain neuropeptide, orexin-A, modulates the sleep-wake cycle of rats. Eur J Neurosci. 2000;12(2):726–730. doi: 10.1046/j.1460-9568.2000.00919.x. [DOI] [PubMed] [Google Scholar]
  • 45.Espana RA, Baldo BA, Kelley AE, Berridge CW. Wake-promoting and sleep-suppressing actions of hypocretin (orexin): basal forebrain sites of action. Neuroscience. 2001;106(4):699–715. doi: 10.1016/S0306-4522(01)00319-0. [DOI] [PubMed] [Google Scholar]
  • 46.Silver R, LeSauter J, Tresco PA, Lehman MN. A diffusible coupling signal from the transplanted suprachiasmatic nucleus controlling circadian locomotor rhythms. Nature. 1996;382(6594):810–813. doi: 10.1038/382810a0. [DOI] [PubMed] [Google Scholar]
  • 47.Kramer A, Yang FC, Snodgrass P, Li X, Scammell TE, Davis FC, Weitz CJ. Regulation of daily locomotor activity and sleep by hypothalamic EGF receptor signaling. Science. 2001;294(5551):2511–2515. doi: 10.1126/science.1067716. [DOI] [PubMed] [Google Scholar]
  • 48.Kraves S, Weitz CJ. A role for cardiotrophin-like cytokine in the circadian control of mammalian locomotor activity. Nat Neurosci. 2006;9(2):212–219. doi: 10.1038/nn1633. [DOI] [PubMed] [Google Scholar]
  • 49.Reiter RJ, Tan DX, Poeggeler B, Menendez-Pelaez A, Chen LD, Saarela S. Melatonin as a free radical scavenger: implications for aging and age-related diseases. Ann N Y Acad Sci. 1994;719:1–12. doi: 10.1111/j.1749-6632.1994.tb56817.x. [DOI] [PubMed] [Google Scholar]
  • 50.Tricoire H, Locatelli A, Chemineau P, Malpaux B. Melatonin enters the cerebrospinal fluid through the pineal recess. Endocrinology. 2002;143(1):84–90. doi: 10.1210/en.143.1.84. [DOI] [PubMed] [Google Scholar]
  • 51.Tan DX, Manchester LC, Sanchez-Barcelo E, Mediavilla MD, Reiter RJ. Significance of high levels of endogenous melatonin in Mammalian cerebrospinal fluid and in the central nervous system. Curr Neuropharmacol. 2010;8(3):162–167. doi: 10.2174/157015910792246182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Seifman MA, Adamides AA, Nguyen PN, Vallance SA, Cooper DJ, Kossmann T, Rosenfeld JV, Morganti-Kossmann MC. Endogenous melatonin increases in cerebrospinal fluid of patients after severe traumatic brain injury and correlates with oxidative stress and metabolic disarray. J Cereb Blood Flow Metab. 2008;28(4):684–696. doi: 10.1038/sj.jcbfm.9600603. [DOI] [PubMed] [Google Scholar]
  • 53.Martin FH, Seoane JR, Baile CA. Feeding in satiated sheep elicited by intraventricular injections of CSF from fasted sheep. Life Sci. 1973;13(2):177–184. doi: 10.1016/0024-3205(73)90193-8. [DOI] [PubMed] [Google Scholar]
  • 54.Woods SC, Lotter EC, McKay LD, Porte D., Jr Chronic intracerebroventricular infusion of insulin reduces food intake and body weight of baboons. Nature. 1979;282(5738):503–505. doi: 10.1038/282503a0. [DOI] [PubMed] [Google Scholar]
  • 55.Schwartz MW, Peskind E, Raskind M, Boyko EJ, Porte D., Jr Cerebrospinal fluid leptin levels: relationship to plasma levels and to adiposity in humans. Nat Med. 1996;2(5):589–593. doi: 10.1038/nm0596-589. [DOI] [PubMed] [Google Scholar]
  • 56.Sawamoto K, Wichterle H, Gonzalez-Perez O, Cholfin JA, Yamada M, Spassky N, Murcia NS, Garcia-Verdugo JM, Marin O, Rubenstein JL, Tessier-Lavigne M, Okano H, Alvarez-Buylla A. New neurons follow the flow of cerebrospinal fluid in the adult brain. Science. 2006;311(5761):629–632. doi: 10.1126/science.1119133. [DOI] [PubMed] [Google Scholar]
  • 57.Cavanagh ME, Cornelis ME, Dziegielewska KM, Evans CA, Lorscheider FL, Mollgard K, Reynolds ML, Saunders NR. Comparison of proteins in CSF of lateral and IVth ventricles during early development of fetal sheep. Brain Res. 1983;313(2):159–167. doi: 10.1016/0165-3806(83)90213-4. [DOI] [PubMed] [Google Scholar]
  • 58.Dziegielewska KM, Evans CA, Lai PC, Lorscheider FL, Malinowska DH, Mollgard K, Saunders NR. Proteins in cerebrospinal fluid and plasma of fetal rats during development. Dev Biol. 1981;83(1):193–200. doi: 10.1016/S0012-1606(81)80024-3. [DOI] [PubMed] [Google Scholar]
  • 59.Parada C, Gato A, Bueno D. Mammalian embryonic cerebrospinal fluid proteome has greater apolipoprotein and enzyme pattern complexity than the avian proteome. J Proteome Res. 2005;4(6):2420–2428. doi: 10.1021/pr050213t. [DOI] [PubMed] [Google Scholar]
  • 60.Zappaterra MD, Lisgo SN, Lindsay S, Gygi SP, Walsh CA, Ballif BA. A comparative proteomic analysis of human and rat embryonic cerebrospinal fluid. J Proteome Res. 2007;6(9):3537–3548. doi: 10.1021/pr070247w. [DOI] [PubMed] [Google Scholar]
  • 61.Zheng, W, Chodobski A (2005) The blood-cerebrospinal fluid barrier. Taylor & Francis, Boca Raton
  • 62.Grove EA, Fukuchi-Shimogori T. Generating the cerebral cortical area map. Annu Rev Neurosci. 2003;26:355–380. doi: 10.1146/annurev.neuro.26.041002.131137. [DOI] [PubMed] [Google Scholar]
  • 63.Wolpert L. One hundred years of positional information. Trends Genet. 1996;12(9):359–364. doi: 10.1016/S0168-9525(96)80019-9. [DOI] [PubMed] [Google Scholar]
