Next Article in Journal
Adeno-Associated Viruses for Modeling Neurological Diseases in Animals: Achievements and Prospects
Next Article in Special Issue
TGF-β as a Key Modulator of Astrocyte Reactivity: Disease Relevance and Therapeutic Implications
Previous Article in Journal
Seizure Susceptibility and Sleep Disturbance as Biomarkers of Epileptogenesis after Experimental TBI
Previous Article in Special Issue
Alcohol-Induced Alterations in the Vascular Basement Membrane in the Substantia Nigra of the Adult Human Brain
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Brain Trauma, Glucocorticoids and Neuroinflammation: Dangerous Liaisons for the Hippocampus

by
Ilia G. Komoltsev
1,2 and
Natalia V. Gulyaeva
1,2,*
1
Department of Functional Biochemistry of the Nervous System, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, 117465 Moscow, Russia
2
Moscow Research and Clinical Center for Neuropsychiatry, 115419 Moscow, Russia
*
Author to whom correspondence should be addressed.
Submission received: 6 April 2022 / Revised: 30 April 2022 / Accepted: 13 May 2022 / Published: 15 May 2022

Abstract

:
Glucocorticoid-dependent mechanisms of inflammation-mediated distant hippocampal damage are discussed with a focus on the consequences of traumatic brain injury. The effects of glucocorticoids on specific neuronal populations in the hippocampus depend on their concentration, duration of exposure and cell type. Previous stress and elevated level of glucocorticoids prior to pro-inflammatory impact, as well as long-term though moderate elevation of glucocorticoids, may inflate pro-inflammatory effects. Glucocorticoid-mediated long-lasting neuronal circuit changes in the hippocampus after brain trauma are involved in late post-traumatic pathology development, such as epilepsy, depression and cognitive impairment. Complex and diverse actions of the hypothalamic–pituitary–adrenal axis on neuroinflammation may be essential for late post-traumatic pathology. These mechanisms are applicable to remote hippocampal damage occurring after other types of focal brain damage (stroke, epilepsy) or central nervous system diseases without obvious focal injury. Thus, the liaisons of excessive glucocorticoids/dysfunctional hypothalamic–pituitary–adrenal axis with neuroinflammation, dangerous to the hippocampus, may be crucial to distant hippocampal damage in many brain diseases. Taking into account that the hippocampus controls both the cognitive functions and the emotional state, further research on potential links between glucocorticoid signaling and inflammatory processes in the brain and respective mechanisms is vital.

1. Introduction

Brain injury is a common cause of death and disability for people of all ages worldwide [1,2,3]. Depending on the biomechanics, brain lesions may occur both in areas of the brain directly adjacent to the place of force application and in remote areas [4]. The mechanisms of hippocampal damage are of particular importance, since they underlie late complications of traumatic brain injury (TBI), such as epilepsy, depression and cognitive impairment. The mechanisms of reorganization of neuronal networks in the hippocampus include long-lasting chronic neuroinflammation and secondary damage to the nervous tissue [5]. Responses and disturbances of the hypothalamic–pituitary–adrenal (HPA) axis may play a critical role in late post-traumatic pathology, in particular by modulation of synaptic activity and neuroinflammation in the hippocampus.
Even though stress-induced neuroinflammation and neurodegeneration in the hippocampus is fairly well described, and secondary hippocampal damage after TBI is studied by several groups, so far there have been no reviews on the glucocorticoid-dependent mechanisms of inflammation-mediated distant hippocampal damage with a focus on the consequences of TBI. PubMed search for the combination of words “TBI” (or “traumatic brain injury” or “brain trauma”) and “corticosterone” shows no review papers. A search query for “TBI” and “glucocorticoids” results in 15 relevant reviews, though none deals with the hippocampus. Ten of them are focused precisely on the clinical aspects of TBI: four reviews are related to pituitary disfunction after TBI [6,7,8,9]; three reviews are devoted to treatment [10,11,12], including usage of glucocorticoids (GCs) as medication; two reviews are focused on post-traumatic stress disorder [13,14]. Only five reviews include experimental studies; three of them are focused on the mechanisms of progesterone, dexanabinol and dexamethasone effects [15], TNF inhibitors [15] and mesenchymal stem cells in TBI [16]. One review describes the blood–brain barrier disfunction and the effects of GCs on its permeability [16]. The last and most relevant review was published in 2019 and was devoted to pro- and anti-inflammatory action of GCs after TBI [17]. Nevertheless, it lacks several important aspects of GC action on synaptic transmission and excitotoxicity in the hippocampus.
The aim of the present review was an analysis of the glucocorticoid-dependent mechanisms of inflammation-mediated distant hippocampal damage with a focus on the consequences of traumatic brain injury. In addition, we have systematically analyzed relevant papers resulting from a PubMed search for the combination of “TBI” and “corticosterone”.

