Traumatic brain injury (TBI) is a serious public health problem. It may result in severe neurological disabilities and in a variety of cellular metabolic alterations for which available therapeutic strategies are limited. In the last decade, the use of estrogenic compounds, which activate protective mechanisms in astrocytes, has been explored as a potential experimental therapeutic approach. Previous works have suggested estradiol (E2) as a neuroprotective hormone that acts in the brain by binding to estrogen receptors (ERs). Several steroidal and nonsteroidal estrogenic compounds can imitate the effects of estradiol on ERs. These include hormonal estrogens, phytoestrogens and synthetic estrogens, such as selective ER modulators or tibolone. Current evidence of the role of astrocytes in mediating protective actions of estrogenic compounds after TBI is reviewed in this paper. We conclude that the use of estrogenic compounds to modulate astrocytic properties is a promising therapeutic approach for the treatment of TBI.

Traumatic brain injury (TBI) is generated by a traumatic event, which causes a strong impact on the brain and results in acute or chronic impairments in the function of neural cells [1]. TBI is a severe public health problem, particularly among male adolescents, young adults, and older people of both sexes [2]. Indeed, TBI is a familiar source of morbidity and mortality all over the world, whereby 69 million people are estimated to be affected by TBI each year [3]. Besides, it is the main cause of mortality and impairment among young people in the low- and middle-income countries [4]. Currently, brain injury has a higher frequency than other highly spread diseases such as cancer, acquired immune deficiency syndrome, Parkinson’s disease and multiple sclerosis [1]. TBI has heterogeneous aetiology, type, severity and outcomes. There are 2 main types of TBI: penetrating and non-penetrating. Penetrating TBI is an open lesion in the head causing intracranial hemorrhage. Non-penetrating TBI causes brain damage because of indirect impact without penetrance of any objects into the brain [5]. As a result of TBI, one or more brain regions are damaged and become swollen [3, 6].

Despite the availability of standardized treatment guidelines and improvements in understanding the mechanisms of cellular damage, therapeutic strategies for patients with TBI remain unsatisfactory because of their reduced efficacy in preventing the metabolic changes and brain dysfunctions caused by the injury. Brain responses to damage and disease involve several cell types that interact with each other in an attempt to preserve cell viability and homeostatic physiological function [7]. In the last few years, protective strategies have been focused on modulating the responses of different brain cells to injury. In this regard, astrocytes may represent relevant candidates, since they play critical roles in the central nervous system (CNS) both under healthy and pathological conditions. In the healthy CNS, astrocytes accomplish a significant number of homeostatic functions, including the control of neuronal metabolism, production of antioxidants and regulation of synaptic transmission and plasticity. In addition, astrocytes respond to brain injury with a series of morphological, metabolic and cellular changes involved in the protection of the damaged neural tissue [8]. Studies with intravital time-lapse imaging have shown that ATP and other metabolites released by astrocytes play a pivotal role in stimulating beneficial cellular responses after brain injury [9]. Therefore, the identification of molecules/compounds that activate protective mechanisms in astrocytes may lead to the development of potential therapeutic strategies for TBI [10-12].

Estrogenic compounds include a wide variety of natural and synthetic steroid and non-steroid molecules that bind to ERs, thereby mimicking the activity of estradiol in different tissues. Estrogenic compounds can affect astrocytes, either directly or indirectly, and exert numerous actions in the CNS under physiological and pathological conditions [13]. For instance, it has been shown that estrogenic compounds regulate oedema, extracellular glutamate levels, reactive astrogliosis and inflammatory response after brain trauma [14, 15]. Therefore, they may be useful molecules for the management of TBI [11]. In the next sections, we review the participation of astrocytes in the response of neural tissue to injury and their role in mediating neuroprotective actions of estrogenic compounds to improve neuronal survival and functional outcomes after TBI.

Depending on the outcome of damage to brain parenchyma, the tissue injury produced by TBI is classified into 2 substantially different stages associated with different mechanisms of neural damage: primary and secondary [16]. The primary injury occurs as a consequence of the initial physical insult [17]. This stage often requires surgical attention and happens at the moment of trauma, when tissues and blood vessels are extended, compacted, and damaged [6]. The secondary stage appears hours to days later as a consequence of the activation of signalling pathways that provoke oedema, inflammation and brain cell death [16]. The secondary injury is characterized by high levels of lactate [18], oxygen-free radicals [1], interleukins [19] and intracellular free Ca2+ in response to the primary injury [20]. In addition, impairment of the blood-brain barrier (BBB), excessive release of the neurotransmitter glutamate and dysfunction of mitochondria in the brain [21] contribute to the secondary injury. Other alterations that can be detected in secondary brain injury are decreased brain blood flow, ischemia, increased reactive oxygen species (ROS) and brain cell death [22].

In comparison with primary injuries, secondary ones are progressive, a fact that can be vital for patient recovery [23]. TBI may be associated with different injuries in other body regions (limb fractures, thoracic, or abdominal injuries) [24], which increase the risk of secondary brain injury due to hypotension, hypoxia, pyrexia and coagulopathy. In addition to brain cell death, TBI is associated with neuroinflammation, axonal damage and vascular abnormalities [25]. TBI includes anomalous ranges of oxygenation, upregulation of inflammatory pathways and irregular endocrine secretion [26]. Interestingly, there is evidence suggesting that neuroendocrine dysfunction might contribute negatively to the severity and the TBI outcome [27]. Recent studies using controlled cortical impacts in mice have demonstrated that the alterations in growth hormone levels after brain injury are associated with a disruption in barrier organization of tanycytes [28, 29]. It has been suggested that these cells play a crucial role in the maintenance of BBB, the control of the pituitary function and the secretion of active compounds. All these functions, which are critical to the protection of the homeostasis of the brain, have highlighted the important role of tanycytes during TBI [30].

The early secondary stages of cerebral injury after TBI are characterized by the accumulation of lactic acid due to an increase in anaerobic glycolysis, excess membrane permeability and oedema [22, 31]. The latest pathological stages are characterized by excessive production of excitatory neurotransmitters (glutamate, aspartate), and activation of α-amino-3-hydroxy-5-methyl-4-isoxazolpro-pionate, N-Methyl-D-aspartic acid and voltage-dependent calcium and sodium channels, which consequently generate terminal membrane depolarization and mitochondrial dysfunction [32, 33]. The imbalance of calcium and sodium intake could trigger autophagy mechanisms [34], activation of caspases, translocases and endonucleases, and inhibition of DNA repair, which may induce structural modifications of biological membranes and DNA damage [35, 36]. Consequently, all these events lead to membrane decoupling and finally cell death by necrosis and apoptosis [37]. Furthermore, the excessive production of excitatory neurotransmitters causes excitotoxicity. In this regard, excitotoxicity could lead to excessive stimulation of glutamate receptors, occasioning damage and death of brain cells. Neurons are especially vulnerable to excitotoxicity [38]. Furthermore, astrocytes are less vulnerable and reuptake glutamate from synapses, thanks to their fast conversion from glutamate into glutamine by the glutamine synthetase, thus preventing the excessive and toxic extracellular glutamate accumulation [39].

Excitotoxicity induces harmful effects on the mitochondrial electron transport chain that generates the oxidation of cellular structures and molecules by the newly generated oxidative species [40]. Indeed, another cell death mechanism elicited by TBI is oxidative stress caused by increased levels of ROS and reactive nitrogen species (superoxide, hydrogen peroxide and nitric oxide) [41]. This mechanism is fundamental due to the fact that the brain is highly susceptible to free radical damage because of its oxidative metabolism and its high levels of polyunsaturated lipids [42]. Some studies have demonstrated that superoxide radicals and nitric oxide impair cerebral vascular function after TBI due to their strong oxidative effects [43, 44].

Increased levels of ROS can be triggered not only by an increase in their generation, but also due to a decreased capacity of the enzymatic antioxidant system (superoxide dismutase, glutathione peroxidase and catalase). In this regard, it is relevant to note that astrocytes maintain high expression of intracellular antioxidant enzymes. This antioxidant activity of astrocytes helps to protect the neuron against the oxidative stress preventing the damage of DNA, RNA, and proteins [45, 46]. In this regard, previous works indicated that astrocytes are pivotal mediators in TBI due to their ability to provide neurons (and other cells) metabolic support [47-49].

Function of Astrocytes under Physiological and Pathological Conditions

Astrocytes are the most abundant group of glial cell, representing approximately around 20–40% of the total brain cell population [48, 50, 51]. Their function depends on their shape, position, subtype, developmental stage and metabolic state [48]. Astrocytes are recognized for being multifunctional housekeeping cells involved in a permanent cross-talk with neurons and other neighbouring glial cells [52]. They regulate the concentration of potassium, sodium and glutamate at the synaptic cleft [47-49], provide lactate and glutamine to neurons and participate in neuron signalling modulation [53-57].

The different physiological functions of astrocytes are listed in Table 1. For instance, in a healthy and uninjured CNS, astrocytes play an essential role in the modulation of potassium, sodium, calcium and glutamate levels in the extracellular space, thus modulating synaptic activity and preventing excitotoxic cell death [58-60]. Astrocytes have cell processes that contact with blood vessels, neuronal perikarya, axons, and synapses. Thus, these cells uptake glucose from blood vessels and supply energy metabolites, such as lactate, to neurons [61]. Besides the production gliotransmitters and growth factors, astrocytes also have the capacity of supplying neuroactive steroids, which modulate synaptic activity [61, 62]. It is noteworthy that astrocytes have a dual role: (1) these cells can be a direct target of several endocrine molecules and (2) also they can be regulatory-endocrine cells that exert influences on nervous tissue. For instance, studies demonstrate that in different physiological and pathological conditions astrocytes are able to synthesize a variety of endogenous neurosteroids [63, 64], which have regulatory effects and affect the neurosecretory activity of other cells of the CNS, such as neurons, microglia, and tanycytes [65]. Likewise, the role of astrocytes may be supported by tanycytes and microglia. For instance, tanycytes are cells with morphological and molecular characteristics similar to those of astrocytes and are responsible for transporting substances from the cerebral spinal fluid and the blood portal [30, 66, 67]. Estrogen receptors (ERs) have been identified in tanycytes and microglial cells and it has been observed that they help in estrogenic communication, as estradiol stimulates the secretory activity of these cells by releasing prostaglandin E, which is related to synaptic alterations in the hippocampus, as well as in astrocytes [67, 68]. As discussed later, the interaction of astrocytes with other brain cells plays a fundamental role in neuroprotection [68].

Table 1.

Astrocyte functions under physiological conditions

Astrocyte functions under physiological conditions
Astrocyte functions under physiological conditions

Under pathological conditions, astrocytes undergo a variety of phenotypic and functional changes, known as reactive astrogliosis. Generally, this pathological phenomenon is related to an enhanced proliferation and secretion of inflammatory mediators, adhesion molecules and growth factors [69-74]. Reactive astrogliosis is elicited by a variety of molecular signals (Table 2) associated with different forms of CNS injuries and diseases such as trauma, ischemia, infection, stroke, neurological disorders and neurodegenerative diseases [71]. Compared to non-reactive astrocytes, reactive astrocytes show various alterations in gene expression, activation of specific signalling cascades, such as signal transducer and activator of transcription 3, suppressor of cytokine signalling 3 or nuclear factor κB (NF-κB) and exhibit morphological and functional changes (Fig. 1) [75-78]. These changes depend on the severity of the damage (Table 3) [51, 72, 79-85]. Previous histopathological studies of the human brain have shown various grades of reactive astrogliosis [84]. Mild to moderate astrogliosis are caused by different insults including TBI, and viral and bacterial infections. When the brain tissue presents mild or moderate injury, astrocytes experiment hypertrophic astrogliosis and process hypertrophy [84]. These changes include modifications in the expression of the glial fibrillary acidic protein (GFAP), vimentin, nestin and other proteins [85], which alter the normal cellular structure, energy metabolism, intracellular signalling, and pump activities of astrocytes [84]. In severe cases of brain injury, astrocytes undergo progressive cellular hypertrophy, proliferation and scar formation [7].

Table 2.

Molecules with the ability to activate astrocytes

Molecules with the ability to activate astrocytes
Molecules with the ability to activate astrocytes
Table 3.

Specific signaling cascades involved in the morphological and functional modifications of reactive astrocytes

Specific signaling cascades involved in the morphological and functional modifications of reactive astrocytes
Specific signaling cascades involved in the morphological and functional modifications of reactive astrocytes
Fig. 1.

Schematic representations of different phenotypic changes in reactive astrogliosis. a Healthy astrocytes have many physiological functions aimed to maintain CNS homeostasis. b Under pathological conditions, astrocytes undergo a variety of phenotypic and functional changes, known as reactive astrogliosis. Reactive astrocytes show a variety of changes in the activation of specific signalling cascades, such as STAT3, SOCS3 or NF-κB, cellular morphology and alterations in the expression of intermediate filament proteins such as nestin, glial fibrillary acidic protein (GFAP) and vimentin. c Astrogliosis progress over the time and this can become maladaptive, whereby scar formation takes place. Glial scar includes alterations in molecular expression and functional features.

Fig. 1.

Schematic representations of different phenotypic changes in reactive astrogliosis. a Healthy astrocytes have many physiological functions aimed to maintain CNS homeostasis. b Under pathological conditions, astrocytes undergo a variety of phenotypic and functional changes, known as reactive astrogliosis. Reactive astrocytes show a variety of changes in the activation of specific signalling cascades, such as STAT3, SOCS3 or NF-κB, cellular morphology and alterations in the expression of intermediate filament proteins such as nestin, glial fibrillary acidic protein (GFAP) and vimentin. c Astrogliosis progress over the time and this can become maladaptive, whereby scar formation takes place. Glial scar includes alterations in molecular expression and functional features.

Close modal

Astrogliosis can be interpreted as a homeostatic response aimed to reduce and repair the initial damage, maintaining brain function [51, 74]. However, depending on the different stages of the insult and its progress over time, astrocytes are capable of playing beneficial or detrimental actions [84, 86]. A number of experimental studies have shown that astrogliosis has an essential role in the recovery after a TBI or ischemia, due to their ability to isolate the still health brain tissue [74, 87-89]. Reactive astrocytes have a beneficial role in the acute response after brain trauma because they may limit the extension of damage, and separate the harmful lesion from the rest of the CNS thus preventing its spreading. Studies using genetic ablation of GFAP and vimentin at the time of injury have shown a significant reduction in astrogliosis, a slower response to damage and a decrease in the integration of new neuronal synapses after TBI [82]. Interestingly, this acute ablation increases the number of leukocytes that cross the BBB, leading to neuronal degeneration [87]. On the other hand, it seems like that reactive astrocytes respond to brain injury by expressing aromatase. This enzyme is involved in the synthesis of different estrogens in humans (E1, E2, E3). The increase in the local expression of estrogens may be involved in the neuroprotective and regenerative effect in the brain [90]. It is important to point out that astrogliosis progresses over the time and becomes harmful in chronic stages. In these stages, reactive astrocytes are known to inhibit axonal regeneration and the integration of neurons after brain injury [86, 91-93].

Reactive astrocytes also can release chemokines, growth factors, proteases and danger-associated molecular patterns, which could be beneficial or detrimental for the BBB integrity in various ways [7, 94, 95]. For example, different studies have shown that astrocytes secrete apolipoprotein E and the vasoactive endothelial growth factor in response to stimulation with inflammatory mediators [96], increasing BBB permeability and causing leukocyte infiltration [7, 94, 97, 98].