  • 64.Chenn A, Zhang YA, Chang BT, McConnell SK. Intrinsic polarity of mammalian neuroepithelial cells. Mol Cell Neurosci. 1998;11(4):183–193. doi: 10.1006/mcne.1998.0680. [DOI] [PubMed] [Google Scholar]
  • 65.Toyoda R, Assimacopoulos S, Wilcoxon J, Taylor A, Feldman P, Suzuki-Hirano A, Shimogori T, Grove EA. FGF8 acts as a classic diffusible morphogen to pattern the neocortex. Development. 2010;137(20):3439–3448. doi: 10.1242/dev.055392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Lehtinen MK, Zappaterra MW, Chen X, Yang YJ, Hill AD, Lun M, Maynard T, Gonzalez D, Kim S, Ye P, D’Ercole AJ, Wong ET, LaMantia AS, Walsh CA. The cerebrospinal fluid provides a proliferative niche for neural progenitor cells. Neuron. 2011;69(5):893–905. doi: 10.1016/j.neuron.2011.01.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Martin C, Bueno D, Alonso MI, Moro JA, Callejo S, Parada C, Martin P, Carnicero E, Gato A. FGF2 plays a key role in embryonic cerebrospinal fluid trophic properties over chick embryo neuroepithelial stem cells. Dev Biol. 2006;297(2):402–416. doi: 10.1016/j.ydbio.2006.05.010. [DOI] [PubMed] [Google Scholar]
  • 68.Salehi Z, Mashayekhi F, Naji M, Pandamooz S. Insulin-like growth factor-1 and insulin-like growth factor binding proteins in cerebrospinal fluid during the development of mouse embryos. J Clin Neurosci. 2009;16(7):950–953. doi: 10.1016/j.jocn.2008.09.018. [DOI] [PubMed] [Google Scholar]
  • 69.Huang X, Liu J, Ketova T, Fleming JT, Grover VK, Cooper MK, Litingtung Y, Chiang C. Transventricular delivery of Sonic hedgehog is essential to cerebellar ventricular zone development. Proc Natl Acad Sci U S A. 2010;107(18):8422–8427. doi: 10.1073/pnas.0911838107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Parada C, Gato A, Bueno D. All-trans retinol and retinol-binding protein from embryonic cerebrospinal fluid exhibit dynamic behaviour during early central nervous system development. NeuroReport. 2008;19(9):945–950. doi: 10.1097/WNR.0b013e3283021c94. [DOI] [PubMed] [Google Scholar]
  • 71.Mashayekhi F, Azari M, Moghadam LM, Yazdankhah M, Naji M, Salehi Z. Changes in cerebrospinal fluid nerve growth factor levels during chick embryonic development. J Clin Neurosci. 2009;16(10):1334–1337. doi: 10.1016/j.jocn.2009.03.023. [DOI] [PubMed] [Google Scholar]
  • 72.Hatta T, Matsumoto A, Ono A, Udagawa J, Nimura M, Hashimoto R, Otani H. Quantitative analyses of leukemia inhibitory factor in the cerebrospinal fluid in mouse embryos. NeuroReport. 2006;17(18):1863–1866. doi: 10.1097/WNR.0b013e3280113962. [DOI] [PubMed] [Google Scholar]
  • 73.Raballo R, Rhee J, Lyn-Cook R, Leckman JF, Schwartz ML, Vaccarino FM. Basic fibroblast growth factor (Fgf2) is necessary for cell proliferation and neurogenesis in the developing cerebral cortex. J Neurosci. 2000;20(13):5012–5023. doi: 10.1523/JNEUROSCI.20-13-05012.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Tao Y, Black IB, DiCicco-Bloom E. In vivo neurogenesis is inhibited by neutralizing antibodies to basic fibroblast growth factor. J Neurobiol. 1997;33(3):289–296. doi: 10.1002/(SICI)1097-4695(199709)33:3<289::AID-NEU7>3.0.CO;2-Y. [DOI] [PubMed] [Google Scholar]
  • 75.Holm NR, Hansen LB, Nilsson C, Gammeltoft S. Gene expression and secretion of insulin-like growth factor-II and insulin-like growth factor binding protein-2 from cultured sheep choroid plexus epithelial cells. Brain Res Mol Brain Res. 1994;21(1–2):67–74. doi: 10.1016/0169-328X(94)90379-4. [DOI] [PubMed] [Google Scholar]
  • 76.Margolis RU, Altszuler N. Insulin in the cerebrospinal fluid. Nature. 1967;215(5108):1375–1376. doi: 10.1038/2151375a0. [DOI] [PubMed] [Google Scholar]
  • 77.Baker J, Liu JP, Robertson EJ, Efstratiadis A. Role of insulin-like growth factors in embryonic and postnatal growth. Cell. 1993;75(1):73–82. [PubMed] [Google Scholar]
  • 78.DeChiara TM, Robertson EJ, Efstratiadis A. Parental imprinting of the mouse insulin-like growth factor II gene. Cell. 1991;64(4):849–859. doi: 10.1016/0092-8674(91)90513-X. [DOI] [PubMed] [Google Scholar]
  • 79.Sutter NB, Bustamante CD, Chase K, Gray MM, Zhao K, Zhu L, Padhukasahasram B, Karlins E, Davis S, Jones PG, Quignon P, Johnson GS, Parker HG, Fretwell N, Mosher DS, Lawler DF, Satyaraj E, Nordborg M, Lark KG, Wayne RK, Ostrander EA. A single IGF1 allele is a major determinant of small size in dogs. Science. 2007;316(5821):112–115. doi: 10.1126/science.1137045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Weber MM, Melmed S, Rosenbloom J, Yamasaki H, Prager D. Rat somatotroph insulin-like growth factor-II (IGF-II) signaling: role of the IGF-I receptor. Endocrinology. 1992;131(5):2147–2153. doi: 10.1210/en.131.5.2147. [DOI] [PubMed] [Google Scholar]