2. TBI, Its Late Consequences and the Hippocampus

Post-traumatic epilepsy (PTE) is a severe complication of traumatic brain injury (TBI). It occurs in 10–20% of patients after TBI [18]. About 57% of patients with PTE suffer from medial temporal lobe epilepsy diagnosed by the semiology of epileptic seizures, signs on the EEG and MRI [19]. Histological analysis reveals a specific epilepsy-related pathology of the hippocampus, hippocampal sclerosis, in at least half of these patients [20]. The main risk factors for PTE include the severity of injury, subdural, intracerebral hematomas and early seizures [18,21,22,23,24].
Major depressive disorder development after TBI is quite frequent. Over 50% of patients met major depression disorder criteria at least once, the disease being associated with poorer health-related quality of life [25]. Pathological changes in the hippocampus may represent a basis for post-traumatic depression [26], and, taking into account the common pathophysiological mechanisms, depression is considered the main comorbid pathology for epilepsy [27]. Interestingly, anxiety and depression in patients can be diagnosed before the diagnosis of epilepsy [28], thus confirming common bases of disease mechanisms but not a unidirectional causal relationship between depression and epilepsy.
The mechanisms of late TBI complications and reorganization of neuronal networks in the hippocampus include long-lasting chronic neuroinflammation and secondary damage to the nervous tissue [5]. The causes of chronic neuroinflammation development and circuit reorganization are obviously complicated and, so far, remain obscure. Recent data suggest that disturbance in the HPA axis function plays a critical role in late post-traumatic pathology.

3. HPA Axis in Patients with TBI

HPA axis (Figure 1) is the main neuroendocrine system of the organism implementing stress response and controlling adaptive mechanisms at different levels, from subcellular to the whole organism [29,30,31]. Normally, physiological stress is realized due to HPA action and release of glucocorticoids (GCs). Clinical studies of cortisol-dependent damage in TBI are limited, and the data are scarce and contradictory. Different groups report that cortisol level after TBI is decreased [32] or increased [33,34]. TBI is an acute physiological stress and is expected to increase cortisol levels, at least in TBI patients with preserved HPA axis function. However, some patients with TBI develop dysfunction of the anterior or posterior pituitary gland, which, in turn, leads to secondary hypocorticism (a decrease in cortisol levels due to a decrease in the production of pituitary adrenocorticotropic hormone, ACTH). Agha et al. [35] showed that ACTH and cortisol production after stimulation by glucagon in patients with TBI may be normal or reduced. In patients with a reduced response, the basal cortisol level after TBI was also decreased, but in patients with a normal response, it was increased. The risk factors for adrenal insufficiency and a decreased cortisol level in the acute period of TBI are basal skull fractures, hypotension and the use of propofol [36]. Hydrocortisone replacement therapy may be associated with a favorable neurologic outcome after TBI, suggesting the involvement of corticosteroids in the consequences of brain trauma [37]. In general, signs of mild TBI, including absence of amnesia and a higher Glasgow coma scale score, are associated with higher cortisol levels [38], while the severity of coma positively correlates with acute cortisol level (within 6 h after TBI) [34]. On the contrary, during the first 3 days after TBI, the cortisol level is higher in patients with lower Glasgow coma scale score and predicts mortality [39].
The time course of cortisol level during the first weeks after TBI also depends on the initial HPA axis state. In patients with stressful events prior to brain injury, cortisol levels were significantly decreased, as compared with patients without stress before TBI [40], indicating stress-induced HPA dysfunction. In patients without stressful events before TBI, HPA function was preserved, and GC levels increased. Chronic HPA disturbances in patients with TBI are studied even less. In mild TBI, hair cortisol did not diverge before and months after TBI, its level reflecting individual coping with stress in general [41]. However, HPA dysregulation was shown two years after TBI, when hypocortisolemia and low diurnal GCs variability were detected [42]. Another study reported normal cortisol and circadian variations for two years after mild-to-moderate TBI, even with the presence of depression [43].
According to the data of basic experimental research, it can be assumed that an altered physiological response to acute stress may underlie some long-term effects of TBI. However, convincing clinical studies in this area are still lacking. Corticoid-related primary and secondary mechanisms of TBI, studied in animal models, are discussed below.