Astrocytes and the Glial Scar

Scar formation takes place in most lesions that cause severe tissue damage, such as penetrating trauma. In this process, astrocytes undergo severe morphological changes with the spread of long branched processes that overlap [84]. In the pathologic process, reactive astrocytes cooperate with the different brain cells and interact with the extracellular matrix to generate the glial scars [76, 84, 99]. These scars persist for long periods of time even when the causes of the brain damage have disappeared [76, 87, 89, 100]. The glial scar is formed in damaged zones that contain surviving neurons where these glial formations can interact with neuronal and non-neuronal cells in the area around the lesion [87, 101-103]. It has been proposed that in this process of scar formation are involved some molecular triggers that initiate the spread of reactive astrocytes in vivo such as ATP, the signal transducer and activator of transcription 3, fibroblast growth factor, epidermal growth factor, and endothelin 1, which can diminish or aggravate the brain damage [104-106]. However, the origin of newly scar-forming astrocytes is not well established. Different hypotheses have been postulated, such as that mature astrocytes have the capability of starting the cell cycle and multiply in the moment of scar formation [87, 104, 107]. There is also evidence that some of the proliferating astrocytes after brain damage derive from polydendrocytes, cells that express the neural/glial antigen 2 proteoglycan [108], in the brain parenchyma and progenitors of ependymocytes [109, 110]. Besides, it is also postulated that other source of scar-forming astrocytes could be multipotent progenitors in ependymal and subependymal tissues expressing GFAP [84]. These progenitors might produce cells with the capacity of migrating to locations of injury [111].

Recent evidence indicates that the glial scar seems to have a beneficial role by forming a compact barrier that isolates the healthy tissue of damaged tissue and does not allow the red and white blood cells to have contact with the healthy tissue [80]. Glial scar plays also a significant role in the regulation and the control of the propagation of CNS inflammation [84]. By contrast, there are studies showing the negative effects of reactive gliosis and glial scar. For instance, experiments in animal models suggest that the suppression of glial scar formation might improve the outcome after diverse types of CNS injuries by reducing excitotoxic neurodegeneration [87, 112, 113], inducing inflammation [76, 87-89, 114, 115] and improving axonal regeneration [116]. The glial scar can also exert other negative effects. For example, there is also evidence showing that glial scar impedes axonal regeneration and reduces axonal re-growth. As described above, the negative effects of astrogliosis and glial scar have been related to late chronic phases, specifically, when reactive astrocytes are not able to appropriately respond to damage during post-acute and early chronic stages after TBI [116]. Consequently, nerve cells are exposed to harmful molecules for a long time, which also have some deleterious long-term effects in the injured brain. Altogether, accumulating evidence has suggested that astrocytes seem to play a critical determinant role in acute and chronic stages after TBI. Their beneficial and detrimental responses can directly affect neuronal survival [117]. For this reason, astrocytes (and astrogliosis) have been postulated as an important therapeutic target for the action of different neuroprotective substances, such as estrogenic compounds. These compounds have been implicated in the activation of different protective cellular pathways in the CNS.

Numerous studies have shown that synthetic or natural estrogens, such as estradiol, phytoestrogens, selective ER modulators (SERMs) and tibolone, exert neuroprotective actions in vitro and in vivo (Fig. 2) [10, 118-137]. Thus, there is evidence showing that estrogens reduce neuronal death in rodent models of TBI [138] and prevent damage in several pathological conditions affecting the CNS such as cerebral ischemia and Alzheimer’s disease [139]. This opens the possibility of the development of neuroprotective estrogenic drugs to support cognitive function and to decrease damage after brain injury [10, 128, 140, 141].

Fig. 2.

Estrogenic actions on astrocytes. Estrogenic compounds (EC) exert different actions on astrocytes through ERα, ERβ and GRP30. (1) Estrogenic compounds may have beneficial effects after brain injury through the regulation of reactive gliosis; (2) increase in the production and release of different molecules, such as neuroestrogens and neurotrophic factors (TrkA, BDNF and ChAT), with the subsequent decrease of neuronal death after brain injury; (3) regulate glutamate transporter GLT-1 and GLAST expression in astrocytes, thus maintaining optimal glutamate levels; (4) reduce the formation of ROS and decrease the loss of antioxidant glutathione, with the subsequent recovery of the mitochondrial membrane potential, ATP production and decreasing cell death; (5) preserve mitochondrial function in astrocytes through ERα/β. Likely, ERs may inhibit (a) apoptotic signalling and (b) regulate the expression of mitochondrial proteins, proton channels and subunits of key enzymes of the respiratory chain, as well as by eliminating radical oxygen species (ROS); and (6) decrease the inflammatory response in astrocytes reducing neuroinflammation through a decrease in the expression of IL-1β, IL-6, tumor necrosis factor (TNF-α) and TNF-β.

Fig. 2.

Estrogenic actions on astrocytes. Estrogenic compounds (EC) exert different actions on astrocytes through ERα, ERβ and GRP30. (1) Estrogenic compounds may have beneficial effects after brain injury through the regulation of reactive gliosis; (2) increase in the production and release of different molecules, such as neuroestrogens and neurotrophic factors (TrkA, BDNF and ChAT), with the subsequent decrease of neuronal death after brain injury; (3) regulate glutamate transporter GLT-1 and GLAST expression in astrocytes, thus maintaining optimal glutamate levels; (4) reduce the formation of ROS and decrease the loss of antioxidant glutathione, with the subsequent recovery of the mitochondrial membrane potential, ATP production and decreasing cell death; (5) preserve mitochondrial function in astrocytes through ERα/β. Likely, ERs may inhibit (a) apoptotic signalling and (b) regulate the expression of mitochondrial proteins, proton channels and subunits of key enzymes of the respiratory chain, as well as by eliminating radical oxygen species (ROS); and (6) decrease the inflammatory response in astrocytes reducing neuroinflammation through a decrease in the expression of IL-1β, IL-6, tumor necrosis factor (TNF-α) and TNF-β.

Close modal

Endogenous Estrogens

Endogenous estrogens are steroid hormones recognized for their participation in the development and maintenance of the female sexual characteristics. There are mainly 3 types of hormonal estrogens: E2, which occurs in higher concentrations in women of fertile age, estrone (E1) that predominates in menopausal women and estriol (E3), which is the primary estrogen during pregnancy [142]. The most potent endogenous estrogen is estradiol, which is produced by the growing follicle and its levels vary across the menstrual cycle, peaking just before ovulation [143]. The placenta, adrenal glands, testicles and adipose tissue are also estrogen-secreting organs [143]. Estrogens are also reported in men in the form of estradiol, which is produced by perifereral aromatization of testosterone in adipocytes and tissues that require the action of estrogens such as bones and the brain [144]. Hormonal estrogens act through their binding to the classical alpha and beta ERs (ERα, ERβ). These classical ERs are ligand-activated transcription factors that once bound to estrogens form dimers that interact with ER response elements (EREs) located in the promoter regions of some genes [145]. In addition, classical ERs can modulate other genes without interacting with ERE regions by the regulation of different signalling pathways [142]. Furthermore, hormonal estrogens may also activate different signalling pathways acting on G-protein coupled ER [146]. ERs are expressed in different tissues, being predominant the ERα in women with greater expression not only in the uterus, but also in liver, adipose tissues, skeletal muscle, pituitary and hypothalamus, while ERβ is present in ovaries, prostate and in the brain [147], suggesting an important role of ERs in different tissues [148].

For many years, it has been known that hormonal estrogens act in different organs such as bones, heart and brain, exerting effects beyond the control of reproductive function [149]. ERs have been identified in diverse brain regions such as the neocortex, amygdala and hippocampus, suggesting an effect of hormonal estrogens on cognition, mood and other general brain functions. Furthermore, the expression of ERs has been identified in microglia, neurons and astrocytes [122, 149-151].

Phytoestrogens

Phytoestrogens are estrogenic molecules produced by plants that are involved in vegetal development, healing and reproduction. Phytoestrogens are mainly found in the Leguminosae family and occur in different parts of the plant such as seeds and fruits [152]. Phytoestrogens are classified into 4 groups: isoflavones, lignans, coumestans and stilbenes, and these can be obtained through diet [153]. Isoflavones have been extensively studied, among which daidzein and genistein are the most recognized compounds and are found mainly in soy and red clover. Lignans are found in linseed, in grains such as wheat, rye and oat, and in various types of berries. Coumestans are present in small amounts in red clover and alfalfa, and lower concentrations are found in lima bean and sunflower seeds [154]. Of the stilbenes, the most recognized is resveratrol that is present in the skin of grapes, peanuts and cranberries [153, 154].

Once in the intestine, intestinal bacteria hydrolyze phytoestrogen to the aglycone form by removing the sugar residue(s). The aglycones are absorbed by the enterocytes, conjugated in the liver and released into the blood. In some cases, metabolites of phytoestrogens are absorbed easier than the parent compound; for example, phytoestrogen enterolactone is produced from lignanosecoisolariciresinol by the action of colon bacteria, which is then absorbed by the enterocytes and released into the bloodstream [152].

Phytoestrogens have attracted the attention because of their structural similarity with estradiol. This particular characteristic has positioned the phytoestrogens as natural alternatives for the treatment of menopausal symptoms. Phytoestrogens are also considered potential complementary treatments for osteoporosis, cardiovascular diseases and CNS diseases [154]. The neuroprotective activity of phytoestrogens may be mediated by the affinity of these compounds to ERs, in particular to ERβ [154].

Few studies have focused on determining the effect of phytoestrogens on astrocytes. Among phytoestrogens, the most studied have been genistein and daidzein, which increase iron transport, increased as a result of oxidative stress [155], protect DNA and mitochondrial function [156], and exert anti-inflammatory effect by inducing the expression of antioxidant enzymes such as inducible nitric oxide synthase (iNOS) and cyclooxygenase (COXs) [157] and peroxisome proliferator-activated receptor (PPARs) [158] in different astrocytic cell lines.

Studies performed by Soltani et al. [159] have revealed a protective effect of the isoflavone genistein against TBI by inhibiting the development of cerebral oedema, suppressing the increase of intracranial pressure and diminishing motor damage. Genistein has also shown to reduce excitotoxic neuronal death in the rat hippocampus, induced by the systemic administration of kainic acid [122]. Similarly, resveratrol has been shown to protect brain functions against hypoxia-ischemia induced by neonatal rat brain injury that causes impairment of sensorimotor abilities, neurological reflexes, learning and memory [160]. Despite the above-mentioned evidence, more research is needed to better understand the mechanisms underlying the action of phytoestrogens in the brain, in order to implement the use of these compounds for controlling the symptoms of estrogen deficiency and protecting brain function in postmenopausal women.

Synthetic Estrogenic Compounds

Selective ER Modulators

SERMs are synthetic molecules, which depending on the tissue act as ER agonists or antagonists. The first synthesized SERM was tamoxifen. This molecule was first considered an antagonist of ERs due to its antagonistic effect in ER-positive breast cancers. Later, it was discovered that tamoxifen acts as an ER agonist in other tissues and the concept of SERM was developed. Nevertheless, it is possible to find in the market first-generation SERMs, such as tamoxifen and toremifene that are used in the treatment of breast cancer. The second-generation drug, raloxifene, is an antagonist of ERs in ER-positive uterine and mammary cancer cells, while it is used as an ER agonist in bone for the prevention of osteoporosis. Finally, there are also third-generation SERMs including bazedoxifene, lasofoxifene and ospemifene that are being used primarily in combination therapies with other estrogenic compounds for the treatment of postmenopausal symptoms [161, 162].

The binding of SERMs to the ligand-binding domain of ERs causes specific conformational changes in the receptors that determine which tissue-specific coregulators of transcription could be recruited to the transcriptional complex [162]. Thus, the transcriptional response to SERMs varies according to the specific SERM, the specific ER and the presence of tissue-specific transcriptional cofactors, resulting in different transcriptional outcomes in different tissues and organs [162, 163].

SERMs have been extensively studied for their agonist activities and selective effects in the brain, acting on neurons, glial cells, astrocytes and microglia. Some SERMs have neuroprotective effects by acting as antioxidants, decreasing neuroinflammation and gliosis, stimulating neurotrophic factors, such as the brain-derived neurotrophic factor (BDNF), regulating mitochondrial functions and promoting neuronal survival [8, 128, 164]. In addition, SERMs, such as raloxifene and tamoxifen, have been shown to be neuroprotective in animal models of Parkinson’s disease by stimulating the phosphatidyl-inositol 3-kinase (PI3K)/Akt/GSK3β signalling pathway [161, 165]. Furthermore, tamoxifen promotes functional recovery in animal models of spinal cord injury [270].

Tibolone

Tibolone is a synthetic steroid that belongs to a group of molecules so-called selective tissue estrogenic activity regulators (STEARs), which have estrogenic, progestogenic and androgenic properties. Selective tissue estrogenic activity regulators, in particular tibolone, are also used in hormone therapy aiming at the reduction of postmenopausal symptoms [166].

When tibolone is metabolized, 3 steroid compounds are produced: delta-4 isomer, a 3-alpha-hydroxy metabolite and a 3-beta-hydroxyl metabolite. The 3-OH metabolites bind to ER, and the delta-4 isomer binds to both androgen and progesterone receptors [167]. A study performed on ovariectomized cynomolgus monkeys treated orally with 0.5 mg/kg/day tibolone for 36 days showed an increase of estrogenic metabolites in the hypothalamus that was correlated with a reduction in the number and severity of hot flushes and perspirations. The combined effect of tibolone on androgen receptor and ERs is responsible for improving the libido, mood and general well-being [168]. Also, it has been reported in an in vivo study that an increase in antioxidant activity may underlie the neuroprotective effects of tibolone. Specifically, the treatment of Wistar adult rats with 1 mg/kg of tibolone for 30 days prevented the oxidation of membrane lipids and proteins in the hippocampus. Furthermore, the administration of the same dose of tibolone for 60 days decreased neuronal cell death caused by ozone in the CA3 region of the hippocampus [264]. Given the above-mentioned findings, tibolone has also been proposed as a therapeutic agent for brain damage including TBI because of the ability of its metabolites to act on different sex steroid receptors, such as ERα and ERβ, and improving physiological functions. In this regard, astrocytes are postulated as mediators of the action of tibolone in the CNS [118]. Indeed, a recent study has shown that tibolone reduces reactive astrogliosis in a model of TBI in ovariectomized female mice [169].

Estrogenic compounds reduce the extent of the injury, the sensorimotor and working memory deficits, and reactive astrogliosis after TBI [123, 124, 133, 170-175]. For instance, estradiol has been shown to reduce intracranial pressure, oedema formation and BBB permeability in female rats after TBI [176]. The SERM raloxifene increases neurotransmitter receptor binding in brain regions associated with cognition and memory, and enhances the transcription and protein levels of neurotrophic factors such as tropomyosin receptor kinase A (TrkA), BDNF and ChAT in experimental models of TBI in rats [177]. Another SERM, tamoxifen, has been shown to reduce tissue damage and promote functional recovery after traumatic injury of the spinal cord [202]. The phytoestrogen resveratrol reduced neuronal loss in all ipsi- and contralateral hippocampal regions while improving exploratory activity and memory abilities in albino Wistar rats submitted to TBI [178]. Another phytoestrogen, genistein, reduced cerebral oedema, BBB permeability and intracranial pressure while improving motor conditions [159]. Similar protective actions have been reported for puerarin, an isoflavone glycoside isolated from the Chinese herb Radix Puerariae [179, 180].