  • 81.Kappeler L, De Magalhaes Filho C, Dupont J, Leneuve P, Cervera P, Perin L, Loudes C, Blaise A, Klein R, Epelbaum J, Le Bouc Y, Holzenberger M. Brain IGF-1 receptors control mammalian growth and lifespan through a neuroendocrine mechanism. PLoS Biol. 2008;6(10):e254. doi: 10.1371/journal.pbio.0060254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Liu W, Ye P, O’Kusky JR, D’Ercole AJ. Type 1 insulin-like growth factor receptor signaling is essential for the development of the hippocampal formation and dentate gyrus. J Neurosci Res. 2009;87(13):2821–2832. doi: 10.1002/jnr.22129. [DOI] [PubMed] [Google Scholar]
  • 83.Hodge RD, D’Ercole AJ, O’Kusky JR. Insulin-like growth factor-I accelerates the cell cycle by decreasing G1 phase length and increases cell cycle reentry in the embryonic cerebral cortex. J Neurosci. 2004;24(45):10201–10210. doi: 10.1523/JNEUROSCI.3246-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Mairet-Coello G, Tury A, DiCicco-Bloom E. Insulin-like growth factor-1 promotes G(1)/S cell cycle progression through bidirectional regulation of cyclins and cyclin-dependent kinase inhibitors via the phosphatidylinositol 3-kinase/Akt pathway in developing rat cerebral cortex. J Neurosci. 2009;29(3):775–788. doi: 10.1523/JNEUROSCI.1700-08.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Popken GJ, Hodge RD, Ye P, Zhang J, Ng W, O’Kusky JR, D’Ercole AJ. In vivo effects of insulin-like growth factor-I (IGF-I) on prenatal and early postnatal development of the central nervous system. Eur J Neurosci. 2004;19(8):2056–2068. doi: 10.1111/j.0953-816X.2004.03320.x. [DOI] [PubMed] [Google Scholar]
  • 86.Joseph D’Ercole A, Ye P. Expanding the mind: insulin-like growth factor I and brain development. Endocrinology. 2008;149(12):5958–5962. doi: 10.1210/en.2008-0920. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Schubert M, Brazil DP, Burks DJ, Kushner JA, Ye J, Flint CL, Farhang-Fallah J, Dikkes P, Warot XM, Rio C, Corfas G, White MF. Insulin receptor substrate-2 deficiency impairs brain growth and promotes tau phosphorylation. J Neurosci. 2003;23(18):7084–7092. doi: 10.1523/JNEUROSCI.23-18-07084.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Chalhoub N, Zhu G, Zhu X, Baker SJ. Cell type specificity of PI3K signaling in Pdk1- and Pten-deficient brains. Genes Dev. 2009;23(14):1619–1624. doi: 10.1101/gad.1799609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Groszer M, Erickson R, Scripture-Adams DD, Lesche R, Trumpp A, Zack JA, Kornblum HI, Liu X, Wu H. Negative regulation of neural stem/progenitor cell proliferation by the Pten tumor suppressor gene in vivo. Science. 2001;294(5549):2186–2189. doi: 10.1126/science.1065518. [DOI] [PubMed] [Google Scholar]
  • 90.Vescovi AL, Reynolds BA, Fraser DD, Weiss S. bFGF regulates the proliferative fate of unipotent (neuronal) and bipotent (neuronal/astroglial) EGF-generated CNS progenitor cells. Neuron. 1993;11(5):951–966. doi: 10.1016/0896-6273(93)90124-A. [DOI] [PubMed] [Google Scholar]
  • 91.Pastrana E, Silva-Vargas V, Doetsch F. Eyes wide open: a critical review of sphere-formation as an assay for stem cells. Cell Stem Cell. 2011;8(5):486–498. doi: 10.1016/j.stem.2011.04.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.McKelvie PA, Rosen KM, Kinney HC, Villa-Komaroff L. Insulin-like growth factor II expression in the developing human brain. J Neuropathol Exp Neurol. 1992;51(4):464–471. doi: 10.1097/00005072-199207000-00009. [DOI] [PubMed] [Google Scholar]
  • 93.Stylianopoulou F, Herbert J, Soares MB, Efstratiadis A. Expression of the insulin-like growth factor II gene in the choroid plexus and the leptomeninges of the adult rat central nervous system. Proc Natl Acad Sci U S A. 1988;85(1):141–145. doi: 10.1073/pnas.85.1.141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Dugas JC, Mandemakers W, Rogers M, Ibrahim A, Daneman R, Barres BA. A novel purification method for CNS projection neurons leads to the identification of brain vascular cells as a source of trophic support for corticospinal motor neurons. J Neurosci. 2008;28(33):8294–8305. doi: 10.1523/JNEUROSCI.2010-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Chell JM, Brand AH. Forever young: death-defying neuroblasts. Cell. 2008;133(5):769–771. doi: 10.1016/j.cell.2008.05.010. [DOI] [PubMed] [Google Scholar]
  • 96.Sousa-Nunes R, Yee LL, Gould AP. Fat cells reactivate quiescent neuroblasts via TOR and glial insulin relays in Drosophila. Nature. 2011;471(7339):508–512. doi: 10.1038/nature09867. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Dahmane N, Ruiz i Altaba A. Sonic hedgehog regulates the growth and patterning of the cerebellum. Development. 1999;126(14):3089–3100. doi: 10.1242/dev.126.14.3089. [DOI] [PubMed] [Google Scholar]
  • 98.Wallace VA. Purkinje-cell-derived Sonic hedgehog regulates granule neuron precursor cell proliferation in the developing mouse cerebellum. Curr Biol. 1999;9(8):445–448. doi: 10.1016/S0960-9822(99)80195-X. [DOI] [PubMed] [Google Scholar]
  • 99.Wechsler-Reya RJ, Scott MP. Control of neuronal precursor proliferation in the cerebellum by Sonic Hedgehog. Neuron. 1999;22(1):103–114. doi: 10.1016/S0896-6273(00)80682-0. [DOI] [PubMed] [Google Scholar]