4. Distant Hippocampal Damage in Rodent TBI Models

Lateral fluid percussion brain injury model in rats [44,45] or mice [46,47] is the most conventional TBI model. A “golden standard” of TBA modeling, it allows studying the mechanisms of primary and secondary brain damage induced by TBI, though a few other models are also used [48].
Primary damage includes direct impact to brain tissue, which is accompanied by rupture of cell membranes, mechanical disruption of the blood–brain barrier, release of albumin and other blood components into the extracellular space. Acute damage causes severe metabolic disturbances inducing deficits in ATP production, energy deficiency and subsequent impairment of Na+/K+ ATPase, as well as an increase in the concentration of extracellular K+. Changes in the extracellular K+ cause depolarization of neuronal membranes and additional opening of voltage-gated calcium channels (VGCC), neurotransmitter release and a fast increase in the level of excitatory amino acids in the extracellular space [49]. Continuous changes in the concentration of extracellular ions further reduce the threshold of neuronal excitability and are aggravated by their repeated excitation. In addition, energy deficiency leads to the generation of free radicals and reactive oxygen species involved in oxidative stress and secondary damage to the nervous tissue [49]. Primary damage results in continued metabolic changes, excitotoxicity and the edema formation, inflammation, apoptosis and necrosis, representing the mechanisms of secondary brain damage.
TBI applied to the neocortex induces secondary, distant damage to the hippocampus. Neuronal death and glial activation are detected in the CA3 field and the dentate gyrus (DG) of the hippocampus [50]. Less pronounced changes are detected in the contralateral hippocampus [51,52]. Bilateral changes in the hippocampus after repeated TBI were also shown [53]. GABAergic neurons (parvalbumin (PV), calretinin, somatostatin and neuropeptide Y-immunoreactive) in the polymorphic layer of the DG are among the most vulnerable populations of hippocampal neuronal cells. Previously, we described the development of distant hippocampal damage after lateral fluid percussion brain injury in rats [54,55]. Selective neuronal cell loss in the polymorph layer of the hippocampal DG was demonstrated bilaterally; in the ipsilateral hippocampus, it was evident on day 3, but in the contralateral hippocampus, these changes were delayed and detected on day 7. Microglial activation was evident in the hippocampus bilaterally on day 7 after TBI, while pro-inflammatory cytokines mRNA levels increased bilaterally from day 1 after TBI.
It is worth noting that distant damage to the hippocampus has been reported to be a result of different extremal factors, including brain ischemia [31]. Remote hippocampal damage is a well-documented consequence of chemoconvulsant injection (kainate [56], dendrotoxin [57], pentylentetrazole [58,59]). Neuronal death [60], excitotoxicity and the involvement of glutamate receptors in distant hippocampal damage were shown after TBI in rats [61,62], indicating the similarity of damage mechanisms, irrespective of primary injury nature. In general, many epileptogenic lesions are characterized by secondary neuronal death in the DG, both in experimental and clinical settings [63]. The involvement of both NMDA and non-NMDA receptors [53,56], excitotoxicity and spreading of epileptiform activity are [64,65] discussed as mechanisms of the damage. Acute and excessive release of glutamate and aspartate leads to the activation of glutamate receptors (primarily AMPA receptors) and depolarization of neuronal membrane. Activation of glutamate, kainate and calcium-permeable AMPA receptors contributes to an increase in intracellular levels of calcium, a universal secondary messenger [49]. Excessive intracellular calcium concentration activates phospholipases, endonucleases and proteases (calpains), accelerating neuronal death as a result of excitotoxicity by apoptotic and necrotic mechanisms [66,67,68].
Thus, excessive glutamate level after TBI is a trigger for secondary neurodegeneration, which involves death of GABAergic neurons. Based on (1) the relative selectivity and distant character of hippocampal damage; (2) lack of specificity regarding the type of primary impact inducing distant hippocampal damage and (3) the involvement of both ipsilateral and contralateral hippocampus, it can be assumed that there are common systemic mechanisms underlying selective death of hippocampal neurons and, possibly, the development of chronic neuroinflammation. This selectivity may be explained by the effects of GCs, affecting the hippocampus and functional properties of the hippocampal networks [26,31].