Mechanisms Involved in the Neuroprotective Actions of Estrogenic Compounds after TBI That Are Mediated by Astrocytes

In vitro studies indicate that astroglial cells are directly or indirectly regulated by estrogenic compounds. Direct actions have been demonstrated in primary rodent astrocytes cultures where, for instance, estradiol regulates the activation of ERK1/2, PI3K and NF-κB signalling [181, 182]. That astrocytes may be direct targets of estrogenic compounds is also suggested by the finding that they express ERs both in vitro [181, 183-188] and in vivo, in particular after brain injury [14, 128, 151, 164, 189-195]. In addition, there is evidence that estradiol anti-inflammatory and protective effects in experimental autoimmune encephalomyelitis is dependent on the expression of ERα in astrocytes [196-198], indicating that these cells may mediate neuroprotective actions of estrogenic compounds. In the next subsections, we review the mechanisms regulated by estrogenic compounds in astrocytes that may mediate neuroprotection after TBI (Table 4).

Table 4.

Estrogenic compounds and mechanisms of astrocytic-induced neuroprotection in TBI

Estrogenic compounds and mechanisms of astrocytic-induced neuroprotection in TBI
Estrogenic compounds and mechanisms of astrocytic-induced neuroprotection in TBI

Regulation of Astrogliosis

Estrogenic compounds may have beneficial effects after brain injury through the regulation of reactive gliosis and the production of different molecules by reactive astrocytes [123]. In vivo studies have shown that several estrogenic compounds, such as estradiol, phytoestrogens (genistein), some SERMs and tibolone, decrease reactive astrogliosis and glial scar formation after TBI [123, 169-171, 199-201]. After spinal cord injury, tamoxifen has been shown to first increase astrogliosis at day 2 after injury and then gradually decrease astrogliosis [202]. This suggests that tamoxifen may accelerate the process of astrogliosis, increasing reactive astrocytes in the acute injury phase, where astrocytes may release trophic factors to protect neurons, and decrease reactive astrocytes in the chronic injury phase.

Regulation of Excitotoxicity

Glutamate transporters are related to excitotoxic brain injury and neurodegenerative diseases. Excitotoxic damage is produced by a dysregulation of astrocytic gluta mate carriers, such as glutamate transporter-1, which is responsible for absorbing the majority of glutamate from the synaptic cleft, maintaining optimal glutamate levels [203]. Excessive extracellular glutamate leads to a dysregulation in calcium, sodium and potassium fluxes, which may open the mitochondrial permeabilty transition pore. The opening of the mitochondrial pore could liberate ROS and proteins that may lead to apoptosis [203, 204].

Estradiol, other several estrogenic compounds and genistein have been shown to decrease excitotoxic damage in the brain in vivo [122, 126, 205-209]. Estradiol and tamoxifen increase the expression of the glutamate transporters glutamate aspartate transporter (GLAST) and glutamate transporter-1, increasing glutamate uptake by astrocytes [181, 196, 197]. This estrogenic action is mediated by ERs, including G-protein coupled ER, and the PI3K and Mitogen-activated protein kinase signalling pathways [185, 196, 197, 210]. The phytoestrogen resveratrol increases glutamate uptake and glutamine synthetase activity in primary cortical astrocytes [211]. This action may be involved in the protective actions of phytoestrogens against excitotoxic damage [159, 211, 212].

Regulation of Oxidative Stress

Different estrogenic compounds, such as tamoxifen, hydroxytamoxifen, genistein and raloxifene, have been shown to control oxidative stress in the brain tissue [155, 159, 213, 214]. This action is exerted by increasing the activities of several detoxifying enzymes such as superoxide dismutase, catalase and glutathione peroxidase [156, 159, 212, 215]. In astrocytes, estrogenic compounds modulate mitochondrial gene expression, increase clearance of ROS and decrease the loss of antioxidant glutathione [10, 179]. In an in vitro model of ischemia (oxygen-glucose deprivation/reperfusion) in primary astrocytes 17β-estradiol and tibolone decreased the production of ROS, recovered the mitochondrial membrane potential and ATP production and decreased cell death induced by mitochondrial dysfunction [216]. The actions of estrogenic compounds on oxidative stress after brain injury may be associated with the reduction in edema formation [217]. For instance, resveratrol decreases lipid peroxidation in the trauma zone and it is known that lipid peroxidation contributes to ischemia and cerebral edema [160, 217], and worsens the prognosis of patients with TBI. Additionally, genistein and daidzein increase iron transport, as a result of oxidative stress [155], protect DNA and mitochondrial function [156], and exert anti-inflammatory effect by inducing the expression of antioxidant enzymes such as thioredoxin, MnSOD, iNOS and COXs [157] and PPARs in different astrocytic cell lines [158].

Regulation of Mitochondrial Function

An important neuroprotective mechanism of estrogenic compounds is the regulation of mitochondrial function [218-220]. Mitochondria are vital for the maintenance and homeostasis of different brain cells and their dysfunction has been associated with several neurological disorders [221, 222]. Some investigations suggest that mitochondria can be regulated by estrogenic compounds and their interactions with ERs. For example, it has been demonstrated that ERβ is located in the mitochondria of various brain cells including astrocytes [223]. Therefore, it has been proposed that the neuroprotective actions of estrogenic compounds linked to mitochondria are mediated by ERβ [223]. ERβ located in the mitochondria may prevent mitochondrial expression of apoptotic molecules and regulate the function and production of other proteins involved in the reduction of ROS [219].

Regulation of Neuroinflammation

Astrocytes and microglia play an important role in inflammatory responses after TBI, as these cells release growth factors, cytokines and chemokines. These molecules could act as modulators for the beginning and progression of post-traumatic inflammation. Thus, the participation of these cells may be causing extra cell death and neurological damage [8, 164, 224]. Estrogenic compounds, like estradiol, SERMs and phytoestrogens, act on astrocytes and microglial cells reducing neuroinflammation through a decrease in the expression of IL-1β, IL-6, tumour necrosis factor (TNF-α) and (TNF-β) [131, 225-228]. The anti-inflammatory actions of estrogenic compounds on these cells begin with the activation of signalling pathways (Mitogen-activated protein kinase, PI3K, and Akt), which in turn control the activity of transcription factors such as cAMP response element-binding and NF-κB [119, 225], a powerful transcriptional regulator of several inflammatory genes [224].

It is clear that the role of astrocytes on TBI is dependent upon the stage of the pathological mechanism. This is especially important when designing potential protective strategies aimed at controlling reactive astrogliosis and astrocytic double-edged functions. This review highlights the neuroprotective actions of estrogenic compounds and their action on astrocytes in the context of TBI. The response of the wounded nervous tissue to estrogenic compounds is contingent on the interface of multiple cell types, like microglia, tanycytes and astrocytes. One of the most important cells is astrocytes that play vital roles in TBI and in post-TBI synaptic plasticity. This response is determined by specific signalling mechanisms and depends on the nature and severity of tissue damage. Besides, astrocytes have shown to be targets of estrogen and estrogenic compounds and are postulated to have a key role in estrogen-mediated protection of the brain. These compounds have a promising potential as a target for therapeutic approaches for the prevention of cognition decline and neurodegeneration caused by brain trauma. However, further studies are needed to deepen the mechanisms of action of estrogenic compounds in astrocytes and how they trigger a neuroprotective response in TBI.

This work is in part funded by Colciencias grant (Contract No. 824-2017) to G.E.B.

The authors have no ethical conflicts to disclose.

The authors declare no conflicts of interest.