  • 100.Chambon P. A decade of molecular biology of retinoic acid receptors. FASEB J. 1996;10(9):940–954. [PubMed] [Google Scholar]
  • 101.Haskell GT, LaMantia AS. Retinoic acid signaling identifies a distinct precursor population in the developing and adult forebrain. J Neurosci. 2005;25(33):7636–7647. doi: 10.1523/JNEUROSCI.0485-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Ribes V, Wang Z, Dolle P, Niederreither K. Retinaldehyde dehydrogenase 2 (RALDH2)-mediated retinoic acid synthesis regulates early mouse embryonic forebrain development by controlling FGF and sonic hedgehog signaling. Development. 2006;133(2):351–361. doi: 10.1242/dev.02204. [DOI] [PubMed] [Google Scholar]
  • 103.Siegenthaler JA, Ashique AM, Zarbalis K, Patterson KP, Hecht JH, Kane MA, Folias AE, Choe Y, May SR, Kume T, Napoli JL, Peterson AS, Pleasure SJ. Retinoic acid from the meninges regulates cortical neuron generation. Cell. 2009;139(3):597–609. doi: 10.1016/j.cell.2009.10.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Radakovits R, Barros CS, Belvindrah R, Patton B, Muller U. Regulation of radial glial survival by signals from the meninges. J Neurosci. 2009;29(24):7694–7705. doi: 10.1523/JNEUROSCI.5537-08.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Konno D, Shioi G, Shitamukai A, Mori A, Kiyonari H, Miyata T, Matsuzaki F. Neuroepithelial progenitors undergo LGN-dependent planar divisions to maintain self-renewability during mammalian neurogenesis. Nat Cell Biol. 2008;10(1):93–101. doi: 10.1038/ncb1673. [DOI] [PubMed] [Google Scholar]
  • 106.Freese JL, Pino D, Pleasure SJ. Wnt signaling in development and disease. Neurobiol Dis. 2010;38(2):148–153. doi: 10.1016/j.nbd.2009.09.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Wang J, Wynshaw-Boris A. The canonical Wnt pathway in early mammalian embryogenesis and stem cell maintenance/differentiation. Curr Opin Genet Dev. 2004;14(5):533–539. doi: 10.1016/j.gde.2004.07.013. [DOI] [PubMed] [Google Scholar]
  • 108.Zhou CJ, Borello U, Rubenstein JL, Pleasure SJ. Neuronal production and precursor proliferation defects in the neocortex of mice with loss of function in the canonical Wnt signaling pathway. Neuroscience. 2006;142(4):1119–1131. doi: 10.1016/j.neuroscience.2006.07.007. [DOI] [PubMed] [Google Scholar]
  • 109.Shimogori T, Banuchi V, Ng HY, Strauss JB, Grove EA. Embryonic signaling centers expressing BMP, WNT and FGF proteins interact to pattern the cerebral cortex. Development. 2004;131(22):5639–5647. doi: 10.1242/dev.01428. [DOI] [PubMed] [Google Scholar]
  • 110.Levine AJ, Brivanlou AH. GDF3, a BMP inhibitor, regulates cell fate in stem cells and early embryos. Development. 2006;133(2):209–216. doi: 10.1242/dev.02192. [DOI] [PubMed] [Google Scholar]
  • 111.Johanson C, Stopa E, Baird A, Sharma H. Traumatic brain injury and recovery mechanisms: peptide modulation of periventricular neurogenic regions by the choroid plexus-CSF nexus. J Neural Transm. 2011;118(1):115–133. doi: 10.1007/s00702-010-0498-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Kazanis I, Giannakopoulou M, Philippidis H, Stylianopoulou F. Alterations in IGF-I, BDNF and NT-3 levels following experimental brain trauma and the effect of IGF-I administration. Exp Neurol. 2004;186(2):221–234. doi: 10.1016/j.expneurol.2003.12.004. [DOI] [PubMed] [Google Scholar]
  • 113.Madathil SK, Evans HN, Saatman KE. Temporal and regional changes in IGF-1/IGF-1R signaling in the mouse brain after traumatic brain injury. J Neurotrauma. 2010;27(1):95–107. doi: 10.1089/neu.2009.1002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Sandberg Nordqvist AC, von Holst H, Holmin S, Sara VR, Bellander BM, Schalling M. Increase of insulin-like growth factor (IGF)-1, IGF binding protein-2 and -4 mRNAs following cerebral contusion. Brain Res Mol Brain Res. 1996;38(2):285–293. doi: 10.1016/0169-328X(95)00346-T. [DOI] [PubMed] [Google Scholar]
  • 115.Walter HJ, Berry M, Hill DJ, Cwyfan-Hughes S, Holly JM, Logan A. Distinct sites of insulin-like growth factor (IGF)-II expression and localization in lesioned rat brain: possible roles of IGF binding proteins (IGFBPs) in the mediation of IGF-II activity. Endocrinology. 1999;140(1):520–532. doi: 10.1210/en.140.1.520. [DOI] [PubMed] [Google Scholar]
  • 116.Endoh M, Pulsinelli WA, Wagner JA. Transient global ischemia induces dynamic changes in the expression of bFGF and the FGF receptor. Brain Res Mol Brain Res. 1994;22(1–4):76–88. doi: 10.1016/0169-328X(94)90034-5. [DOI] [PubMed] [Google Scholar]
  • 117.Lin TN, Te J, Lee M, Sun GY, Hsu CY. Induction of basic fibroblast growth factor (bFGF) expression following focal cerebral ischemia. Brain Res Mol Brain Res. 1997;49(1–2):255–265. doi: 10.1016/S0169-328X(97)00152-6. [DOI] [PubMed] [Google Scholar]