5. Glucocorticoid Signaling, Hippocampus and Neuronal Death

GCs (corticosterone, CS, in most animals, and cortisol in humans) are essential hormones in all vertebrates, regulating two basic systems: glucose metabolism and immune response. In general, GCs suppress inflammation and increase blood glucose level by stimulating gluconeogenesis and inhibition of glucose uptake by cells [69]. Specific receptors mediating signals of GCs are present in most cells of the organism. GCs regulate the behavioral response to stress, and their receptors are widely expressed in the brain. The effects of GCs are critically determined by the specific aspects of their action [70]. During the stress response, GCs modulate the hippocampal function, affecting numerous signaling and metabolic systems [31]. It is also important that, unlike other brain structures, the basal membrane covers only 30% of the vascular surface in the hippocampus [71], which facilitates the penetration of hormones into hippocampal neurons.
The functions of the GCs in the hippocampus are mediated by high-affinity mineralocorticoid (MR) and low-affinity glucocorticoid receptors (GR). MRs bind GCs at low hormone levels, while the affinity of GRs to GCs is much lower, and the activation of these receptors occurs when GC levels increase, for example, during stress response. Each type of corticosteroid receptor is presented by two forms: intracellular cytoplasmic/nuclear receptors, exerting primarily slow genomic action (iMR, iGR), and non-genomic membrane-bound receptors (mMR, mGR), rapidly altering excitatory neurotransmission [29,72,73]. The affinity of membrane-bound and intracellular receptors decreases in the order: iMR > iGR = mMR > mGR [30] (Figure 2). The genomic binding loci of GR and MR comprise hundreds of partially overlapping DNA sites changing during the circadian cycle and stress [74]. Intracellular MRs and GRs translocate to the nucleus, where they act as nuclear transcription factors and modify gene expression, affecting protein synthesis. The genomic effects are realized within hours and may persist for many days, underlying adaptation, synaptic and cellular plasticity. Membrane-associated GRs and MRs act through G-proteins and affect the ion channels, rapidly modulating cell excitability. In general, the effects of GCs on specific cell populations depend on: (1) their concentration, (2) duration of exposure, (3) cell type with definite balance of specific intracellular and extracellular GRs and MRs.
MRs are expressed mainly in the brain regions that are crucial to the formation of memory and emotions, such as the hippocampus, amygdala, frontal, enthorinal and insular cortex. GCs trigger rapid non-genomic effects on excitability of neurons in brain through mMRs, thus influencing the cognitive and emotional functions and adaptive behavior within minutes. Besides limbic structures, GRs are also expressed in the prefrontal cortex and are involved in cognitive and executive functions, such as reasoning and attention [30,75]. GRs are involved in negative feedback on HPA axis; the amygdala stimulates the HPA axis, whereas both the hippocampus and prefrontal cortex have inhibitory effects [76,77]. It is noteworthy that after TBI in rats, CRH may increase the excitability of the amygdala and hippocampus [78].
The group of R. Sapolsky studied the effects of stress in animals for many years and showed that sustained exposure to stress induces neuronal loss in the hippocampus [79,80]. Chronic stress resulted in about 20% loss of neurons in CA3 field of the hippocampus of rats, and GCs also worsened other types of damage produced by ischemia, seizures or excitatory amino acids in CA1 and CA3 fields. The authors explained these effects by suppression of glucose transport, changes in calcium metabolism and suppression of neurotrophic factors expression [79,81,82]. Since neuronal energy metabolism is almost exclusively dependent on oxidative phosphorylation, and neurons have almost no glucose storage, they are the cells most vulnerable to energy restriction. Adaptive processes are highly energy dependent, and GCs may worsen neuronal survival.
Figure 2. Receptors of glucocorticoids [29,30,31,83]. See details in Chapter 5. Green circles—glucocorticoid molecules.
Figure 2. Receptors of glucocorticoids [29,30,31,83]. See details in Chapter 5. Green circles—glucocorticoid molecules.
Biomedicines 10 01139 g002
Another mechanism of GCs-dependent neurodegeneration may be the elevation of intracellular calcium and glutamate excitotoxicity. GCs dose-dependently affect the excitability of neurons: glutamatergic synaptic transmission is enhanced by intermediate doses of GCs acting on mMR (rapid response to stress) and is reduced by higher doses acting on GR in addition to the already activated MRs (recovery after a stressful situation) [72,84] (Figure 2).
The influence of GCs on neuronal excitability was extensively studied by the M. Jöels group. The effects of GCs on CA1 pyramidal neurons are explained by a U-shaped curve: very low (not physiological, e.g., after adrenalectomy), as well as very high CS levels suppress neuronal activity, but intermediate doses increase the amplitude of excitatory postsynaptic potentials (EPSPs) [83,85,86] (Figure 3). In animals subjected to chronic stress, changes of pyramidal neurons are rarely found. In pyramidal neurons of CA3 field, EPSP amplitude increased, likely due to NMDA action in chronically stressed animals [87] (Figure 4).
In granular neurons of the DG, MR-dependent effects on field potentials caused by the activation of AMPA receptors were demonstrated [83,85]; however, they are almost insensitive to physiological GS changes, including acute stress. In adrenalectomized rats, extremely low level of GCs reduced neuronal activity of granular cells [88] (Figure 3). In contrast, in animals exposed to chronic stress, GCs enhanced glutamatergic AMPA-mediated signaling in granular cells [89] (Figure 4).
Voltage-gated calcium channels (VGCC) are among the principal players involved in the control of calcium homeostasis. They are activated at depolarized membrane potentials and become permeable for calcium. Amplitudes of VGCC currents increased after 1–4 h of exposure to CSs, more likely through iGR signaling [90]. In chronic stress, GCs also increased calcium currents through VGCC, both in granular cells of the DG [91] and in pyramidal cells of CA1 field [92]. Thus, VGCC activation increases risk of cell death at glutamate excess, especially in chronic stress.