1.
Prins
M
,
Greco
T
,
Alexander
D
,
Giza
CC
.
The pathophysiology of traumatic brain injury at a glance
.
Dis Model Mech
.
2013
Nov
;
6
(
6
):
1307
15
.
[PubMed]
1754-8403
2.
Barreto
GE
,
Gonzalez
J
,
Torres
Y
,
Morales
L
.
Astrocytic-neuronal crosstalk: implications for neuroprotection from brain injury
.
Neurosci Res
.
2011
Oct
;
71
(
2
):
107
13
.
[PubMed]
0168-0102
3.
Heegaard
W
,
Biros
M
.
Traumatic brain injury
.
Emerg Med Clin North Am
.
2007
Aug
;
25
(
3
):
655
78
.
[PubMed]
0733-8627
4.
Lovasik
D
,
Kerr
ME
,
Alexander
S
.
Traumatic brain injury research: a review of clinical studies
.
Crit Care Nurs Q
.
2001
Feb
;
23
(
4
):
24
41
.
[PubMed]
0887-9303
5.
Santiago
LA
,
Oh
BC
,
Dash
PK
,
Holcomb
JB
,
Wade
CE
.
A clinical comparison of penetrating and blunt traumatic brain injuries
.
Brain Inj
.
2012
;
26
(
2
):
107
25
.
[PubMed]
0269-9052
6.
Giustini
A
,
Pistarini
C
,
Pisoni
C
.
Traumatic and nontraumatic brain injury
.
Handb Clin Neurol
.
2013
;
110
:
401
9
.
[PubMed]
0072-9752
7.
Burda
JE
,
Bernstein
AM
,
Sofroniew
MV
.
Astrocyte roles in traumatic brain injury
.
Exp Neurol
.
2015
.
[PubMed]
0014-4886
8.
Arevalo
MA
,
Santos-Galindo
M
,
Lagunas
N
,
Azcoitia
I
,
Garcia-Segura
LM
.
Selective estrogen receptor modulators as brain therapeutic agents
.
J Mol Endocrinol
.
2011
Jan
;
46
(
1
):
R1
9
.
[PubMed]
0952-5041
9.
Kim
JV
,
Dustin
ML
.
Innate response to focal necrotic injury inside the blood-brain barrier
.
J Immunol
.
2006
Oct
;
177
(
8
):
5269
77
.
[PubMed]
0022-1767
10.
Zhao
L
,
O’Neill
K
,
Diaz Brinton
R
.
Selective estrogen receptor modulators (SERMs) for the brain: current status and remaining challenges for developing NeuroSERMs
.
Brain Res Brain Res Rev
.
2005
Nov
;
49
(
3
):
472
93
.
[PubMed]
0165-0173
11.
de Medeiros
AR
,
Lamas
AZ
,
Caliman
IF
,
Dalpiaz
PL
,
Firmes
LB
,
de Abreu
GR
, et al
Tibolone has anti-inflammatory effects in estrogen-deficient female rats on the natriuretic peptide system and TNF-alpha
.
Regul Pept
.
2012
Nov
;
179
(
1-3
):
55
60
.
[PubMed]
0167-0115
12.
Khan
MM
,
Wakade
C
,
de Sevilla
L
,
Brann
DW
.
Selective estrogen receptor modulators (SERMs) enhance neurogenesis and spine density following focal cerebral ischemia
.
J Steroid Biochem Mol Biol
.
2015
Feb
;
146
:
38
47
.
[PubMed]
0960-0760
13.
Dhandapani
K
,
Brann
D
.
Neuroprotective effects of estrogen and tamoxifen in vitro: a facilitative role for glia?
Endocrine
.
2003
Jun
;
21
(
1
):
59
66
.
[PubMed]
1355-008X
14.
Acaz-Fonseca
E
,
Sanchez-Gonzalez
R
,
Azcoitia
I
,
Arevalo
MA
,
Garcia-Segura
LM
.
Role of astrocytes in the neuroprotective actions of 17β-estradiol and selective estrogen receptor modulators
.
Mol Cell Endocrinol
.
2014
May
;
389
(
1-2
):
48
57
.
[PubMed]
0303-7207
15.
Arevalo
MA
,
Azcoitia
I
,
Garcia-Segura
LM
.
The neuroprotective actions of oestradiol and oestrogen receptors
.
Nat Rev Neurosci
.
2015
Jan
;
16
(
1
):
17
29
.
[PubMed]
1471-003X
16.
Escobedo
LV
,
Habboushe
J
,
Kaafarani
H
,
Velmahos
G
,
Shah
K
,
Lee
J
.
Traumatic brain injury: A case-based review
.
World J Emerg Med
.
2013
;
4
(
4
):
252
9
.
[PubMed]
1920-8642
17.
Dinsmore
J
.
Traumatic brain injury: an evidence-based review of management
.
Contin Educ Anaesth Crit Care Pain
.
2013
;
13
(
6
):
189
95
. 1743-1816
18.
Andersen
LW
,
Mackenhauer
J
,
Roberts
JC
,
Berg
KM
,
Cocchi
MN
,
Donnino
MW
.
Etiology and therapeutic approach to elevated lactate levels
.
Mayo Clin Proc
.
2013
Oct
;
88
(
10
):
1127
40
.
[PubMed]
0025-6196
19.
Yousefzadeh-Chabok
S
,
Dehnadi Moghaddam
A
,
Kazemnejad-Leili
E
,
Saneei
Z
,
Hosseinpour
M
,
Kouchakinejad-Eramsadati
L
, et al
The Relationship Between Serum Levels of Interleukins 6, 8, 10 and Clinical Outcome in Patients With Severe Traumatic Brain Injury
.
Arch Trauma Res
.
2015
Feb
;
4
(
1
):
e18357
18357
.
[PubMed]
2251-953X
20.
Ray
SK
,
Dixon
CE
,
Banik
NL
.
Molecular mechanisms in the pathogenesis of traumatic brain injury
.
Histol Histopathol
.
2002
Oct
;
17
(
4
):
1137
52
.
[PubMed]
0213-3911
21.
Downard
C
,
Hulka
F
,
Mullins
RJ
,
Piatt
J
,
Chesnut
R
,
Quint
P
, et al
Relationship of cerebral perfusion pressure and survival in pediatric brain-injured patients
.
J Trauma
.
2000
Oct
;
49
(
4
):
654
8
.
[PubMed]
0022-5282
22.
Saatman
KE
,
Duhaime
AC
,
Bullock
R
,
Maas
AI
,
Valadka
A
,
Manley
GT
;
Workshop Scientific Team and Advisory Panel Members
.
Classification of traumatic brain injury for targeted therapies
.
J Neurotrauma
.
2008
Jul
;
25
(
7
):
719
38
.
[PubMed]
0897-7151
23.
Greve
MW
,
Zink
BJ
.
Pathophysiology of traumatic brain injury
.
Mt Sinai J Med
.
2009
Apr
;
76
(
2
):
97
104
.
[PubMed]
0027-2507
24.
Graham
DI
,
Gennarelli
TA
,
McIntosh
TK
. Trauma. In:
Graham
DI
, editor
.
Greenfield’s Neuropathology, L.P.L
.
London
:
Arnold Publishers
;
2002
. pp.
823
98
.
25.
Donkin
JJ
,
Vink
R
.
Mechanisms of cerebral edema in traumatic brain injury: therapeutic developments
.
Curr Opin Neurol
.
2010
Jun
;
23
(
3
):
293
9
.
[PubMed]
1350-7540
26.
Cook
LG
,
Chapman
SB
,
Levin
HS
.
Self-regulation abilities in children with severe traumatic brain injury: a preliminary investigation of naturalistic action
.
NeuroRehabilitation
.
2008
;
23
(
6
):
467
75
.
[PubMed]
1053-8135
27.
Molaie
AM
,
Maguire
J
.
Neuroendocrine Abnormalities Following Traumatic Brain Injury: An Important Contributor to Neuropsychiatric Sequelae
.
Front Endocrinol (Lausanne)
.
2018
Apr
;
9
:
176
.
[PubMed]
1664-2392
28.
Osterstock
G
,
El Yandouzi
T
,
Romanò
N
,
Carmignac
D
,
Langlet
F
,
Coutry
N
, et al
Sustained alterations of hypothalamic tanycytes during posttraumatic hypopituitarism in male mice
.
Endocrinology
.
2014
May
;
155
(
5
):
1887
98
.
[PubMed]
0013-7227
29.
Mullier
A
,
Bouret
SG
,
Prevot
V
,
Dehouck
B
.
Differential distribution of tight junction proteins suggests a role for tanycytes in blood-hypothalamus barrier regulation in the adult mouse brain
.
J Comp Neurol
.
2010
Apr
;
518
(
7
):
943
62
.
[PubMed]
0021-9967
30.
Prevot
V
,
Dehouck
B
,
Sharif
A
,
Ciofi
P
,
Giacobini
P
,
Clasadonte
J
.
The Versatile Tanycyte: A Hypothalamic Integrator of Reproduction and Energy Metabolism
.
Endocr Rev
.
2018
Jun
;
39
(
3
):
333
68
.
[PubMed]
0163-769X
31.
Stiefel
MF
,
Tomita
Y
,
Marmarou
A
.
Secondary ischemia impairing the restoration of ion homeostasis following traumatic brain injury
.
J Neurosurg
.
2005
Oct
;
103
(
4
):
707
14
.
[PubMed]
0022-3085
32.
Floyd
CL
,
Gorin
FA
,
Lyeth
BG
.
Mechanical strain injury increases intracellular sodium and reverses Na+/Ca2+ exchange in cortical astrocytes
.
Glia
.
2005
Jul
;
51
(
1
):
35
46
.
[PubMed]
0894-1491
33.
Yi
JH
,
Hazell
AS
.
Excitotoxic mechanisms and the role of astrocytic glutamate transporters in traumatic brain injury
.
Neurochem Int
.
2006
Apr
;
48
(
5
):
394
403
.
[PubMed]
0197-0186
34.
DeWitt
DS
,
Prough
DS
.
Traumatic cerebral vascular injury: the effects of concussive brain injury on the cerebral vasculature
.
J Neurotrauma
.
2003
Sep
;
20
(
9
):
795
825
.
[PubMed]
0897-7151
35.
Choi
DW
.
Ischemia-induced neuronal apoptosis
.
Curr Opin Neurobiol
.
1996
Oct
;
6
(
5
):
667
72
.
[PubMed]
0959-4388
36.
Eldadah
BA
,
Faden
AI
.
Caspase pathways, neuronal apoptosis, and CNS injury
.
J Neurotrauma
.
2000
Oct
;
17
(
10
):
811
29
.
[PubMed]
0897-7151
37.
Werner
C
,
Engelhard
K
.
Pathophysiology of traumatic brain injury
.
Br J Anaesth
.
2007
Jul
;
99
(
1
):
4
9
.
[PubMed]
0007-0912
38.
Mattson
MP
,
Magnus
T
.
Ageing and neuronal vulnerability
.
Nat Rev Neurosci
.
2006
Apr
;
7
(
4
):
278
94
.
[PubMed]
1471-003X
39.
Chen
Y
,
Swanson
RA
.
Astrocytes and brain injury
.
J Cereb Blood Flow Metab
.
2003
Feb
;
23
(
2
):
137
49
.
[PubMed]
0271-678X
40.
Bayir
H
,
Kagan
VE
,
Borisenko
GG
,
Tyurina
YY
,
Janesko
KL
,
Vagni
VA
, et al
Enhanced oxidative stress in iNOS-deficient mice after traumatic brain injury: support for a neuroprotective role of iNOS
.
J Cereb Blood Flow Metab
.
2005
Jun
;
25
(
6
):
673
84
.
[PubMed]
0271-678X
41.
Palmer
AM
,
Marion
DW
,
Botscheller
ML
,
Swedlow
PE
,
Styren
SD
,
DeKosky
ST
.
Traumatic brain injury-induced excitotoxicity assessed in a controlled cortical impact model
.
J Neurochem
.
1993
Dec
;
61
(
6
):
2015
24
.
[PubMed]
0022-3042
42.
Toklu
H
,
Tumer
N
. Oxidative Stress, Brain Edema, Blood–Brain Barrier Permeability, and Autonomic Dysfunction from Traumatic Brain Injury.
SourceBrain Neurotrauma: Molecular, Neuropsychological, and Rehabilitation Aspects
.
Boca Raton (FL)
:
CRC Press/Taylor & Francis
;
2015
. pp.
725
725
.
43.
DeWitt
DS
,
Prough
DS
.
Blast-induced brain injury and posttraumatic hypotension and hypoxemia
.
J Neurotrauma
.
2009
Jun
;
26
(
6
):
877
87
.
[PubMed]
0897-7151
44.
Vuceljić
M
,
Zunić
G
,
Romić
P
,
Jevtić
M
.
Relation between both oxidative and metabolic-osmotic cell damages and initial injury severity in bombing casualties
.
Vojnosanit Pregl
.
2006
Jun
;
63
(
6
):
545
51
.
[PubMed]
0042-8450
45.
Raps
SP
,
Lai
JC
,
Hertz
L
,
Cooper
AJ
.
Glutathione is present in high concentrations in cultured astrocytes but not in cultured neurons
.
Brain Res
.
1989
Jul
;
493
(
2
):
398
401
.
[PubMed]
0006-8993
46.
Wang
J
,
Ma
JH
,
Giffard
RG
.
Overexpression of copper/zinc superoxide dismutase decreases ischemia-like astrocyte injury
.
Free Radic Biol Med
.
2005
Apr
;
38
(
8
):
1112
8
.
[PubMed]
0891-5849
47.
Cabezas
R
,
Avila
M
,
Gonzalez
J
,
El-Bachá
RS
,
Báez
E
,
García-Segura
LM
, et al
Astrocytic modulation of blood brain barrier: perspectives on Parkinson’s disease
.
Front Cell Neurosci
.
2014
Aug
;
8
(
AUG
):
211
211
.
[PubMed]
1662-5102
48.
Iglesias
J
,
Morales
L
,
Barreto
GE
.
Metabolic and Inflammatory Adaptation of Reactive Astrocytes: role of PPARs
.
Mol Neurobiol
.
2016
;(
May
):
1
21
.
[PubMed]
0893-7648
49.
Posada-Duque
RA
,
Barreto
GE
,
Cardona-Gomez
GP
.
Protection after stroke: cellular effectors of neurovascular unit integrity
.
Front Cell Neurosci
.
2014
Aug
;
8
(
August
):
231
231
.
[PubMed]
1662-5102
50.
Suzumura
A
.
Neuron-microglia interaction in neuroinflammation
.
Curr Protein Pept Sci
.
2013
Feb
;
14
(
1
):
16
20
.
[PubMed]
1389-2037
51.
Verkhratsky
A
,
Butt
A
.
Glial Physiology and Pathophysiology
.
John Wiley & Sons, Ltd.
;
2013
.
52.
Parpura
V
,
Heneka
MT
,
Montana
V
,
Oliet
SH
,
Schousboe
A
,
Haydon
PG
, et al
Glial cells in (patho)physiology
.
J Neurochem
.
2012
Apr
;
121
(
1
):
4
27
.
[PubMed]
0022-3042
53.
Gavillet
M
,
Allaman
I
,
Magistretti
PJ
.
Modulation of astrocytic metabolic phenotype by proinflammatory cytokines
.
Glia
.
2008
Jul
;
56
(
9
):
975
89
.
[PubMed]
0894-1491
54.
Volterra
A
,
Meldolesi
J
.
Astrocytes, from brain glue to communication elements: the revolution continues
.
Nat Rev Neurosci
.
2005
Aug
;
6
(
8
):
626
40
.
[PubMed]
1471-003X
55.
Navarrete
M
,
Araque
A
.
Basal synaptic transmission: astrocytes rule!
Cell
.
2011
Sep
;
146
(
5
):
675
7
.
[PubMed]
0092-8674
56.
Perea
G
,
Navarrete
M
,
Araque
A
.
Tripartite synapses: astrocytes process and control synaptic information
.
Trends Neurosci
.
2009
Aug
;
32
(
8
):
421
31
.
[PubMed]
0166-2236
57.
Pérez-Alvarez
A
,
Araque
A
.
Astrocyte-neuron interaction at tripartite synapses
.
Curr Drug Targets
.
2013
Oct
;
14
(
11
):
1220
4
.
[PubMed]
1389-4501
58.
Liberto
CM
,
Albrecht
PJ
,
Herx
LM
,
Yong
VW
,
Levison
SW
.
Pro-regenerative properties of cytokine-activated astrocytes
.
J Neurochem
.
2004
Jun
;
89
(
5
):
1092
100
.
[PubMed]
0022-3042
59.
Parpura
V
,
Scemes
E
,
Spray
DC
.
Mechanisms of glutamate release from astrocytes: gap junction “hemichannels”, purinergic receptors and exocytotic release
.
Neurochem Int
.
2004
Jul-Aug
;
45
(
2-3
):
259
64
.
[PubMed]
0197-0186
60.
Volterra
A
,
Steinhäuser
C
.
Glial modulation of synaptic transmission in the hippocampus
.
Glia
.
2004
Aug
;
47
(
3
):
249
57
.
[PubMed]
0894-1491
61.
Sofroniew
MV
,
Vinters
HV
.
Astrocytes: biology and pathology
.
Acta Neuropathol
.
2010
Jan
;
119
(
1
):
7
35
.
[PubMed]
0001-6322
62.
Garcia-Segura
LM
,
Melcangi
RC
.
Steroids and glial cell function
.
Glia
.
2006
Nov
;
54
(
6
):
485
98
.
[PubMed]
0894-1491
63.
Fuente-Martin
E
,
Garcia-Caceres
C
,
Morselli
E
,
Clegg
DJ
,
Chowen
JA
,
Finan
B
, et al
Estrogen, astrocytes and the neuroendocrine control of metabolism
.
Rev Endocr Metab Disord
.
2013
Dec
;
14
(
4
):
331
8