  • 118.Chiaretti A, Antonelli A, Riccardi R, Genovese O, Pezzotti P, Di Rocco C, Tortorolo L, Piedimonte G. Nerve growth factor expression correlates with severity and outcome of traumatic brain injury in children. Eur J Paediatr Neurol. 2008;12(3):195–204. doi: 10.1016/j.ejpn.2007.07.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Patterson SL, Grady MS, Bothwell M. Nerve growth factor and a fibroblast growth factor-like neurotrophic activity in cerebrospinal fluid of brain injured human patients. Brain Res. 1993;605(1):43–49. doi: 10.1016/0006-8993(93)91354-U. [DOI] [PubMed] [Google Scholar]
  • 120.Morganti-Kossmann MC, Hans VH, Lenzlinger PM, Dubs R, Ludwig E, Trentz O, Kossmann T. TGF-beta is elevated in the CSF of patients with severe traumatic brain injuries and parallels blood-brain barrier function. J Neurotrauma. 1999;16(7):617–628. doi: 10.1089/neu.1999.16.617. [DOI] [PubMed] [Google Scholar]
  • 121.Pasinetti GM, Nichols NR, Tocco G, Morgan T, Laping N, Finch CE. Transforming growth factor beta 1 and fibronectin messenger RNA in rat brain: responses to injury and cell-type localization. Neuroscience. 1993;54(4):893–907. doi: 10.1016/0306-4522(93)90583-2. [DOI] [PubMed] [Google Scholar]
  • 122.Doyle KP, Cekanaviciute E, Mamer LE, Buckwalter MS. TGFbeta signaling in the brain increases with aging and signals to astrocytes and innate immune cells in the weeks after stroke. J neuroinflammation. 2010;7:62. doi: 10.1186/1742-2094-7-62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Cheng Q, Di Liberto V, Caniglia G, Mudo G. Time-course of GDNF and its receptor expression after brain injury in the rat. Neurosci Lett. 2008;439(1):24–29. doi: 10.1016/j.neulet.2008.04.089. [DOI] [PubMed] [Google Scholar]
  • 124.Yang K, Perez-Polo JR, Mu XS, Yan HQ, Xue JJ, Iwamoto Y, Liu SJ, Dixon CE, Hayes RL. Increased expression of brain-derived neurotrophic factor but not neurotrophin-3 mRNA in rat brain after cortical impact injury. J Neurosci Res. 1996;44(2):157–164. doi: 10.1002/(SICI)1097-4547(19960415)44:2<157::AID-JNR8>3.0.CO;2-C. [DOI] [PubMed] [Google Scholar]
  • 125.Korhonen L, Riikonen R, Nawa H, Lindholm D. Brain derived neurotrophic factor is increased in cerebrospinal fluid of children suffering from asphyxia. Neurosci Lett. 1998;240(3):151–154. doi: 10.1016/S0304-3940(97)00937-3. [DOI] [PubMed] [Google Scholar]
  • 126.Mocchetti I, Wrathall JR. Neurotrophic factors in central nervous system trauma. J Neurotrauma. 1995;12(5):853–870. doi: 10.1089/neu.1995.12.853. [DOI] [PubMed] [Google Scholar]
  • 127.Pasarica D, Gheorghiu M, Toparceanu F, Bleotu C, Ichim L, Trandafir T. Neurotrophin-3, TNF-alpha and IL-6 relations in serum and cerebrospinal fluid of ischemic stroke patients. Roum Arch Microbiol Immunol. 2005;64(1–4):27–33. [PubMed] [Google Scholar]
  • 128.Royo NC, Conte V, Saatman KE, Shimizu S, Belfield CM, Soltesz KM, Davis JE, Fujimoto ST, McIntosh TK. Hippocampal vulnerability following traumatic brain injury: a potential role for neurotrophin-4/5 in pyramidal cell neuroprotection. Eur J Neurosci. 2006;23(5):1089–1102. doi: 10.1111/j.1460-9568.2006.04642.x. [DOI] [PubMed] [Google Scholar]
  • 129.Shore PM, Jackson EK, Wisniewski SR, Clark RS, Adelson PD, Kochanek PM. Vascular endothelial growth factor is increased in cerebrospinal fluid after traumatic brain injury in infants and children. Neurosurgery. 2004;54(3):605–611. doi: 10.1227/01.neu.0000108642.88724.db. [DOI] [PubMed] [Google Scholar]
  • 130.Brines ML, Ghezzi P, Keenan S, Agnello D, de Lanerolle NC, Cerami C, Itri LM, Cerami A. Erythropoietin crosses the blood-brain barrier to protect against experimental brain injury. Proc Nat Acad Sci USA. 2000;97(19):10526–10531. doi: 10.1073/pnas.97.19.10526. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Juul SE, Stallings SA, Christensen RD. Erythropoietin in the cerebrospinal fluid of neonates who sustained CNS injury. Pediatr Res. 1999;46(5):543–547. doi: 10.1203/00006450-199911000-00009. [DOI] [PubMed] [Google Scholar]
  • 132.Sakanaka M, Wen TC, Matsuda S, Masuda S, Morishita E, Nagao M, Sasaki R. In vivo evidence that erythropoietin protects neurons from ischemic damage. Proc Nat Acad Sci USA. 1998;95(8):4635–4640. doi: 10.1073/pnas.95.8.4635. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Ross SA, Halliday MI, Campbell GC, Byrnes DP, Rowlands BJ. The presence of tumour necrosis factor in CSF and plasma after severe head injury. Br J Neurosurg. 1994;8(4):419–425. doi: 10.3109/02688699408995109. [DOI] [PubMed] [Google Scholar]
  • 134.Sriram K, O’Callaghan JP. Divergent roles for tumor necrosis factor-alpha in the brain. J neuroimmune pharmacol: Off J Soc Neuroimmune Pharmacol. 2007;2(2):140–153. doi: 10.1007/s11481-007-9070-6. [DOI] [PubMed] [Google Scholar]
  • 135.Stover JF, Schoning B, Beyer TF, Woiciechowsky C, Unterberg AW. Temporal profile of cerebrospinal fluid glutamate, interleukin-6, and tumor necrosis factor-alpha in relation to brain edema and contusion following controlled cortical impact injury in rats. Neurosci Lett. 2000;288(1):25–28. doi: 10.1016/S0304-3940(00)01187-3. [DOI] [PubMed] [Google Scholar]