Figure 3. Corticosterone effects in the hippocampus at rest and during acute stress. 1, Glutamatergic synapse on granular cell. In normal conditions, granular cells are almost insensitive to physiological GSs changes, including acute stress, but extremely low levels of GCs reduce neuronal activity of granular cells [88]. 2, Glutamatergic collateral to an interneuron. 3, GABAergic synapse on granular neuron. Though little data on the modulation by GCs of collateral inhibition in the DG are available, the effects of GCs on pyramidal cells of CA1 field may be similar. 4, Glutamatergic synapse on pyramidal neuron of CA3 field. 5, Glutamatergic synapse on pyramidal neuron of CA1 field. Pyramidal neurons demonstrate U-shaped modulation by GCs: very low (not physiological), as well as very high CS levels suppress neuronal activity, but intermediate doses increase EPSP amplitude [83,85,86]. 6, GABAergic synapse on pyramidal neuron. GCs may temporarily reduce IPSP with subsequent rapid or slow elevation of inhibitory postsynaptic potential (IPSP) amplitude [93,94]. pp, perforant path; gc, granular cell; mf, mossy fibers; pc, pyramidal cells (CA3 and CA1 fields), sc, Schaffer collateral; in, interneuron. Red circle (+)—activating action; red circle (*)—activation by very low GC levels; blue circle (−)—inhibiting action; arrows show changes in GC action with increasing concentration or over time.
Figure 3. Corticosterone effects in the hippocampus at rest and during acute stress. 1, Glutamatergic synapse on granular cell. In normal conditions, granular cells are almost insensitive to physiological GSs changes, including acute stress, but extremely low levels of GCs reduce neuronal activity of granular cells [88]. 2, Glutamatergic collateral to an interneuron. 3, GABAergic synapse on granular neuron. Though little data on the modulation by GCs of collateral inhibition in the DG are available, the effects of GCs on pyramidal cells of CA1 field may be similar. 4, Glutamatergic synapse on pyramidal neuron of CA3 field. 5, Glutamatergic synapse on pyramidal neuron of CA1 field. Pyramidal neurons demonstrate U-shaped modulation by GCs: very low (not physiological), as well as very high CS levels suppress neuronal activity, but intermediate doses increase EPSP amplitude [83,85,86]. 6, GABAergic synapse on pyramidal neuron. GCs may temporarily reduce IPSP with subsequent rapid or slow elevation of inhibitory postsynaptic potential (IPSP) amplitude [93,94]. pp, perforant path; gc, granular cell; mf, mossy fibers; pc, pyramidal cells (CA3 and CA1 fields), sc, Schaffer collateral; in, interneuron. Red circle (+)—activating action; red circle (*)—activation by very low GC levels; blue circle (−)—inhibiting action; arrows show changes in GC action with increasing concentration or over time.
Biomedicines 10 01139 g003
Thus, GC excess plays an essential role in the selective vulnerability of the hippocampus, promoting calcium overload, energy deficits and secondary death of neurons [95] by increasing the susceptibility of neurons to glutamate excitotoxicity [96]. Importantly, glutamatergic axons terminate on interneurons (Figure 3), and excitotoxic damage affects primarily GABAergic interneurons, the most vulnerable population of cells in the hippocampus.
On the other hand, GCs impair the protective activation of inhibitory neurotransmitter systems during insults [79]. Patch-clamp recordings show that GCs, through the activation of MRs, reduced the frequency of spontaneous inhibitory postsynaptic potentials (IPSP) in pyramidal cells of CA1 field of the ventral hippocampus (more likely due to membrane-associated receptors) but not in the dorsal part. The effect of a GR agonist was different: it slowly increased IPSP magnitude in the hippocampus, more likely through iGRs [93]. Another group demonstrated rapid increase in spontaneous inhibitory postsynaptic currents (IPSCs) via mGR in CA1 pyramidal cells [94] (Figure 3). The authors explained that the controversial result of rapid GC action is likely due to the difference in experimental conditions.
In chronic stress, rhythmic IPSCs originating from the PV-positive GABAergic neurons was impaired due to selective PV-positive cell loss, demonstrating lack of inhibition in CA1 [94] (Figure 4). The authors explained the selective loss of PV-positive neurons by the sustained activation of interneurons and imbalance in perisomatic inhibition.
Thus, GCs modulate the excitability of the hippocampus in acute and chronic stress and enhance glutamate excitotoxicity, potentially causing selective neurodegeneration in the hippocampus. Since TBI increases GC levels in humans and in animal models [54,55], the consequences of stress can be considered as one of the secondary brain injury mechanisms.
Figure 4. Effects of GCs in chronic stress and structural post-traumatic changes in the hippocampus. GABAergic neuronal loss in the DG and neuroinflammation are histological hallmarks of late post-traumatic changes. 1, Glutamatergic synapse on granular cell. GCs enhance glutamatergic AMPA-mediated signaling [89]. 2, Glutamatergic collateral on granular neuron (mossy fiber sprouting) enhances DG excitability [97]. 4, Glutamatergic synapse on pyramidal neuron of CA3 field. Chronic stress increases EPSP amplitude via NMDA-dependent signaling [87]. Failure of inhibition due to GABAergic neuronal loss is demonstrated [50,98]. 5, Glutamatergic synapse on pyramidal neuron of CA1 field. Though little data on the modulation by chronically elevated GCs of glutamatergic synapses in the CA1 are available, the effects of GCs on pyramidal cells of CA3 field may be similar. 6, GABAergic synapse on pyramidal neuron. Rhythmic IPSCs due to loss of interneurons are demonstrated [94]. pp, perforant path; gc, granular cell; mf, mossy fibers; pc, pyramidal cells (CA3 and CA1 fields); sc, Schaffer collateral; in, interneuron. Red circle (+)—activating action; blue circle (−)—inhibiting action.
Figure 4. Effects of GCs in chronic stress and structural post-traumatic changes in the hippocampus. GABAergic neuronal loss in the DG and neuroinflammation are histological hallmarks of late post-traumatic changes. 1, Glutamatergic synapse on granular cell. GCs enhance glutamatergic AMPA-mediated signaling [89]. 2, Glutamatergic collateral on granular neuron (mossy fiber sprouting) enhances DG excitability [97]. 4, Glutamatergic synapse on pyramidal neuron of CA3 field. Chronic stress increases EPSP amplitude via NMDA-dependent signaling [87]. Failure of inhibition due to GABAergic neuronal loss is demonstrated [50,98]. 5, Glutamatergic synapse on pyramidal neuron of CA1 field. Though little data on the modulation by chronically elevated GCs of glutamatergic synapses in the CA1 are available, the effects of GCs on pyramidal cells of CA3 field may be similar. 6, GABAergic synapse on pyramidal neuron. Rhythmic IPSCs due to loss of interneurons are demonstrated [94]. pp, perforant path; gc, granular cell; mf, mossy fibers; pc, pyramidal cells (CA3 and CA1 fields); sc, Schaffer collateral; in, interneuron. Red circle (+)—activating action; blue circle (−)—inhibiting action.
Biomedicines 10 01139 g004