.
[PubMed]
1389-9155
64.
Micevych
P
,
Bondar
G
,
Kuo
J
.
Estrogen actions on neuroendocrine glia
.
Neuroendocrinology
.
2010
;
91
(
3
):
211
22
.
[PubMed]
0028-3835
65.
Wicher
G
,
Norlin
M
.
Estrogen-mediated regulation of steroid metabolism in rat glial cells; effects on neurosteroid levels via regulation of CYP7B1-mediated catalysis
.
J Steroid Biochem Mol Biol
.
2015
Jan
;
145
:
21
7
.
[PubMed]
0960-0760
66.
Bolborea
M
,
Dale
N
.
Hypothalamic tanycytes: potential roles in the control of feeding and energy balance
.
Trends Neurosci
.
2013
Feb
;
36
(
2
):
91
100
.
[PubMed]
0166-2236
67.
Rodríguez
EM
,
Blázquez
JL
,
Pastor
FE
,
Peláez
B
,
Peña
P
,
Peruzzo
B
, et al
Hypothalamic tanycytes: a key component of brain-endocrine interaction
.
Int Rev Cytol
.
2005
;
247
:
89
164
.
[PubMed]
0074-7696
68.
Duncan
SR
.
Estrogen Effects on Traumatic Brain Injury
. 1st ed.
2018
.
69.
Gorina
R
,
Font-Nieves
M
,
Márquez-Kisinousky
L
,
Santalucia
T
,
Planas
AM
.
Astrocyte TLR4 activation induces a proinflammatory environment through the interplay between MyD88-dependent NFκB signaling, MAPK, and Jak1/Stat1 pathways
.
Glia
.
2011
Feb
;
59
(
2
):
242
55
.
[PubMed]
0894-1491
70.
Paintlia
MK
,
Paintlia
AS
,
Singh
AK
,
Singh
I
.
S-nitrosoglutathione induces ciliary neurotrophic factor expression in astrocytes, which has implications to protect the central nervous system under pathological conditions
.
J Biol Chem
.
2013
Feb
;
288
(
6
):
3831
43
.
[PubMed]
0021-9258
71.
Pekny
M
,
Johansson
CB
,
Eliasson
C
,
Stakeberg
J
,
Wallén
A
,
Perlmann
T
, et al
Abnormal reaction to central nervous system injury in mice lacking glial fibrillary acidic protein and vimentin
.
J Cell Biol
.
1999
May
;
145
(
3
):
503
14
.
[PubMed]
0021-9525
72.
Pekny
M
,
Nilsson
M
.
Astrocyte activation and reactive gliosis
.
Glia
.
2005
Jun
;
50
(
4
):
427
34
.
[PubMed]
0894-1491
73.
Silver
J
,
Miller
JH
.
Regeneration beyond the glial scar
.
Nat Rev Neurosci
.
2004
Feb
;
5
(
2
):
146
56
.
[PubMed]
1471-003X
74.
Zamanian
JL
,
Xu
L
,
Foo
LC
,
Nouri
N
,
Zhou
L
,
Giffard
RG
, et al
Genomic analysis of reactive astrogliosis
.
J Neurosci
.
2012
May
;
32
(
18
):
6391
410
.
[PubMed]
0270-6474
75.
Brambilla
R
,
Bracchi-Ricard
V
,
Hu
WH
,
Frydel
B
,
Bramwell
A
,
Karmally
S
, et al
Inhibition of astroglial nuclear factor kappaB reduces inflammation and improves functional recovery after spinal cord injury
.
J Exp Med
.
2005
Jul
;
202
(
1
):
145
56
.
[PubMed]
0022-1007
76.
Herrmann
JE
,
Imura
T
,
Song
B
,
Qi
J
,
Ao
Y
,
Nguyen
TK
, et al
STAT3 is a critical regulator of astrogliosis and scar formation after spinal cord injury
.
J Neurosci
.
2008
Jul
;
28
(
28
):
7231
43
.
[PubMed]
0270-6474
77.
Okada
S
,
Nakamura
M
,
Katoh
H
,
Miyao
T
,
Shimazaki
T
,
Ishii
K
, et al
Conditional ablation of Stat3 or Socs3 discloses a dual role for reactive astrocytes after spinal cord injury
.
Nat Med
.
2006
Jul
;
12
(
7
):
829
34
.
[PubMed]
1078-8956
78.
Wanner
IB
,
Anderson
MA
,
Song
B
,
Levine
J
,
Fernandez
A
,
Gray-Thompson
Z
, et al
Glial scar borders are formed by newly proliferated, elongated astrocytes that interact to corral inflammatory and fibrotic cells via STAT3-dependent mechanisms after spinal cord injury
.
J Neurosci
.
2013
Jul
;
33
(
31
):
12870
86
.
[PubMed]
0270-6474
79.
Eng
LF
,
Ghirnikar
RS
.
GFAP and astrogliosis
.
Brain Pathol
.
1994
Jul
;
4
(
3
):
229
37
.
[PubMed]
1015-6305
80.
Hamby
ME
,
Sofroniew
MV
.
Reactive astrocytes as therapeutic targets for CNS disorders
.
Neurotherapeutics
.
2010
Oct
;
7
(
4
):
494
506
.
[PubMed]
1933-7213
81.
Mucke
L
,
Eddleston
M
.
Astrocytes in infectious and immune-mediated diseases of the central nervous system
.
FASEB J
.
1993
Oct
;
7
(
13
):
1226
32
.
[PubMed]
0892-6638
82.
Pekny
M
,
Pekna
M
.
Astrocyte reactivity and reactive astrogliosis: costs and benefits
.
Physiol Rev
.
2014
Oct
;
94
(
4
):
1077
98
.
[PubMed]
0031-9333
83.
Hernandez
MR
,
Agapova
OA
,
Yang
P
,
Salvador-Silva
M
,
Ricard
CS
,
Aoi
S
.
Differential gene expression in astrocytes from human normal and glaucomatous optic nerve head analyzed by cDNA microarray
.
Glia
.
2002
Apr
;
38
(
1
):
45
64
.
[PubMed]
0894-1491
84.
Sofroniew
MV
.
Molecular dissection of reactive astrogliosis and glial scar formation
.
Trends Neurosci
.
2009
Dec
;
32
(
12
):
638
47
.
[PubMed]
0166-2236
85.
Wilhelmsson
U
,
Bushong
EA
,
Price
DL
,
Smarr
BL
,
Phung
V
,
Terada
M
, et al
Redefining the concept of reactive astrocytes as cells that remain within their unique domains upon reaction to injury
.
Proc Natl Acad Sci USA
.
2006
Nov
;
103
(
46
):
17513
8
.
[PubMed]
0027-8424
86.
McKeon
RJ
,
Schreiber
RC
,
Rudge
JS
,
Silver
J
.
Reduction of neurite outgrowth in a model of glial scarring following CNS injury is correlated with the expression of inhibitory molecules on reactive astrocytes
.
J Neurosci
.
1991
Nov
;
11
(
11
):
3398
411
.
[PubMed]
0270-6474
87.
Bush
TG
,
Puvanachandra
N
,
Horner
CH
,
Polito
A
,
Ostenfeld
T
,
Svendsen
CN
, et al
Leukocyte infiltration, neuronal degeneration, and neurite outgrowth after ablation of scar-forming, reactive astrocytes in adult transgenic mice
.
Neuron
.
1999
Jun
;
23
(
2
):
297
308
.
[PubMed]
0896-6273
88.
Faulkner
JR
,
Herrmann
JE
,
Woo
MJ
,
Tansey
KE
,
Doan
NB
,
Sofroniew
MV
.
Reactive astrocytes protect tissue and preserve function after spinal cord injury
.
J Neurosci
.
2004
Mar
;
24
(
9
):
2143
55
.
[PubMed]
0270-6474
89.
Voskuhl
RR
,
Peterson
RS
,
Song
B
,
Ao
Y
,
Morales
LB
,
Tiwari-Woodruff
S
, et al
Reactive astrocytes form scar-like perivascular barriers to leukocytes during adaptive immune inflammation of the CNS
.
J Neurosci
.
2009
Sep
;
29
(
37
):
11511
22
.
[PubMed]
0270-6474
90.
Garcia-Segura
LM
,
Wozniak
A
,
Azcoitia
I
,
Rodriguez
JR
,
Hutchison
RE
,
Hutchison
JB
.
Aromatase expression by astrocytes after brain injury: implications for local estrogen formation in brain repair
.
Neuroscience
.
1999
Mar
;
89
(
2
):
567
78
.
[PubMed]
0306-4522
91.
Alilain
WJ
,
Horn
KP
,
Hu
H
,
Dick
TE
,
Silver
J
.
Functional regeneration of respiratory pathways after spinal cord injury
.
Nature
.
2011
Jul
;
475
(
7355
):
196
200
.
[PubMed]
0028-0836
92.
Bradbury
EJ
,
Moon
LD
,
Popat
RJ
,
King
VR
,
Bennett
GS
,
Patel
PN
, et al
Chondroitinase ABC promotes functional recovery after spinal cord injury
.
Nature
.
2002
Apr
;
416
(
6881
):
636
40
.
[PubMed]
0028-0836
93.
Fitch
MT
,
Silver
J
.
CNS injury, glial scars, and inflammation: inhibitory extracellular matrices and regeneration failure
.
Exp Neurol
.
2008
Feb
;
209
(
2
):
294
301
.
[PubMed]
0014-4886
94.
Burda
JE
,
Sofroniew
MV
.
Reactive gliosis and the multicellular response to CNS damage and disease
.
Neuron
.
2014
Jan
;
81
(
2
):
229
48
.
[PubMed]
0896-6273
95.
Sofroniew
MV
.
Astrogliosis
.
Cold Spring Harb Perspect Biol
.
2014
Nov
;
7
(
2
):
a020420
.
[PubMed]
1943-0264
96.
Zador
Z
,
Stiver
S
,
Wang
V
,
Manley
GT
.
Role of aquaporin-4 in cerebral edema and stroke
.
Handb Exp Pharmacol
.
2009
;
190
(
190
):
159
70
.
[PubMed]
0171-2004
97.
Argaw
AT
,
Gurfein
BT
,
Zhang
Y
,
Zameer
A
,
John
GR
.
VEGF-mediated disruption of endothelial CLN-5 promotes blood-brain barrier breakdown
.
Proc Natl Acad Sci USA
.
2009
Feb
;
106
(
6
):
1977
82
.
[PubMed]
0027-8424
98.
Bell
RD
,
Winkler
EA
,
Singh
I
,
Sagare
AP
,
Deane
R
,
Wu
Z
, et al
Apolipoprotein E controls cerebrovascular integrity via cyclophilin A
.
Nature
.
2012
May
;
485
(
7399
):
512
6
.
[PubMed]
0028-0836
99.
De Keyser
J
,
Mostert
JP
,
Koch
MW
.
Dysfunctional astrocytes as key players in the pathogenesis of central nervous system disorders
.
J Neurol Sci
.
2008
Apr
;
267
(
1-2
):
3
16
.
[PubMed]
0022-510X
100.
Gris
P
,
Tighe
A
,
Levin
D
,
Sharma
R
,
Brown
A
.
Transcriptional regulation of scar gene expression in primary astrocytes
.
Glia
.
2007
Aug
;
55
(
11
):
1145
55
.
[PubMed]
0894-1491
101.
Colodner
KJ
,
Montana
RA
,
Anthony
DC
,
Folkerth
RD
,
De Girolami
U
,
Feany
MB
.
Proliferative potential of human astrocytes
.
J Neuropathol Exp Neurol
.
2005
Feb
;
64
(
2
):
163
9
.
[PubMed]
0022-3069
102.
Horner
PJ
,
Power
AE
,
Kempermann
G
,
Kuhn
HG
,
Palmer
TD
,
Winkler
J
, et al
Proliferation and differentiation of progenitor cells throughout the intact adult rat spinal cord
.
J Neurosci
.
2000
Mar
;
20
(
6
):
2218
28
.
[PubMed]
0270-6474
103.
Norton
WT
,
Aquino
DA
,
Hozumi
I
,
Chiu
FC
,
Brosnan
CF
.
Quantitative aspects of reactive gliosis: a review
.
Neurochem Res
.
1992
Sep
;
17
(
9
):
877
85
.
[PubMed]
0364-3190
104.
Gadea
A
,
Schinelli
S
,
Gallo
V
.
Endothelin-1 regulates astrocyte proliferation and reactive gliosis via a JNK/c-Jun signaling pathway
.
J Neurosci
.
2008
Mar
;
28
(
10
):
2394
408
.
[PubMed]
0270-6474
105.
Levison
SW
,
Jiang
FJ
,
Stoltzfus
OK
,
Ducceschi
MH
.
IL-6-type cytokines enhance epidermal growth factor-stimulated astrocyte proliferation
.
Glia
.
2000
Dec
;
32
(
3
):
328
37
.
[PubMed]
0894-1491
106.
Neary
JT
,
Zimmermann
H
.
Trophic functions of nucleotides in the central nervous system
.
Trends Neurosci
.
2009
Apr
;
32
(
4
):
189
98
.
[PubMed]
0166-2236
107.
Buffo
A
,
Rite
I
,
Tripathi
P
,
Lepier
A
,
Colak
D
,
Horn
AP
, et al
Origin and progeny of reactive gliosis: A source of multipotent cells in the injured brain
.
Proc Natl Acad Sci USA
.
2008
Mar
;
105
(
9
):
3581
6
.
[PubMed]
0027-8424
108.
Magnus
T
,
Carmen
J
,
Deleon
J
,
Xue
H
,
Pardo
AC
,
Lepore
AC
, et al
Adult glial precursor proliferation in mutant SOD1G93A mice
.
Glia
.
2008
Jan
;
56
(
2
):
200
8
.
[PubMed]
0894-1491
109.
Carlén
M
,
Meletis
K
,
Göritz
C
,
Darsalia
V
,
Evergren
E
,
Tanigaki
K
, et al
Forebrain ependymal cells are Notch-dependent and generate neuroblasts and astrocytes after stroke
.
Nat Neurosci
.
2009
Mar
;
12
(
3
):
259
67
.
[PubMed]
1097-6256
110.
Meletis
K
,
Barnabé-Heider
F
,
Carlén
M
,
Evergren
E
,
Tomilin
N
,
Shupliakov
O
, et al
Spinal cord injury reveals multilineage differentiation of ependymal cells
.
PLoS Biol
.
2008
Jul
;
6
(
7
):
e182
.
[PubMed]
1544-9173
111.
Ohab
JJ
,
Carmichael
ST
.
Poststroke neurogenesis: emerging principles of migration and localization of immature neurons
.
Neuroscientist
.
2008
Aug
;
14
(
4
):
369
80
.
[PubMed]
1073-8584
112.
Rothstein
JD
,
Dykes-Hoberg
M
,
Pardo
CA
,
Bristol
LA
,
Jin
L
,
Kuncl
RW
, et al
Knockout of glutamate transporters reveals a major role for astroglial transport in excitotoxicity and clearance of glutamate
.
Neuron
.
1996
Mar
;
16
(
3
):
675
86
.
[PubMed]
0896-6273
113.
Swanson
RA
,
Ying
W
,
Kauppinen
TM
.
Astrocyte influences on ischemic neuronal death
.
Curr Mol Med
.
2004
Mar
;
4
(
2
):
193
205
.
[PubMed]
1566-5240
114.
Colangelo
AM
,
Alberghina
L
,
Papa
M
.
Astrogliosis as a therapeutic target for neurodegenerative diseases
.
Neurosci Lett
.
2014
Apr
;
565
:
59
64
.
[PubMed]
0304-3940
115.
Drögemüller
K
,
Helmuth
U
,
Brunn
A
,
Sakowicz-Burkiewicz
M
,
Gutmann
DH
,
Mueller
W
, et al
Astrocyte gp130 expression is critical for the control of Toxoplasma encephalitis
.
J Immunol
.
2008
Aug
;
181
(
4
):
2683
93
.
[PubMed]
0022-1767
116.
Tian
DS
,
Yu
ZY
,
Xie
MJ
,
Bu
BT
,
Witte
OW
,
Wang
W
.
Suppression of astroglial scar formation and enhanced axonal regeneration associated with functional recovery in a spinal cord injury rat model by the cell cycle inhibitor olomoucine
.
J Neurosci Res
.
2006
Oct
;
84
(
5
):
1053
63
.
[PubMed]
0360-4012
117.
Montgomery
DL
.
Astrocytes: form, functions, and roles in disease
.
Vet Pathol
.
1994
Mar
;
31
(
2
):
145
67
.
[PubMed]
0300-9858
118.
Acaz-Fonseca
E
,
Avila-Rodriguez
M
,
Garcia-Segura
LM
,
Barreto
GE
.
Regulation of astroglia by gonadal steroid hormones under physiological and pathological conditions
.
Prog Neurobiol
.
2016
Sep
;
144
:
5
26
.
[PubMed]
0301-0082
119.
Arevalo
MA
,
Santos-Galindo
M
,
Bellini
MJ
,
Azcoitia
I
,
Garcia-Segura
LM
.
Actions of estrogens on glial cells: implications for neuroprotection
.
Biochim Biophys Acta
.
2010
Oct
;
1800
(
10
):
1106
12
.
[PubMed]
0006-3002
120.
Avila Rodriguez
,
M.
, et al
, Tibolone protects T98G cells from glucose deprivation. Journal of Steroid Biochemistry and Molecular Biology,
2014
. 144(PART B): p. 294-303.
121.
Avila-Rodriguez
M
,
Garcia-Segura
LM
,
Hidalgo-Lanussa
O
,
Baez
E
,
Gonzalez
J
,
Barreto
GE
.