  • 136.Chasseigneaux S, Allinquant B. Functions of Abeta, sAPPalpha and sAPPbeta: similarities and differences. J Neurochem. 2012;120(Suppl 1):99–108. doi: 10.1111/j.1471-4159.2011.07584.x. [DOI] [PubMed] [Google Scholar]
  • 137.Olsson A, Csajbok L, Ost M, Hoglund K, Nylen K, Rosengren L, Nellgard B, Blennow K. Marked increase of beta-amyloid (1–42) and amyloid precursor protein in ventricular cerebrospinal fluid after severe traumatic brain injury. J Neurol. 2004;251(7):870–876. doi: 10.1007/s00415-004-0451-y. [DOI] [PubMed] [Google Scholar]
  • 138.Chiaretti A, Antonelli A, Genovese O, Pezzotti P, Rocco CD, Viola L, Riccardi R. Nerve growth factor and doublecortin expression correlates with improved outcome in children with severe traumatic brain injury. J Trauma. 2008;65(1):80–85. doi: 10.1097/TA.0b013e31805f7036. [DOI] [PubMed] [Google Scholar]
  • 139.Chiaretti A, Barone G, Riccardi R, Antonelli A, Pezzotti P, Genovese O, Tortorolo L, Conti G. NGF, DCX, and NSE upregulation correlates with severity and outcome of head trauma in children. Neurology. 2009;72(7):609–616. doi: 10.1212/01.wnl.0000342462.51073.06. [DOI] [PubMed] [Google Scholar]
  • 140.Rotwein P, Burgess SK, Milbrandt JD, Krause JE. Differential expression of insulin-like growth factor genes in rat central nervous system. Proc Natl Acad Sci U S A. 1988;85(1):265–269. doi: 10.1073/pnas.85.1.265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Valentino KL, Ocrant I, Rosenfeld RG. Developmental expression of insulin-like growth factor-II receptor immunoreactivity in the rat central nervous system. Endocrinology. 1990;126(2):914–920. doi: 10.1210/endo-126-2-914. [DOI] [PubMed] [Google Scholar]
  • 142.Wood TL, Brown AL, Rechler MM, Pintar JE. The expression pattern of an insulin-like growth factor (IGF)-binding protein gene is distinct from IGF-II in the midgestational rat embryo. Mol Endocrinol. 1990;4(8):1257–1263. doi: 10.1210/mend-4-8-1257. [DOI] [PubMed] [Google Scholar]
  • 143.Kelley KM, Oh Y, Gargosky SE, Gucev Z, Matsumoto T, Hwa V, Ng L, Simpson DM, Rosenfeld RG. Insulin-like growth factor-binding proteins (IGFBPs) and their regulatory dynamics. Int J Biochem Cell Biol. 1996;28(6):619–637. doi: 10.1016/1357-2725(96)00005-2. [DOI] [PubMed] [Google Scholar]
  • 144.Chesik D, De Keyser J, Wilczak N. Insulin-like growth factor binding protein-2 as a regulator of IGF actions in CNS: implications in multiple sclerosis. Cytokine Growth Factor Rev. 2007;18(3–4):267–278. doi: 10.1016/j.cytogfr.2007.04.001. [DOI] [PubMed] [Google Scholar]
  • 145.Gonzalez AM, Podvin S, Lin SY, Miller MC, Botfield H, Leadbeater WE, Roberton A, Dang X, Knowling SE, Cardenas-Galindo E, Donahue JE, Stopa EG, Johanson CE, Coimbra R, Eliceiri BP, Baird A. Ecrg4 expression and its product augurin in the choroid plexus: impact on fetal brain development, cerebrospinal fluid homeostasis and neuroprogenitor cell response to CNS injury. Fluids Barriers CNS. 2011;8(1):6. doi: 10.1186/2045-8118-8-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Podvin S, Gonzalez AM, Miller MC, Dang X, Botfield H, Donahue JE, Kurabi A, Boissaud-Cooke M, Rossi R, Leadbeater WE, Johanson CE, Coimbra R, Stopa EG, Eliceiri BP, Baird A. Esophageal cancer related gene-4 is a choroid plexus-derived injury response gene: evidence for a biphasic response in early and late brain injury. PLoS ONE. 2011;6(9):e24609. doi: 10.1371/journal.pone.0024609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Li LW, Yu XY, Yang Y, Zhang CP, Guo LP, Lu SH. Expression of esophageal cancer related gene 4 (ECRG4), a novel tumor suppressor gene, in esophageal cancer and its inhibitory effect on the tumor growth in vitro and in vivo. Int J Cancer. 2009;125(7):1505–1513. doi: 10.1002/ijc.24513. [DOI] [PubMed] [Google Scholar]
  • 148.Sabatier R, Finetti P, Adelaide J, Guille A, Borg JP, Chaffanet M, Lane L, Birnbaum D, Bertucci F. Down-regulation of ECRG4, a candidate tumor suppressor gene, in human breast cancer. PLoS ONE. 2011;6(11):e27656. doi: 10.1371/journal.pone.0027656. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Kujuro Y, Suzuki N, Kondo T. Esophageal cancer-related gene 4 is a secreted inducer of cell senescence expressed by aged CNS precursor cells. Proc Natl Acad Sci U S A. 2010;107(18):8259–8264. doi: 10.1073/pnas.0911446107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Sun D, Bullock MR, McGinn MJ, Zhou Z, Altememi N, Hagood S, Hamm R, Colello RJ. Basic fibroblast growth factor-enhanced neurogenesis contributes to cognitive recovery in rats following traumatic brain injury. Exp Neurol. 2009;216(1):56–65. doi: 10.1016/j.expneurol.2008.11.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Wada K, Sugimori H, Bhide PG, Moskowitz MA, Finklestein SP. Effect of basic fibroblast growth factor treatment on brain progenitor cells after permanent focal ischemia in rats. Stroke. 2003;34(11):2722–2728. doi: 10.1161/01.STR.0000094421.61917.71. [DOI] [PubMed] [Google Scholar]