6. Neuroinflammation and TBI

Both primary and secondary mechanisms of brain damage discussed above are associated with neuroinflammatory response. Neuroinflammation is one of the essential mechanisms of brain damage modulated by GCs. Under normal conditions, microglial activity and cytokine release are intimately associated with neuroplasticity and memory; however, uncontrolled excessive neuroinflammation leads to cell death and further progression of brain pathology [99]. In pathological conditions, microglia play a crucial role in the expression of both pro-inflammatory (IL-1ß, IL-6, TNFα) and anti-inflammatory (IL-4, IL-10) cytokines, chemokines, as well as molecular fragments associated with damage (DAMP, damage-associated molecular patterns, such as HMGB1, ATP, S100ß). These substances enable microglial modulation of cyclooxygenase-2 and the components of the complement system [49]. Cytokines produced by inflammatory cells are released within minutes after TBI and alter the functioning of glutamate and GABAergic receptors, as well as potential-dependent ion channels, inhibit the reuptake of glutamate by astrocytes and provoke an increase in extracellular K+. Thus, cytokines can participate in hypersynchronization of neurons and the occurrence of epileptiform activity [100], as well as contribute to further neurodegeneration [101]. Microglial activation also occurs in remote areas of the brain, microglial properties and cytokine profile changing over time [102].
In the area of TBI, neutrophils and other cells of the immune system are recruited as well. These cells, along with glia, take part in the production of cytokines, chemokines, free radicals, prostaglandins and components of the complement system. The profile of peripheral immune system cells changes over time. Neutrophils first appear in the focus of injury; after 3–5 days they are replaced by mononuclear leukocytes and, to a lesser degree, by T cells, dendritic cells and natural killers [49]. The peripheral immune system is also activated. It has been shown that 1 day after TBI, the number of CD4+ and CD8+ T cells in rat spleen increases, indicating an activation of adaptive immunity. Suppression of adaptive immunity improves TBI outcomes [103]. Thus, autoimmune mechanisms are involved in the development of post-traumatic pathology [49], though their role has not been studied in detail yet.
Astrocytic gliosis in the neocortex develops about 1 week after TBI and, in the long-term period of injury astrogliosis, serves as an important histopathological marker of hippocampal sclerosis [104,105,106]. Astrocyte dysfunction may be involved in increasing the excitability of neurons and circuit reorganization via several mechanisms. Astrocytes normally participate in the utilization of extracellular K+ (due to active transport into the cell and distribution through the astrocyte system) and utilization/metabolism of glutamate. Changes in K+ homeostasis and an increase in its concentration lead to a decrease in neuronal excitability threshold, while impairment of glutamate utilization results in an increase in its toxic effects. In addition, astrocytes play an important role in water homeostasis of the brain [107] and form the brain glymphatic system involved in the development and resorption of edema, transport of metabolites and immune cells [108].
The transition from acute activation of the brain immune system to chronic neuroinflammation in TBI is the subject of quite a few studies [5,17,109]. Chronic neuroinflammation caused by TBI induces progressive edema and neurodegeneration associated with cognitive and emotional disorders [110]. The first week after TBI is an important time interval, day 7 being considered a borderline between acute and chronic post-traumatic changes. It is noteworthy that edema resorption and the early development of astrogliosis in the focus of direct impact to the neocortex was shown 7 days after TBI [104,105,106].