Tibolone protects astrocytic cells from glucose deprivation through a mechanism involving estrogen receptor beta and the upregulation of neuroglobin expression
.
Mol Cell Endocrinol
.
2016
Sep
;
433
:
35
46
.
[PubMed]
0303-7207
122.
Azcoitia
I
,
Moreno
A
,
Carrero
P
,
Palacios
S
,
Garcia-Segura
LM
.
Neuroprotective effects of soy phytoestrogens in the rat brain
.
Gynecol Endocrinol
.
2006
Feb
;
22
(
2
):
63
9
.
[PubMed]
0951-3590
123.
Barreto
G
,
Santos-Galindo
M
,
Diz-Chaves
Y
,
Pernía
O
,
Carrero
P
,
Azcoitia
I
, et al
Selective estrogen receptor modulators decrease reactive astrogliosis in the injured brain: effects of aging and prolonged depletion of ovarian hormones
.
Endocrinology
.
2009
Nov
;
150
(
11
):
5010
5
.
[PubMed]
0013-7227
124.
Barreto
GE
,
Santos-Galindo
M
,
Garcia-Segura
LM
.
Selective estrogen receptor modulators regulate reactive microglia after penetrating brain injury
.
Front Aging Neurosci
.
2014
Jun
;
6
(
JUN
):
132
.
[PubMed]
1663-4365
125.
Callier
S
,
Morissette
M
,
Grandbois
M
,
Pélaprat
D
,
Di Paolo
T
.
Neuroprotective properties of 17beta-estradiol, progesterone, and raloxifene in MPTP C57Bl/6 mice
.
Synapse
.
2001
Aug
;
41
(
2
):
131
8
.
[PubMed]
0887-4476
126.
Ciriza
I
,
Carrero
P
,
Azcoitia
I
,
Lundeen
SG
,
Garcia-Segura
LM
.
Selective estrogen receptor modulators protect hippocampal neurons from kainic acid excitotoxicity: differences with the effect of estradiol
.
J Neurobiol
.
2004
Nov
;
61
(
2
):
209
21
.
[PubMed]
0022-3034
127.
Dhandapani
KM
,
Brann
DW
.
Protective effects of estrogen and selective estrogen receptor modulators in the brain
.
Biol Reprod
.
2002
Nov
;
67
(
5
):
1379
85
.
[PubMed]
0006-3363
128.
DonCarlos
,
L.L.A.I.
and
L.M.
Garcia-Segura
,
Neuroprotective actions of selective estrogen receptor modulators.
Psychoneuroendocrinology,
2009
. 34S1: p. S113--S122.
129.
Garcia-Ovejero
D
,
Azcoitia
I
,
Doncarlos
LL
,
Melcangi
RC
,
Garcia-Segura
LM
.
Glia-neuron crosstalk in the neuroprotective mechanisms of sex steroid hormones
.
Brain Res Brain Res Rev
.
2005
Apr
;
48
(
2
):
273
86
.
[PubMed]
0165-0173
130.
González-Giraldo
Y
,
Garcia-Segura
LM
,
Echeverria
V
,
Barreto
GE
.
Tibolone Preserves Mitochondrial Functionality and Cell Morphology in Astrocytic Cells Treated with Palmitic Acid
.
Mol Neurobiol
.
2018
May
;
55
(
5
):
4453
62
.
[PubMed]
1559-1182
131.
Hidalgo-Lanussa
O
, et al
Tibolone Reduces Oxidative Damage and Inflammation in Microglia Stimulated with Palmitic Acid through Mechanisms Involving Estrogen Receptor Beta
.
Mol Neurobiol
.
2017
.
[PubMed]
0893-7648
132.
Kimelberg
HK
,
Jin
Y
,
Charniga
C
,
Feustel
PJ
.
Neuroprotective activity of tamoxifen in permanent focal ischemia
.
J Neurosurg
.
2003
Jul
;
99
(
1
):
138
42
.
[PubMed]
0022-3085
133.
Kokiko
ON
,
Murashov
AK
,
Hoane
MR
.
Administration of raloxifene reduces sensorimotor and working memory deficits following traumatic brain injury
.
Behav Brain Res
.
2006
Jun
;
170
(
2
):
233
40
.
[PubMed]
0166-4328
134.
Mehta
SH
,
Dhandapani
KM
,
De Sevilla
LM
,
Webb
RC
,
Mahesh
VB
,
Brann
DW
.
Tamoxifen, a selective estrogen receptor modulator, reduces ischemic damage caused by middle cerebral artery occlusion in the ovariectomized female rat
.
Neuroendocrinology
.
2003
Jan
;
77
(
1
):
44
50
.
[PubMed]
0028-3835
135.
Mickley
KR
,
Dluzen
DE
.
Dose-response effects of estrogen and tamoxifen upon methamphetamine-induced behavioral responses and neurotoxicity of the nigrostriatal dopaminergic system in female mice
.
Neuroendocrinology
.
2004
;
79
(
6
):
305
16
.
[PubMed]
0028-3835
136.
Rossberg
MI
,
Murphy
SJ
,
Traystman
RJ
,
Hurn
PD
.
LY353381.HCl, a selective estrogen receptor modulator, and experimental stroke
.
Stroke
.
2000
Dec
;
31
(
12
):
3041
6
.
[PubMed]
0039-2499
137.
Zhao
L
,
O’Neill
K
,
Brinton
RD
.
Estrogenic agonist activity of ICI 182,780 (Faslodex) in hippocampal neurons: implications for basic science understanding of estrogen signaling and development of estrogen modulators with a dual therapeutic profile
.
J Pharmacol Exp Ther
.
2006
Dec
;
319
(
3
):
1124
32
.
[PubMed]
0022-3565
138.
Green
PS
,
Simpkins
JW
.
Neuroprotective effects of estrogens: potential mechanisms of action
.
Int J Dev Neurosci
.
2000
Jul-Aug
;
18
(
4-5
):
347
58
.
[PubMed]
0736-5748
139.
Maximov
PY
,
Lee
TM
,
Jordan
VC
.
The discovery and development of selective estrogen receptor modulators (SERMs) for clinical practice
.
Curr Clin Pharmacol
.
2013
May
;
8
(
2
):
135
55
.
[PubMed]
1574-8847
140.
Brinton
RD
.
Selective estrogen receptor modulators (SERM) for the brain: recent advances and remaining challenges for developing a NeuroSERM™
.
Drug Dev Res
.
2002
;
56
(
3
):
380
92
. 0272-4391
141.
Brinton
RD
.
Requirements of a brain selective estrogen: advances and remaining challenges for developing a NeuroSERM
.
J Alzheimers Dis
.
2004
Dec
;
6
(
6
Suppl
):
S27
35
.
[PubMed]
1387-2877
142.
López
M
,
Tena-Sempere
M
.
Estrogens and the control of energy homeostasis: a brain perspective
.
Trends Endocrinol Metab
.
2015
Aug
;
26
(
8
):
411
21
.
[PubMed]
1043-2760
143.
Kim
JH
,
Cho
HT
,
Kim
YJ
.
The role of estrogen in adipose tissue metabolism: insights into glucose homeostasis regulation
.
Endocr J
.
2014
;
61
(
11
):
1055
67
.
[PubMed]
0918-8959
144.
Stanikova
D
,
Luck
T
,
Bae
YJ
,
Thiery
J
,
Ceglarek
U
,
Engel
C
, et al
Increased estrogen level can be associated with depression in males
.
Psychoneuroendocrinology
.
2018
Jan
;
87
:
196
203
.
[PubMed]
0306-4530
145.
Engler-chiurazzi
EB
, et al
Progress in Neurobiology Estrogens as neuroprotectants : estrogenic actions in the context of cognitive aging and brain injury
.
Prog Neurobiol
.
2016
.0301-0082
146.
Santolla
MF
,
Lappano
R
,
De Marco
P
,
Pupo
M
,
Vivacqua
A
,
Sisci
D
, et al
G protein-coupled estrogen receptor mediates the up-regulation of fatty acid synthase induced by 17β-estradiol in cancer cells and cancer-associated fibroblasts
.
J Biol Chem
.
2012
Dec
;
287
(
52
):
43234
45
.
[PubMed]
0021-9258
147.
Verderame
M
,
Scudiero
R
.
A comparative review on estrogen receptors in the reproductive male tract of non mammalian vertebrates
.
Steroids
.
2018
Jun
;
134
:
1
8
.
[PubMed]
0039-128X
148.
Dubal
DB
,
Zhu
H
,
Yu
J
,
Rau
SW
,
Shughrue
PJ
,
Merchenthaler
I
, et al
Estrogen receptor alpha, not beta, is a critical link in estradiol-mediated protection against brain injury
.
Proc Natl Acad Sci USA
.
2001
Feb
;
98
(
4
):
1952
7
.
[PubMed]
0027-8424
149.
Born
G
, et al
Estrogens and Antiestrogens I
.
Volume 135
.
Springer-Verlag
;
1999
.
150.
Blurton-Jones
M
,
Tuszynski
MH
.
Reactive astrocytes express estrogen receptors in the injured primate brain
.
J Comp Neurol
.
2001
Apr
;
433
(
1
):
115
23
.
[PubMed]
0021-9967
151.
García-Ovejero
D
,
Veiga
S
,
García-Segura
LM
,
Doncarlos
LL
.
Glial expression of estrogen and androgen receptors after rat brain injury
.
J Comp Neurol
.
2002
Aug
;
450
(
3
):
256
71
.
[PubMed]
0021-9967
152.
Sirotkin
AV
,
Harrath
AH
.
Phytoestrogens and their effects
.
Eur J Pharmacol
.
2014
Oct
;
741
:
230
6
.
[PubMed]
0014-2999
153.
Moreira
AC
,
Silva
AM
,
Santos
MS
,
Sardão
VA
.
Phytoestrogens as alternative hormone replacement therapy in menopause: what is real, what is unknown
.
J Steroid Biochem Mol Biol
.
2014
Sep
;
143
:
61
71
.
[PubMed]
0960-0760
154.
Cvejic
J
,
Bursac
M
,
Atanackovic
M
. Phytoestrogens: “Estrogene-Like” Phytochemicals.
Studies in Natural Products Chemsitry
.
Elsevier B.V.
;
2012
. pp.
1
35
.
155.
Persichini
T
,
Maio
N
,
di Patti
MC
,
Rizzo
G
,
Colasanti
M
,
Musci
G
.
Genistein up-regulates the iron efflux system in glial cells
.
Neurosci Lett
.
2010
Feb
;
470
(
2
):
145
9
.
[PubMed]
0304-3940
156.
Ma
WW
,
Hou
CC
,
Zhou
X
,
Yu
HL
,
Xi
YD
,
Ding
J
, et al
Genistein alleviates the mitochondria-targeted DNA damage induced by β-amyloid peptides 25-35 in C6 glioma cells
.
Neurochem Res
.
2013
Jul
;
38
(
7
):
1315
23
.
[PubMed]
0364-3190
157.
Lu
H
,
Shi
JX
,
Zhang
DM
,
Wang
HD
,
Hang
CH
,
Chen
HL
, et al
Inhibition of hemolysate-induced iNOS and COX-2 expression by genistein through suppression of NF-small ka, CyrillicB activation in primary astrocytes
.
J Neurol Sci
.
2009
Mar
;
278
(
1-2
):
91
5
.
[PubMed]
0022-510X
158.
Valles
SL
,
Dolz-Gaiton
P
,
Gambini
J
,
Borras
C
,
Lloret
A
,
Pallardo
FV
, et al
Estradiol or genistein prevent Alzheimer’s disease-associated inflammation correlating with an increase PPAR gamma expression in cultured astrocytes
.
Brain Res
.
2010
Feb
;
1312
:
138
44
.
[PubMed]
0006-8993
159.
Soltani
Z
,
Khaksari
M
,
Jafari
E
,
Iranpour
M
,
Shahrokhi
N
.
Is genistein neuroprotective in traumatic brain injury?
Physiol Behav
.
2015
Dec
;
152
Pt A
:
26
31
.
[PubMed]
0031-9384
160.
Sinha
K
,
Chaudhary
G
,
Gupta
YK
.
Protective effect of resveratrol against oxidative stress in middle cerebral artery occlusion model of stroke in rats
.
Life Sci
.
2002
Jun
;
71
(
6
):
655
65
.
[PubMed]
0024-3205
161.
Morissette
M
,
Al Sweidi
S
,
Callier
S
,
Di Paolo
T
.
Estrogen and SERM neuroprotection in animal models of Parkinson’s disease
.
Mol Cell Endocrinol
.
2008
Aug
;
290
(
1-2
):
60
9
.
[PubMed]
0303-7207
162.
Pinkerton
JV
,
Thomas
S
.
Use of SERMs for treatment in postmenopausal women
.
J Steroid Biochem Mol Biol
.
2014
Jul
;
142
:
142
54
.
[PubMed]
0960-0760
163.
Mirkin
S
,
Pickar
JH
.
Selective estrogen receptor modulators (SERMs): a review of clinical data
.
Maturitas
.
2015
Jan
;
80
(
1
):
52
7
.
[PubMed]
0378-5122
164.
Arevalo
MA
,
Diz-Chaves
Y
,
Santos-Galindo
M
,
Bellini
MJ
,
Garcia-Segura
LM
.
Selective oestrogen receptor modulators decrease the inflammatory response of glial cells
.
J Neuroendocrinol
.
2012
Jan
;
24
(
1
):
183
90
.
[PubMed]
0953-8194
165.
Bourque
M
,
Dluzen
DE
,
Di Paolo
T
.
Signaling pathways mediating the neuroprotective effects of sex steroids and SERMs in Parkinson’s disease
.
Front Neuroendocrinol
.
2012
Apr
;
33
(
2
):
169
78
.
[PubMed]
0091-3022
166.
Goldstein
JS
,
Sites
CK
.
Selective modulation of sex steroids
.
Ageing Res Rev
.
2002
Feb
;
1
(
1
):
17
28
.
[PubMed]
1568-1637
167.
Kloosterboer
HJ
.
Tibolone: a steroid with a tissue-specific mode of action
.
J Steroid Biochem Mol Biol
.
2001
Jan-Mar
;
76
(
1-5
):
231
8
.
[PubMed]
0960-0760
168.
Verheul
HA
,
Kloosterboer
HJ
.
Metabolism of exogenous sex steroids and effect on brain functions with a focus on tibolone
.
J Steroid Biochem Mol Biol
.
2006
Dec
;
102
(
1-5
):
195
204
.
[PubMed]
0960-0760
169.
Crespo-Castrillo
A
,
Yanguas-Casás
N
,
Arevalo
MA
,
Azcoitia
I
,
Barreto
GE
,
Garcia-Segura
LM
.
The Synthetic Steroid Tibolone Decreases Reactive Gliosis and Neuronal Death in the Cerebral Cortex of Female Mice After a Stab Wound Injury
.
Mol Neurobiol
.
2018
Nov
;
55
(
11
):
8651
67
.
[PubMed]
0893-7648
170.
Franco Rodríguez
NE
,
Dueñas Jiménez
JM
,
De la Torre Valdovinos
B
,
López Ruiz
JR
,
Hernández Hernández
L
,
Dueñas Jiménez
SH
.
Tamoxifen favoured the rat sensorial cortex regeneration after a penetrating brain injury
.
Brain Res Bull
.
2013
Sep
;
98
:
64
75
.
[PubMed]
0361-9230
171.
López Rodríguez
AB
,
Mateos Vicente
B
,
Romero-Zerbo
SY
,
Rodriguez-Rodriguez
N
,
Bellini
MJ
,
Rodriguez de Fonseca
F
, et al
Estradiol decreases cortical reactive astrogliosis after brain injury by a mechanism involving cannabinoid receptors
.
Cereb Cortex
.
2011
Sep
;
21
(
9
):
2046
55
.
[PubMed]
1047-3211
172.
Rzemieniec
J
,
Litwa
E
,
Wnuk
A
,
Lason
W
,
Gołas
A
,
Krzeptowski
W
, et al
Neuroprotective action of raloxifene against hypoxia-induced damage in mouse hippocampal cells depends on ERα but not ERβ or GPR30 signalling
.
J Steroid Biochem Mol Biol
.
2015
Feb
;
146
:
26
37
.
[PubMed]
0960-0760
173.
Rzemieniec
J
, et al
Anti-apoptotic caspase-3-independent actions
.
J Steroid Biochem Mol Biol
.
2014
;
146
:
26
37
.
[PubMed]
0960-0760
174.
Tsai
YT
,
Wang
CC
,
Leung
PO
,
Lin
KC
,
Chio
CC
,
Hu
CY
, et al
Extracellular signal-regulated kinase 1/2 is involved in a tamoxifen neuroprotective effect in a lateral fluid percussion injury rat model
.
J Surg Res
.
2014
Jun
;
189
(
1
):
106
16
.
[PubMed]
0022-4804
175.
Zhou
W
,
Koldzic-Zivanovic
N
,
Clarke
CH
,
de Beun
R
,
Wassermann
K
,
Bury
PS
, et al
Selective estrogen receptor modulator effects in the rat brain
.
Neuroendocrinology
.
2002
Jan
;
75
(
1
):
24
33
.
[PubMed]