  • 152.Watanabe T, Okuda Y, Nonoguchi N, Zhao MZ, Kajimoto Y, Furutama D, Yukawa H, Shibata MA, Otsuki Y, Kuroiwa T, Miyatake S. Postischemic intraventricular administration of FGF-2 expressing adenoviral vectors improves neurologic outcome and reduces infarct volume after transient focal cerebral ischemia in rats. J Cereb Blood Flow Metab. 2004;24(11):1205–1213. doi: 10.1097/01.WCB.0000136525.75839.41. [DOI] [PubMed] [Google Scholar]
  • 153.Sun D, Bullock MR, Altememi N, Zhou Z, Hagood S, Rolfe A, McGinn MJ, Hamm R, Colello RJ. The effect of epidermal growth factor in the injured brain after trauma in rats. J Neurotrauma. 2010;27(5):923–938. doi: 10.1089/neu.2009.1209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Dixon CE, Flinn P, Bao J, Venya R, Hayes RL. Nerve growth factor attenuates cholinergic deficits following traumatic brain injury in rats. Exp Neurol. 1997;146(2):479–490. doi: 10.1006/exnr.1997.6557. [DOI] [PubMed] [Google Scholar]
  • 155.Araujo DM, Hilt DC. Glial cell line-derived neurotrophic factor attenuates the excitotoxin-induced behavioral and neurochemical deficits in a rodent model of Huntington’s disease. Neuroscience. 1997;81(4):1099–1110. doi: 10.1016/S0306-4522(97)00079-1. [DOI] [PubMed] [Google Scholar]
  • 156.Lee C, Agoston DV. Vascular endothelial growth factor is involved in mediating increased de novo hippocampal neurogenesis in response to traumatic brain injury. J Neurotrauma. 2010;27(3):541–553. doi: 10.1089/neu.2009.0905. [DOI] [PubMed] [Google Scholar]
  • 157.Schabitz WR, Schwab S, Spranger M, Hacke W. Intraventricular brain-derived neurotrophic factor reduces infarct size after focal cerebral ischemia in rats. J Cereb Blood Flow Metab. 1997;17(5):500–506. doi: 10.1097/00004647-199705000-00003. [DOI] [PubMed] [Google Scholar]
  • 158.Sharma HS, Johanson CE. Intracerebroventricularly administered neurotrophins attenuate blood cerebrospinal fluid barrier breakdown and brain pathology following whole-body hyperthermia: an experimental study in the rat using biochemical and morphological approaches. Ann N Y Acad Sci. 2007;1122:112–129. doi: 10.1196/annals.1403.008. [DOI] [PubMed] [Google Scholar]
  • 159.Storkebaum E, Lambrechts D, Dewerchin M, Moreno-Murciano MP, Appelmans S, Oh H, Van Damme P, Rutten B, Man WY, De Mol M, Wyns S, Manka D, Vermeulen K, Van Den Bosch L, Mertens N, Schmitz C, Robberecht W, Conway EM, Collen D, Moons L, Carmeliet P. Treatment of motoneuron degeneration by intracerebroventricular delivery of VEGF in a rat model of ALS. Nat Neurosci. 2005;8(1):85–92. doi: 10.1038/nn1360. [DOI] [PubMed] [Google Scholar]
  • 160.Takeshima Y, Nakamura M, Miyake H, Tamaki R, Inui T, Horiuchi K, Wajima D, Nakase H. Neuroprotection with intraventricular brain-derived neurotrophic factor in rat venous occlusion model. Neurosurgery. 2011;68(5):1334–1341. doi: 10.1227/NEU.0b013e31820c048e. [DOI] [PubMed] [Google Scholar]
  • 161.Thau-Zuchman O, Shohami E, Alexandrovich AG, Leker RR. Vascular endothelial growth factor increases neurogenesis after traumatic brain injury. J Cereb Blood Flow Metab. 2010;30(5):1008–1016. doi: 10.1038/jcbfm.2009.271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Kolb B, Morshead C, Gonzalez C, Kim M, Gregg C, Shingo T, Weiss S. Growth factor-stimulated generation of new cortical tissue and functional recovery after stroke damage to the motor cortex of rats. J Cereb Blood Flow Metab. 2007;27(5):983–997. doi: 10.1038/sj.jcbfm.9600402. [DOI] [PubMed] [Google Scholar]
  • 163.Sun L, Lee J, Fine HA. Neuronally expressed stem cell factor induces neural stem cell migration to areas of brain injury. J Clin Invest. 2004;113(9):1364–1374. doi: 10.1172/JCI20001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Alcala-Barraza SR, Lee MS, Hanson LR, McDonald AA, Frey WH, 2nd, McLoon LK. Intranasal delivery of neurotrophic factors BDNF, CNTF, EPO, and NT-4 to the CNS. J Drug Target. 2010;18(3):179–190. doi: 10.3109/10611860903318134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Cheng X, Wang Z, Yang J, Ma M, Lu T, Xu G, Liu X. Acidic fibroblast growth factor delivered intranasally induces neurogenesis and angiogenesis in rats after ischemic stroke. Neurol Res. 2011;33(7):675–680. doi: 10.1179/1743132810Y.0000000004. [DOI] [PubMed] [Google Scholar]
  • 166.Craft S, Baker LD, Montine TJ, Minoshima S, Watson GS, Claxton A, Arbuckle M, Callaghan M, Tsai E, Plymate SR, Green PS, Leverenz J, Cross D, Gerton B. Intranasal insulin therapy for Alzheimer disease and amnestic mild cognitive impairment: a pilot clinical trial. Arch Neurol. 2012;69(1):29–38. doi: 10.1001/archneurol.2011.233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Lochhead JJ, Thorne RG (2011) Intranasal delivery of biologics to the central nervous system. Adv Drug Deliv Rev (in press) [DOI] [PubMed]