7. Neuroinflammation and GCs

Chronic neuroinflammation is a recognized consequence of chronic stress; its definitive association with GCs is rigorously discussed but still remains obscure [111]. The available data indicate dual effects of GCs, both anti- and pro-inflammatory. Suppression of inflammation is among the well-established systemic effects of GCs. This ability of GCs is widely used in clinical practice for treatment of inflammatory and autoimmune diseases. The activation of GRs and MRs in peripheral tissues results in inhibition of immune cell activity and induction of apoptosis in lymphocytes [112]. GCs also inhibit inflammation via several other mechanisms, including inhibition of tissue infiltration by cells from the blood, inhibition of cytokine expression, changes of lymphocyte functioning and others [111]. In the brain, GCs realize either pro- or anti-inflammatory properties depending on the degree and duration of exposure, external factors preceding injury, injury characteristics and the specific brain region [31,70,111].
The order and time period between GC increase and immune challenge may be important for the effects of GCs on neuroinflammation (Figure 5). This was confirmed in a study with administration of GCs and lipopolysaccharide (LPS, immunogenic component of Gram-negative bacteria) in a different order [113]. If GCs were injected prior to LPS (2 and 24 h), they potentiated pro-neuroinflammatory effects (TNFa, IL-1b, IL-6 expression). In contrast, GCs injected 1 h after LPS had an anti-inflammatory action in the brain. LPS injection directly into the hippocampus of the stressed animals also increased the number of reactive microglial cells and expression of pro-inflammatory cytokines [114] as compared to non-stressed animals. The second factor affecting GCs action is the duration of their exposure (Figure 5). Many groups have demonstrated that chronic stress is definitely a pro-inflammatory condition [111]. Chronic stress potentiated LPS-induced activation of several pro-inflammatory pathways, including nuclear factor kappa B (NF-κB) [115], and increased basal activation of other intracellular pathways, such as ERK1/2, p38, SAPK/JNK and AKT [116].
The interaction between GCs and the inflammatory mechanisms seems really intricate. Dexamethasone injected directly into the rat hippocampus was able to induce weak neuroinflammation but, when applied during LPS-induced neuroinflammation, evoked differential effects on pro-inflammatory cytokines expression [117]. Systemic administration of dexamethasone for 3 weeks in mice, mimicking chronic stress, induced depressive-like behavior and glucocorticoid resistance, a potential priming factor enhancing inflammatory response [118]. After ten days of corticosterone exposure in adrenalectomized rats, GCs, in a dose-dependent manner, primed microglia to pro-inflammatory stimuli by gene expression associated with inflammation (NLRP3, Iba-1, MHCII and NF-κB), thus potentiating microglial pro-inflammatory response to LPS [119]. Interestingly, diffuse TBI also primes microglia and promotes depressive-like behavior after secondary LPS-induced inflammatory challenge 1 month after trauma [120].
Recent information about relationships between inflammation, GCs and TBI is scarce. CS increased 1 hour after TBI, and its level negatively correlated with the number of peripheral T cells, confirming the anti-inflammatory effect of GCs [121]. The number of circulating T cells positively correlated with TBI core infiltration and destructive neuroinflammatory response in the brain.
Using the lateral fluid percussion model of TBI, we showed CS elevation in the blood and the hippocampus on day 3 after TBI [54,55]. The correlations between CS and neuroinflammatory response in the hippocampus were time dependent and vague. On day 3, the blood CS level negatively correlated with microglial cell count in the hippocampus. In contrast, on day 7 after TBI, when CS almost returned to baseline, noticeable and bilateral microglial activation was detected. The levels of IL-1β in the contralateral hippocampus positively correlated with CS in the same region. These results may reflect an early anti-inflammatory and latter pro-inflammatory effect of CS in TBI (Figure 6).
Since GCs modulate the secondary mechanisms of damage, the HPA axis state during trauma is also an important factor for GC action. On the one hand, the time course of cortisol levels after TBI depends on the initial HPA state; in patients experiencing stressful events before brain injury, acute cortisol levels significantly decreased during the acute period of TBI [40]. Thus, patients with previously activated HPA demonstrate impaired stress reactivity. This may defeat the positive effects of GCs (early anti-inflammatory action) and enhance negative ones (e.g., enhancement of excitotoxicity). On the other hand, moderate stress may increase the resistance of neurons to brain insults and protect from further excitotoxic damage; the expression of cytokines and neurotrophic factors may underlie the protective effects of mild stress [122].