0028-3835
176.
Dehghan
F
,
Khaksari
M
,
Abbasloo
E
,
Shahrokhi
N
.
The Effects of Estrogen Receptors’ Antagonist on Brain Edema, Intracranial Pressure and Neurological Outcomes after Traumatic Brain Injury in Rat
.
Iran Biomed J
.
2015
;
19
(
3
):
165
71
.
[PubMed]
2008-823X
177.
Littleton-Kearney
MT
,
Ostrowski
NL
,
Cox
DA
,
Rossberg
MI
,
Hurn
PD
.
Selective estrogen receptor modulators: tissue actions and potential for CNS protection
.
CNS Drug Rev
.
2002
;
8
(
3
):
309
30
.
[PubMed]
1080-563X
178.
Sönmez
U
,
Sönmez
A
,
Erbil
G
,
Tekmen
I
,
Baykara
B
.
Neuroprotective effects of resveratrol against traumatic brain injury in immature rats
.
Neurosci Lett
.
2007
Jun
;
420
(
2
):
133
7
.
[PubMed]
0304-3940
179.
Wang
YP
,
Lin
HP
,
Chen
HM
,
Kuo
YS
,
Lang
MJ
,
Sun
A
.
Hemoglobin, iron, and vitamin B12 deficiencies and high blood homocysteine levels in patients with anti-thyroid autoantibodies
.
J Formos Med Assoc
.
2014
Mar
;
113
(
3
):
155
60
.
[PubMed]
0929-6646
180.
Zhou
XM
,
Zhou
ML
,
Zhang
XS
,
Zhuang
Z
,
Li
T
,
Shi
JX
, et al
Resveratrol prevents neuronal apoptosis in an early brain injury model
.
J Surg Res
.
2014
Jun
;
189
(
1
):
159
65
.
[PubMed]
0022-4804
181.
Pawlak
J
,
Karolczak
M
,
Krust
A
,
Chambon
P
,
Beyer
C
.
Estrogen receptor-α is associated with the plasma membrane of astrocytes and coupled to the MAP/Src-kinase pathway
.
Glia
.
2005
May
;
50
(
3
):
270
5
.
[PubMed]
0894-1491
182.
Zhang
L
,
Li
B
,
Zhao
W
,
Chang
YH
,
Ma
W
,
Dragan
M
, et al
Sex-related differences in MAPKs activation in rat astrocytes: effects of estrogen on cell death
.
Brain Res Mol Brain Res
.
2002
Jun
;
103
(
1-2
):
1
11
.
[PubMed]
0169-328X
183.
Buchanan
CD
,
Mahesh
VB
,
Brann
DW
.
Estrogen-astrocyte-luteinizing hormone-releasing hormone signaling: a role for transforming growth factor-beta(1)
.
Biol Reprod
.
2000
Jun
;
62
(
6
):
1710
21
.
[PubMed]
0006-3363
184.
Hösli
E
,
Rühl
W
,
Hösli
L
.
Histochemical and electrophysiological evidence for estrogen receptors on cultured astrocytes: colocalization with cholinergic receptors
.
Int J Dev Neurosci
.
2000
Feb
;
18
(
1
):
101
11
.
[PubMed]
0736-5748
185.
Lee
D
,
Smallbone
K
,
Dunn
WB
,
Murabito
E
,
Winder
CL
,
Kell
DB
, et al
Improving metabolic flux predictions using absolute gene expression data
.
BMC Syst Biol
.
2012
Jun
;
6
(
1
):
73
73
.
[PubMed]
1752-0509
186.
Marino
M
,
Galluzzo
P
,
Ascenzi
P
.
Estrogen signaling multiple pathways to impact gene transcription
.
Curr Genomics
.
2006
;
7
(
8
):
497
508
.
[PubMed]
1389-2029
187.
Paterni
I
,
Granchi
C
,
Katzenellenbogen
JA
,
Minutolo
F
.
Estrogen receptors alpha (ERα) and beta (ERβ): subtype-selective ligands and clinical potential
.
Steroids
.
2014
Nov
;
90
:
13
29
.
[PubMed]
0039-128X
188.
Santagati
S
,
Melcangi
RC
,
Celotti
F
,
Martini
L
,
Maggi
A
.
Estrogen receptor is expressed in different types of glial cells in culture
.
J Neurochem
.
1994
Dec
;
63
(
6
):
2058
64
.
[PubMed]
0022-3042
189.
Arimoto
JM
,
Wong
A
,
Rozovsky
I
,
Lin
SW
,
Morgan
TE
,
Finch
CE
.
Age increase of estrogen receptor-α (ERα) in cortical astrocytes impairs neurotrophic support in male and female rats
.
Endocrinology
.
2013
Jun
;
154
(
6
):
2101
13
.
[PubMed]
0013-7227
190.
Azcoitia
I
,
Sierra
A
,
Garcia-Segura
LM
.
Localization of estrogen receptor beta-immunoreactivity in astrocytes of the adult rat brain
.
Glia
.
1999
May
;
26
(
3
):
260
7
.
[PubMed]
0894-1491
191.
Langub
MC
 Jr
,
Watson
RE
 Jr
.
Estrogen receptor-immunoreactive glia, endothelia, and ependyma in guinea pig preoptic area and median eminence: electron microscopy
.
Endocrinology
.
1992
Jan
;
130
(
1
):
364
72
.
[PubMed]
0013-7227
192.
Donahue
JE
,
Stopa
EG
,
Chorsky
RL
,
King
JC
,
Schipper
HM
,
Tobet
SA
, et al
Cells containing immunoreactive estrogen receptor-alpha in the human basal forebrain
.
Brain Res
.
2000
Feb
;
856
(
1-2
):
142
51
.
[PubMed]
0006-8993
193.
Jakab
RL
,
Wong
JK
,
Belcher
SM
.
Estrogen receptor beta immunoreactivity in differentiating cells of the developing rat cerebellum
.
J Comp Neurol
.
2001
Feb
;
430
(
3
):
396
409
.
[PubMed]
0021-9967
194.
Milner
TA
,
McEwen
BS
,
Hayashi
S
,
Li
CJ
,
Reagan
LP
,
Alves
SE
.
Ultrastructural evidence that hippocampal alpha estrogen receptors are located at extranuclear sites
.
J Comp Neurol
.
2001
Jan
;
429
(
3
):
355
71
.
[PubMed]
0021-9967
195.
Savaskan
E
,
Olivieri
G
,
Meier
F
,
Ravid
R
,
Müller-Spahn
F
.
Hippocampal estrogen β-receptor immunoreactivity is increased in Alzheimer’s disease
.
Brain Res
.
2001
Jul
;
908
(
2
):
113
9
.
[PubMed]
0006-8993
196.
Karki
P
,
Smith
K
,
Johnson
J
 Jr
,
Lee
E
.
Astrocyte-derived growth factors and estrogen neuroprotection: role of transforming growth factor-α in estrogen-induced upregulation of glutamate transporters in astrocytes
.
Mol Cell Endocrinol
.
2014
May
;
389
(
1-2
):
58
64
.
[PubMed]
0303-7207
197.
Karki
P
,
Webb
A
,
Zerguine
A
,
Choi
J
,
Son
DS
,
Lee
E
.
Mechanism of raloxifene-induced upregulation of glutamate transporters in rat primary astrocytes
.
Glia
.
2014
Aug
;
62
(
8
):
1270
83
.
[PubMed]
0894-1491
198.
Spence
RD
,
Wisdom
AJ
,
Cao
Y
,
Hill
HM
,
Mongerson
CR
,
Stapornkul
B
, et al
Estrogen mediates neuroprotection and anti-inflammatory effects during EAE through ERα signaling on astrocytes but not through ERβ signaling on astrocytes or neurons
.
J Neurosci
.
2013
Jun
;
33
(
26
):
10924
33
.
[PubMed]
0270-6474
199.
Garcia-Estrada
J
,
Del Rio
JA
,
Luquin
S
,
Soriano
E
,
Garcia-Segura
LM
.
Gonadal hormones down-regulate reactive gliosis and astrocyte proliferation after a penetrating brain injury
.
Brain Res
.
1993
Nov
;
628
(
1-2
):
271
8
.
[PubMed]
0006-8993
200.
García-Estrada
J
,
Luquín
S
,
Fernández
AM
,
Garcia-Segura
LM
.
Dehydroepiandrosterone, pregnenolone and sex steroids down-regulate reactive astroglia in the male rat brain after a penetrating brain injury
.
Int J Dev Neurosci
.
1999
Apr
;
17
(
2
):
145
51
.
[PubMed]
0736-5748
201.
Bagheri
M
,
Roghani
M
,
Joghataei
MT
,
Mohseni
S
.
Genistein inhibits aggregation of exogenous amyloid-beta₁₋₄₀ and alleviates astrogliosis in the hippocampus of rats
.
Brain Res
.
2012
Jan
;
1429
:
145
54
.
[PubMed]
0006-8993
202.
Colón
JM
,
González
PA
,
Cajigas
Á
,
Maldonado
WI
,
Torrado
AI
,
Santiago
JM
, et al
Continuous tamoxifen delivery improves locomotor recovery 6h after spinal cord injury by neuronal and glial mechanisms in male rats
.
Exp Neurol
.
2018
Jan
;
299
Pt A
:
109
21
.
[PubMed]
0014-4886
203.
Danbolt
NC
.
Glutamate uptake
.
Prog Neurobiol
.
2001
Sep
;
65
(
1
):
1
105
.
[PubMed]
0301-0082
204.
Yuzefovych
LV
,
Musiyenko
SI
,
Wilson
GL
,
Rachek
LI
.
Mitochondrial DNA damage and dysfunction, and oxidative stress are associated with endoplasmic reticulum stress, protein degradation and apoptosis in high fat diet-induced insulin resistance mice
.
PLoS One
.
2013
;
8
(
1
):
e54059
.
[PubMed]
1932-6203
205.
Azcoitia
I
,
Sierra
A
,
Garcia-Segura
LM
.
Estradiol prevents kainic acid-induced neuronal loss in the rat dentate gyrus
.
Neuroreport
.
1998
Sep
;
9
(
13
):
3075
9
.
[PubMed]
0959-4965
206.
Azcoitia
I
,
Sierra
A
,
Garcia-Segura
LM
.
Neuroprotective effects of estradiol in the adult rat hippocampus: interaction with insulin-like growth factor-I signalling
.
J Neurosci Res
.
1999
Dec
;
58
(
6
):
815
22
.
[PubMed]
0360-4012
207.
Azcoitia
I
,
Fernandez-Galaz
C
,
Sierra
A
,
Garcia-Segura
LM
.
Gonadal hormones affect neuronal vulnerability to excitotoxin-induced degeneration
.
J Neurocytol
.
1999
Sep
;
28
(
9
):
699
710
.
[PubMed]
0300-4864
208.
Picazo
O
,
Azcoitia
I
,
Garcia-Segura
LM
.
Neuroprotective and neurotoxic effects of estrogens
.
Brain Res
.
2003
Nov
;
990
(
1-2
):
20
7
.
[PubMed]
0006-8993
209.
Picazo
O
,
Becerril-Montes
A
,
Huidobro-Perez
D
,
Garcia-Segura
LM
.
Neuroprotective actions of the synthetic estrogen 17alpha-ethynylestradiol in the hippocampus
.
Cell Mol Neurobiol
.
2010
Jul
;
30
(
5
):
675
82
.
[PubMed]
0272-4340
210.
Shy
H
,
Malaiyandi
L
,
Timiras
PS
.
Protective action of 17beta-estradiol and tamoxifen on glutamate toxicity in glial cells
.
Int J Dev Neurosci
.
2000
Apr-Jun
;
18
(
2-3
):
289
97
.
[PubMed]
0736-5748
211.
dos Santos
AQ
,
Nardin
P
,
Funchal
C
,
de Almeida
LM
,
Jacques-Silva
MC
,
Wofchuk
ST
, et al
Resveratrol increases glutamate uptake and glutamine synthetase activity in C6 glioma cells
.
Arch Biochem Biophys
.
2006
Sep
;
453
(
2
):
161
7
.
[PubMed]
0003-9861
212.
de Almeida
LM
,
Piñeiro
CC
,
Leite
MC
,
Brolese
G
,
Tramontina
F
,
Feoli
AM
, et al
Resveratrol increases glutamate uptake, glutathione content, and S100B secretion in cortical astrocyte cultures
.
Cell Mol Neurobiol
.
2007
Aug
;
27
(
5
):
661
8
.
[PubMed]
0272-4340
213.
Moreira
PI
,
Custódio
JB
,
Oliveira
CR
,
Santos
MS
.
Hydroxytamoxifen protects against oxidative stress in brain mitochondria
.
Biochem Pharmacol
.
2004
Jul
;
68
(
1
):
195
204
.
[PubMed]
0006-2952
214.
Moreira
PI
,
Custódio
JB
,
Oliveira
CR
,
Santos
MS
.
Brain mitochondrial injury induced by oxidative stress-related events is prevented by tamoxifen
.
Neuropharmacology
.
2005
Mar
;
48
(
3
):
435
47
.
[PubMed]
0028-3908
215.
Thiede
A
,
Gellerich
FN
,
Schönfeld
P
,
Siemen
D
.
Complex effects of 17β-estradiol on mitochondrial function
.
Biochim Biophys Acta
.
2012
Oct
;
1817
(
10
):
1747
53
.
[PubMed]
0006-3002
216.
Guo
J
,
Duckles
SP
,
Weiss
JH
,
Li
X
,
Krause
DN
.
17β-Estradiol prevents cell death and mitochondrial dysfunction by an estrogen receptor-dependent mechanism in astrocytes after oxygen-glucose deprivation/reperfusion
.
Free Radic Biol Med
.
2012
Jun
;
52
(
11-12
):
2151
60
.
[PubMed]
0891-5849
217.
Ates
O
,
Cayli
S
,
Altinoz
E
,
Gurses
I
,
Yucel
N
,
Sener
M
, et al
Neuroprotection by resveratrol against traumatic brain injury in rats
.
Mol Cell Biochem
.
2007
Jan
;
294
(
1-2
):
137
44
.
[PubMed]
0300-8177
218.
Klinge
CM
.
Estrogens regulate life and death in mitochondria
.
J Bioenerg Biomembr
.
2017
Aug
;
49
(
4
):
307
24
.
[PubMed]
0145-479X
219.
Simpkins
JW
,
Yang
SH
,
Sarkar
SN
,
Pearce
V
.
Estrogen actions on mitochondria—physiological and pathological implications
.
Mol Cell Endocrinol
.
2008
Aug
;
290
(
1-2
):
51
9
.
[PubMed]
0303-7207
220.
Simpkins
JW
,
Dykens
JA
.
Mitochondrial mechanisms of estrogen neuroprotection
.
Brain Res Brain Res Rev
.
2008
Mar
;
57
(
2
):
421
30
.
[PubMed]
0165-0173
221.
Kubik
LL
,
Philbert
MA
.
The role of astrocyte mitochondria in differential regional susceptibility to environmental neurotoxicants: tools for understanding neurodegeneration
.
Toxicol Sci
.
2015
Mar
;
144
(
1
):
7
16
.
[PubMed]
1096-6080
222.
Moreira
PI
,
Custódio
JB
,
Nunes
E
,
Oliveira
PJ
,
Moreno
A
,
Seiça
R
, et al
Mitochondria from distinct tissues are differently affected by 17β-estradiol and tamoxifen
.
J Steroid Biochem Mol Biol
.
2011
Jan
;
123
(
1-2
):
8
16
.
[PubMed]
0960-0760
223.
Yang
SH
,
Liu
R
,
Perez
EJ
,
Wen
Y
,
Stevens
SM
 Jr
,
Valencia
T
, et al
Mitochondrial localization of estrogen receptor beta
.
Proc Natl Acad Sci USA
.
2004
Mar
;
101
(
12
):
4130
5
.
[PubMed]
0027-8424
224.
Aoki
E
,
Yano
R
,
Yokoyama
H
,
Kato
H
,
Araki
T
.
Role of nuclear transcription factor kappa B (NF-kappaB) for MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahyropyridine)-induced apoptosis in nigral neurons of mice
.
Exp Mol Pathol
.
2009
Feb
;
86
(
1
):
57
64
.
[PubMed]
0014-4800
225.
Cerciat
M
,
Unkila
M
,
Garcia-Segura
LM
,
Arevalo
MA
.
Selective estrogen receptor modulators decrease the production of interleukin-6 and interferon-γ-inducible protein-10 by astrocytes exposed to inflammatory challenge in vitro
.
Glia
.
2010
Jan
;
58
(
1
):
93
102
.
[PubMed]
0894-1491
226.
Khaksari
M
,
Soltani
Z
,
Shahrokhi
N
,
Moshtaghi
G
,
Asadikaram
G
.
The role of estrogen and progesterone, administered alone and in combination, in modulating cytokine concentration following traumatic brain injury
.
Can J Physiol Pharmacol
.
2011
Jan
;
89
(
1
):
31
40
.
[PubMed]
0008-4212
227.
Liu
MH
,
Lin
YS
,
Sheu
SY
,
Sun
JS
.
Anti-inflammatory effects of daidzein on primary astroglial cell culture
.
Nutr Neurosci
.
2009
Jun
;
12
(
3
):
123
34
.
[PubMed]
1028-415X
228.
Sarkaki
AR
,
Khaksari Haddad
M
,
Soltani
Z
,
Shahrokhi
N
,
Mahmoodi
M
.