  • 168.Ma M, Ma Y, Yi X, Guo R, Zhu W, Fan X, Xu G, Frey WH, 2nd, Liu X. Intranasal delivery of transforming growth factor-beta1 in mice after stroke reduces infarct volume and increases neurogenesis in the subventricular zone. BMC Neurosci. 2008;9:117. doi: 10.1186/1471-2202-9-117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Reger MA, Watson GS, Green PS, Baker LD, Cholerton B, Fishel MA, Plymate SR, Cherrier MM, Schellenberg GD, Frey WH, 2nd, Craft S. Intranasal insulin administration dose-dependently modulates verbal memory and plasma amyloid-beta in memory-impaired older adults. J Alzheimers Dis. 2008;13(3):323–331. doi: 10.3233/jad-2008-13309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Reger MA, Watson GS, Green PS, Wilkinson CW, Baker LD, Cholerton B, Fishel MA, Plymate SR, Breitner JC, DeGroodt W, Mehta P, Craft S. Intranasal insulin improves cognition and modulates beta-amyloid in early AD. Neurology. 2008;70(6):440–448. doi: 10.1212/01.WNL.0000265401.62434.36. [DOI] [PubMed] [Google Scholar]
  • 171.Thorne RG, Pronk GJ, Padmanabhan V, Frey WH., 2nd Delivery of insulin-like growth factor-I to the rat brain and spinal cord along olfactory and trigeminal pathways following intranasal administration. Neuroscience. 2004;127(2):481–496. doi: 10.1016/j.neuroscience.2004.05.029. [DOI] [PubMed] [Google Scholar]
  • 172.Hanson LR, Frey WH., 2nd Intranasal delivery bypasses the blood-brain barrier to target therapeutic agents to the central nervous system and treat neurodegenerative disease. BMC Neurosci. 2008;9(Suppl 3):S5. doi: 10.1186/1471-2202-9-S3-S5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Liu XF, Fawcett JR, Thorne RG, DeFor TA, Frey WH., 2nd Intranasal administration of insulin-like growth factor-I bypasses the blood-brain barrier and protects against focal cerebral ischemic damage. J Neurol Sci. 2001;187(1–2):91–97. doi: 10.1016/S0022-510X(01)00532-9. [DOI] [PubMed] [Google Scholar]
  • 174.Liu XF, Fawcett JR, Thorne RG, Frey WH., 2nd Non-invasive intranasal insulin-like growth factor-I reduces infarct volume and improves neurologic function in rats following middle cerebral artery occlusion. Neurosci Lett. 2001;308(2):91–94. doi: 10.1016/S0304-3940(01)01982-6. [DOI] [PubMed] [Google Scholar]
  • 175.Born J, Lange T, Kern W, McGregor GP, Bickel U, Fehm HL. Sniffing neuropeptides: a transnasal approach to the human brain. Nat Neurosci. 2002;5(6):514–516. doi: 10.1038/nn0602-849. [DOI] [PubMed] [Google Scholar]
  • 176.Parada C, Gato A, Aparicio M, Bueno D. Proteome analysis of chick embryonic cerebrospinal fluid. Proteomics. 2006;6(1):312–320. doi: 10.1002/pmic.200500085. [DOI] [PubMed] [Google Scholar]
  • 177.Hussein MH, Daoud GA, Kakita H, Kato S, Goto T, Kamei M, Goto K, Nobata M, Ozaki Y, Ito T, Fukuda S, Kato I, Suzuki S, Sobajima H, Hara F, Hashimoto T, Togari H. High cerebrospinal fluid antioxidants and interleukin 8 are protective of hypoxic brain damage in newborns. Free Radic Res. 2010;44(4):422–429. doi: 10.3109/10715760903548245. [DOI] [PubMed] [Google Scholar]
  • 178.Borlongan CV, Skinner SJ, Geaney M, Vasconcellos AV, Elliott RB, Emerich DF. CNS grafts of rat choroid plexus protect against cerebral ischemia in adult rats. NeuroReport. 2004;15(10):1543–1547. doi: 10.1097/01.wnr.0000133298.84901.cf. [DOI] [PubMed] [Google Scholar]
  • 179.Borlongan CV, Skinner SJ, Geaney M, Vasconcellos AV, Elliott RB, Emerich DF. Intracerebral transplantation of porcine choroid plexus provides structural and functional neuroprotection in a rodent model of stroke. Stroke. 2004;35(9):2206–2210. doi: 10.1161/01.STR.0000138954.25825.0b. [DOI] [PubMed] [Google Scholar]
  • 180.Regev L, Ezrielev E, Gershon E, Gil S, Chen A. Genetic approach for intracerebroventricular delivery. Proc Natl Acad Sci U S A. 2010;107(9):4424–4429. doi: 10.1073/pnas.0907059107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Ohta M, Suzuki Y, Noda T, Kataoka K, Chou H, Ishikawa N, Kitada M, Matsumoto N, Dezawa M, Suzuki S, Ide C. Implantation of neural stem cells via cerebrospinal fluid into the injured root. NeuroReport. 2004;15(8):1249–1253. doi: 10.1097/01.wnr.0000129998.72184.e1. [DOI] [PubMed] [Google Scholar]
  • 182.Wu S, Suzuki Y, Kitada M, Kataoka K, Kitaura M, Chou H, Nishimura Y, Ide C. New method for transplantation of neurosphere cells into injured spinal cord through cerebrospinal fluid in rat. Neurosci Lett. 2002;318(2):81–84. doi: 10.1016/S0304-3940(01)02488-0. [DOI] [PubMed] [Google Scholar]
  • 183.Wu S, Suzuki Y, Kitada M, Kitaura M, Kataoka K, Takahashi J, Ide C, Nishimura Y. Migration, integration, and differentiation of hippocampus-derived neurosphere cells after transplantation into injured rat spinal cord. Neurosci Lett. 2001;312(3):173–176. doi: 10.1016/S0304-3940(01)02219-4. [DOI] [PubMed] [Google Scholar]
  • 184.Park D, Joo SS, Kim TK, Lee SH, Kang H, Lee HJ, Lim I, Matsuo A, Tooyama I, Kim YB, Kim SU (2011) Human neural stem cells overexpressing choline acetyltransferase restore cognitive function of kainic acid-induced learning and memory deficit animals. Cell Transplant (in press) [DOI] [PubMed]

Articles from Cellular and Molecular Life Sciences: CMLS are provided here courtesy of Springer

RESOURCES