Time- and dose-dependent neuroprotective effects of sex steroid hormones on inflammatory cytokines after a traumatic brain injury
.
J Neurotrauma
.
2013
Jan
;
30
(
1
):
47
54
.
[PubMed]
0897-7151
229.
Zonta
M
,
Angulo
MC
,
Gobbo
S
,
Rosengarten
B
,
Hossmann
KA
,
Pozzan
T
, et al
Neuron-to-astrocyte signaling is central to the dynamic control of brain microcirculation
.
Nat Neurosci
.
2003
Jan
;
6
(
1
):
43
50
.
[PubMed]
1097-6256
230.
Levi
G
,
Wilkin
GP
,
Ciotti
MT
,
Johnstone
S
.
Enrichment of differentiated, stellate astrocytes in cerebellar interneuron cultures as studied by GFAP immunofluorescence and autoradiographic uptake patterns with [3H]D-aspartate and [3H]GABA
.
Brain Res
.
1983
Nov
;
312
(
2
):
227
41
.
[PubMed]
0006-8993
231.
Newman
DJ
,
Cragg
GM
.
Natural products as sources of new drugs over the last 25 years
.
J Nat Prod
.
2007
Mar
;
70
(
3
):
461
77
.
[PubMed]
0163-3864
232.
Aschner
M
.
Neuron-astrocyte interactions: implications for cellular energetics and antioxidant levels
.
Neurotoxicology
.
2000
Dec
;
21
(
6
):
1101
7
.
[PubMed]
0161-813X
233.
Nedergaard
M
,
Ransom
B
,
Goldman
SA
.
New roles for astrocytes: redefining the functional architecture of the brain
.
Trends Neurosci
.
2003
Oct
;
26
(
10
):
523
30
.
[PubMed]
0166-2236
234.
Myer
DJ
,
Gurkoff
GG
,
Lee
SM
,
Hovda
DA
,
Sofroniew
MV
.
Essential protective roles of reactive astrocytes in traumatic brain injury
.
Brain
.
2006
Oct
;
129
(
Pt 10
):
2761
72
.
[PubMed]
0006-8950
235.
Gee
JR
,
Keller
JN
.
Astrocytes: regulation of brain homeostasis via apolipoprotein E
.
Int J Biochem Cell Biol
.
2005
Jun
;
37
(
6
):
1145
50
.
[PubMed]
1357-2725
236.
Di Giorgio
FP
,
Carrasco
MA
,
Siao
MC
,
Maniatis
T
,
Eggan
K
.
Non-cell autonomous effect of glia on motor neurons in an embryonic stem cell-based ALS model
.
Nat Neurosci
.
2007
May
;
10
(
5
):
608
14
.
[PubMed]
1097-6256
237.
John
GR
,
Lee
SC
,
Brosnan
CF
.
Cytokines: powerful regulators of glial cell activation
.
Neuroscientist
.
2003
Feb
;
9
(
1
):
10
22
.
[PubMed]
1073-8584
238.
Smith
GM
,
Strunz
C
.
Growth factor and cytokine regulation of chondroitin sulfate proteoglycans by astrocytes
.
Glia
.
2005
Nov
;
52
(
3
):
209
18
.
[PubMed]
0894-1491
239.
Streit
WJ
,
Hurley
SD
,
McGraw
TS
,
Semple-Rowland
SL
.
Comparative evaluation of cytokine profiles and reactive gliosis supports a critical role for interleukin-6 in neuron-glia signaling during regeneration
.
J Neurosci Res
.
2000
Jul
;
61
(
1
):
10
20
.
[PubMed]
0360-4012
240.
Farina
C
,
Aloisi
F
,
Meinl
E
.
Astrocytes are active players in cerebral innate immunity
.
Trends Immunol
.
2007
Mar
;
28
(
3
):
138
45
.
[PubMed]
1471-4906
241.
Hamby
ME
,
Coppola
G
,
Ao
Y
,
Geschwind
DH
,
Khakh
BS
,
Sofroniew
MV
.
Inflammatory mediators alter the astrocyte transcriptome and calcium signaling elicited by multiple G-protein-coupled receptors
.
J Neurosci
.
2012
Oct
;
32
(
42
):
14489
510
.
[PubMed]
0270-6474
242.
Bekar
LK
,
He
W
,
Nedergaard
M
.
Locus coeruleus alpha-adrenergic-mediated activation of cortical astrocytes in vivo
.
Cereb Cortex
.
2008
Dec
;
18
(
12
):
2789
95
.
[PubMed]
1047-3211
243.
Braissant
O
,
McLin
VA
,
Cudalbu
C
.
Ammonia toxicity to the brain
.
J Inherit Metab Dis
.
2013
Jul
;
36
(
4
):
595
612
.
[PubMed]
0141-8955
244.
Chandra
K
, et al
Protection against FCA induced oxidative stress induced DNA damage as a model of arthritis and In vitro anti-arthritic potential of Costus speciosus Rhizome extract
.
International Journal of Pharmacognosy and Phytochemical Research
.
2015
;
7
(
2
):
383
9
.
245.
Lee
HJ
,
Suk
JE
,
Patrick
C
,
Bae
EJ
,
Cho
JH
,
Rho
S
, et al
Direct transfer of α-synuclein from neuron to astroglia causes inflammatory responses in synucleinopathies
.
J Biol Chem
.
2010
Mar
;
285
(
12
):
9262
72
.
[PubMed]
0021-9258
246.
Simpson
JE
,
Ince
PG
,
Lace
G
,
Forster
G
,
Shaw
PJ
,
Matthews
F
, et al;
MRC Cognitive Function and Ageing Neuropathology Study Group
.
Astrocyte phenotype in relation to Alzheimer-type pathology in the ageing brain
.
Neurobiol Aging
.
2010
Apr
;
31
(
4
):
578
90
.
[PubMed]
0197-4580
247.
Kang
W
,
Hébert
JM
.
Signaling pathways in reactive astrocytes, a genetic perspective
.
Mol Neurobiol
.
2011
Jun
;
43
(
3
):
147
54
.
[PubMed]
0893-7648
248.
Chen
Y
,
Miles
DK
,
Hoang
T
,
Shi
J
,
Hurlock
E
,
Kernie
SG
, et al
The basic helix-loop-helix transcription factor olig2 is critical for reactive astrocyte proliferation after cortical injury
.
J Neurosci
.
2008
Oct
;
28
(
43
):
10983
9
.
[PubMed]
0270-6474
249.
Beck
H
,
Semisch
M
,
Culmsee
C
,
Plesnila
N
,
Hatzopoulos
AK
.
Egr-1 regulates expression of the glial scar component phosphacan in astrocytes after experimental stroke
.
Am J Pathol
.
2008
Jul
;
173
(
1
):
77
92
.
[PubMed]
0002-9440
250.
Bundesen
LQ
,
Scheel
TA
,
Bregman
BS
,
Kromer
LF
.
Ephrin-B2 and EphB2 regulation of astrocyte-meningeal fibroblast interactions in response to spinal cord lesions in adult rats
.
J Neurosci
.
2003
Aug
;
23
(
21
):
7789
800
.
[PubMed]
0270-6474
251.
Brambilla
R
,
Hurtado
A
,
Persaud
T
,
Esham
K
,
Pearse
DD
,
Oudega
M
, et al
Transgenic inhibition of astroglial NF-κ B leads to increased axonal sparing and sprouting following spinal cord injury
.
J Neurochem
.
2009
Jul
;
110
(
2
):
765
78
.
[PubMed]
0022-3042
252.
Panenka
W
,
Jijon
H
,
Herx
LM
,
Armstrong
JN
,
Feighan
D
,
Wei
T
, et al
P2X7-like receptor activation in astrocytes increases chemokine monocyte chemoattractant protein-1 expression via mitogen-activated protein kinase
.
J Neurosci
.
2001
Sep
;
21
(
18
):
7135
42
.
[PubMed]
0270-6474
253.
Qin
H
,
Niyongere
SA
,
Lee
SJ
,
Baker
BJ
,
Benveniste
EN
.
Expression and functional significance of SOCS-1 and SOCS-3 in astrocytes
.
J Immunol
.
2008
Sep
;
181
(
5
):
3167
76
.
[PubMed]
0022-1767
254.
Shih
AY
,
Johnson
DA
,
Wong
G
,
Kraft
AD
,
Jiang
L
,
Erb
H
, et al
Coordinate regulation of glutathione biosynthesis and release by Nrf2-expressing glia potently protects neurons from oxidative stress
.
J Neurosci
.
2003
Apr
;
23
(
8
):
3394
406
.
[PubMed]
0270-6474
255.
Vargas
MR
,
Johnson
DA
,
Sirkis
DW
,
Messing
A
,
Johnson
JA
.
Nrf2 activation in astrocytes protects against neurodegeneration in mouse models of familial amyotrophic lateral sclerosis
.
J Neurosci
.
2008
Dec
;
28
(
50
):
13574
81
.
[PubMed]
0270-6474
256.
Hamby
ME
,
Hewett
JA
,
Hewett
SJ
.
TGF-beta1 potentiates astrocytic nitric oxide production by expanding the population of astrocytes that express NOS-2
.
Glia
.
2006
Nov
;
54
(
6
):
566
77
.
[PubMed]
0894-1491
257.
Figiel
M
,
Maucher
T
,
Rozyczka
J
,
Bayatti
N
,
Engele
J
.
Regulation of glial glutamate transporter expression by growth factors
.
Exp Neurol
.
2003
Sep
;
183
(
1
):
124
35
.
[PubMed]
0014-4886
258.
Li
LB
,
Toan
SV
,
Zelenaia
O
,
Watson
DJ
,
Wolfe
JH
,
Rothstein
JD
, et al
Regulation of astrocytic glutamate transporter expression by Akt: evidence for a selective transcriptional effect on the GLT-1/EAAT2 subtype
.
J Neurochem
.
2006
May
;
97
(
3
):
759
71
.
[PubMed]
0022-3042
259.
Khaksari
M
,
Hajializadeh
Z
,
Shahrokhi
N
,
Esmaeili-Mahani
S
.
Changes in the gene expression of estrogen receptors involved in the protective effect of estrogen in rat’s trumatic brain injury
.
Brain Res
.
2015
Aug
;
1618
:
1
8
.
[PubMed]
0006-8993
260.
Day
NL
,
Floyd
CL
,
D’Alessandro
TL
,
Hubbard
WJ
,
Chaudry
IH
.
17β-estradiol confers protection after traumatic brain injury in the rat and involves activation of G protein-coupled estrogen receptor 1
.
J Neurotrauma
.
2013
Sep
;
30
(
17
):
1531
41
.
[PubMed]
0897-7151
261.
Zhang
YQ
,
Shi
J
,
Rajakumar
G
,
Day
AL
,
Simpkins
JW
.
Effects of gender and estradiol treatment on focal brain ischemia
.
Brain Res
.
1998
Feb
;
784
(
1-2
):
321
4
.
[PubMed]
0006-8993
262.
Espinosa-Raya
J
,
Neri-Gómez
T
,
Orozco-Suárez
S
,
Campos
MG
,
Guerra-Araiza
C
.
Chronic administration of tibolone modulates anxiety-like behavior and enhances cognitive performance in ovariectomized rats
.
Horm Behav
.
2012
Jan
;
61
(
1
):
76
83
.
[PubMed]
0018-506X
263.
Farfán-García
ED
,
Castillo-Hernández
MC
,
Pinto-Almazán
R
,
Rivas-Arancibia
S
,
Gallardo
JM
,
Guerra-Araiza
C
.
Tibolone prevents oxidation and ameliorates cholinergic deficit induced by ozone exposure in the male rat hippocampus
.
Neurochem Res
.
2014
Sep
;
39
(
9
):
1776
86
.
[PubMed]
0364-3190
264.
Pinto-Almazán
R
,
Rivas-Arancibia
S
,
Farfán-García
ED
,
Rodríguez-Martínez
E
,
Guerra-Araiza
C
.
Neuroprotective effects of tibolone against oxidative stress induced by ozone exposure
.
Rev Neurol
.
2014
May
;
58
(
10
):
441
8
.
[PubMed]
1576-6578
265.
Fluck
E
,
File
SE
,
Rymer
J
.
Cognitive effects of 10 years of hormone-replacement therapy with tibolone
.
J Clin Psychopharmacol
.
2002
Feb
;
22
(
1
):
62
7
.
[PubMed]
0271-0749
266.
Sato
K
,
Saito
Y
,
Oka
J
,
Ohwada
T
,
Nakazawa
K
.
Effects of tamoxifen on L-glutamate transporters of astrocytes
.
J Pharmacol Sci
.
2008
Jun
;
107
(
2
):
226
30
.
[PubMed]
1347-8613
267.
Karki
P
,
Webb
A
,
Smith
K
,
Lee
K
,
Son
DS
,
Aschner
M
, et al
cAMP response element-binding protein (CREB) and nuclear factor κB mediate the tamoxifen-induced up-regulation of glutamate transporter 1 (GLT-1) in rat astrocytes
.
J Biol Chem
.
2013
Oct
;
288
(
40
):
28975
86
.
[PubMed]
0021-9258
268.
Hung
KL
,
Wang
CC
,
Wang
SJ
.
Cellular mechanisms of acute decrease of glutamate release induced by raloxifene in rat cerebral cortex
.
Neuropharmacology
.
2011
Jul-Aug
;
61
(
1-2
):
293
304
.
[PubMed]
0028-3908
269.
Ismailoğlu
Ö
,
Oral
B
,
Sütcü
R
,
Kara
Y
,
Tomruk
O
,
Demir
N
.
Neuroprotective effects of raloxifene on experimental spinal cord injury in rats
.
Am J Med Sci
.
2013
Jan
;
345
(
1
):
39
44
.
[PubMed]
0002-9629
270.
Mosquera
L
,
Colón
JM
,
Santiago
JM
,
Torrado
AI
,
Meléndez
M
,
Segarra
AC
, et al
Tamoxifen and estradiol improved locomotor function and increased spared tissue in rats after spinal cord injury: their antioxidant effect and role of estrogen receptor alpha
.
Brain Res
.
2014
May
;
1561
:
11
22
.
[PubMed]
0006-8993
271.
Tian
DS
,
Liu
JL
,
Xie
MJ
,
Zhan
Y
,
Qu
WS
,
Yu
ZY
, et al
Tamoxifen attenuates inflammatory-mediated damage and improves functional outcome after spinal cord injury in rats
.
J Neurochem
.
2009
Jun
;
109
(
6
):
1658
67
.
[PubMed]
0022-3042
272.
Shi
R
,
Wang
S
,
Qi
X
,
Chen
S
,
Chen
P
,
Zhang
Q
.
Lose dose genistein inhibits glucocorticoid receptor and ischemic brain injury in female rats
.
Neurochem Int
.
2014
Jan
;
65
(
1
):
14
22
.
[PubMed]
0197-0186
273.
Liu
R
,
Zhong
X
,
Zeng
J
,
Huang
Z
,
Li
X
,
Xiao
H
, et al
3′-Daidzein sulfonate sodium inhibits neuronal apoptosis induced by cerebral ischemia-reperfusion
.
Int J Mol Med
.
2017
Apr
;
39
(
4
):
1021
8
.
[PubMed]
1107-3756
Copyright / Drug Dosage / Disclaimer
Copyright: All rights reserved. No part of this publication may be translated into other languages, reproduced or utilized in any form or by any means, electronic or mechanical, including photocopying, recording, microcopying, or by any information storage and retrieval system, without permission in writing from the publisher.
Drug Dosage: The authors and the publisher have exerted every effort to ensure that drug selection and dosage set forth in this text are in accord with current recommendations and practice at the time of publication. However, in view of ongoing research, changes in government regulations, and the constant flow of information relating to drug therapy and drug reactions, the reader is urged to check the package insert for each drug for any changes in indications and dosage and for added warnings and precautions. This is particularly important when the recommended agent is a new and/or infrequently employed drug.
Disclaimer: The statements, opinions and data contained in this publication are solely those of the individual authors and contributors and not of the publishers and the editor(s). The appearance of advertisements or/and product references in the publication is not a warranty, endorsement, or approval of the products or services advertised or of their effectiveness, quality or safety. The publisher and the editor(s) disclaim responsibility for any injury to persons or property resulting from any ideas, methods, instructions or products referred to in the content or advertisements.