8. CS Changes and Associated Events in Animal Models of TBI: Summary Table

Additionally, we have summarized the data generated from systematic analysis of all 48 relevant papers resulting from the PubMed search for the combination of “TBI” and “corticosterone” (21 of them combined with “hippocampus” and 7 of them with “neuroinflammation”). The results on CS changes are presented in Table S1 (Supplementary Material). In addition to CS alterations, the cellular, molecular and behavioral changes revealed in these papers are shown in the last column. The data are different and sometimes appear contradictory; however, the main reasons for discrepancies seem to be significant differences in the aims of the studies and, hence, in the experimental designs used by different groups.

9. Conclusions: TBI and Beyond

In this review, we discussed GC-dependent common mechanisms of stress- and inflammation-mediated distant hippocampal damage, focusing on the consequences of TBI. The effects of GCs on specific neuronal populations in the hippocampus depend on GC levels, duration of GC exposure and cell type (in particular, the balance of specific intracellular and extracellular GR and MR). Pro- or anti-inflammatory effects of GCs also depend on their concentration and exposure duration. Previous stress and elevated GC level prior to pro-inflammatory impact may inflate pro-inflammatory effects. Long-term and moderate elevation of GCs may also enhance neuroinflammatory response. GC-mediated long-lasting neuronal circuit changes in the hippocampus after TBI are involved in late post-traumatic pathology development, such as epilepsy, depression and cognitive impairment. Complex and diverse actions of HPA axis on neuroinflammation may be essential for late post-traumatic pathology.
Importantly, these mechanisms are applicable to remote hippocampal damage occurring after other types of focal brain damage (stroke, epilepsy) or central nervous system diseases without obvious focal injury (e.g., infections). Secondary damage to the hippocampus is shown in the middle cerebral artery (MCAO) model in rats [123,124]. MCAO induces accumulation of the pro-inflammatory cytokine IL-1β accompanied by elevated CS at the early and delayed stages of stroke [124]. High initial level of GCs and previous stress exacerbate damage to the hippocampus after brain strokes in humans [125] and rats [126].
Thus, the liaisons of excessive GCs /dysfunctional HPA axis with neuroinflammation, dangerous to the hippocampus, may be crucial for distant hippocampal damage in many brain diseases. Taking into account that the hippocampus controls both the cognitive functions and the emotional state, further research of potential links between GC signaling and the inflammatory processes in the brain and respective mechanisms is vital.

Supplementary Materials

The following supporting information can be downloaded at: https://0-www-mdpi-com.brum.beds.ac.uk/article/10.3390/biomedicines10051139/s1, Table S1: Corticosterone changes in TBI models [127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155,156,157,158,159,160,161,162,163,164,165,166,167,168,169,170,171].

Author Contributions

Conceptualization, N.V.G.; writing—original draft preparation, I.G.K.; writing—review and editing, N.V.G. All authors have read and agreed to the published version of the manuscript.

Funding

This review was funded by Russian Science Foundation, grant # 22-25-00713.

Data Availability Statement

Not applicable.

Acknowledgments

Figures are created using BioRender.com, 27 March–14 May 2022.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ACTHadrenocorticotropic hormone
AMPAα-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid
BDNFbrain-derived neurotrophic factor
CRHcorticotropin-releasing hormone
CScorticosterone
DAMPdamage-associated molecular patterns
DGdentate gyrus, hippocampal field
EPSPexcitatory postsynaptic potential
GABAgamma-Aminobutyric acid
GCsglucocorticoids
GRglucocorticoid receptor
HPAhypothalamo-pituitary axis
IL-1ßinterleukin 1 beta
IL-6interleukin 6
iMR, iGRintracellular cytoplasmic/nuclear receptors subtype
IPSCinhibitory postsynaptic current
IPSPinhibitory postsynaptic potential
LPSlipopolysaccharide
MCAOmiddle cerebral artery
mMR, mGRmembrane-associated receptors subtype
MRmineralocorticoid receptor
NF-κBnuclear factor kappa B
NMDAN-methyl-D-aspartate
PTEpost-traumatic epilepsy
PVparvalbumin
TBItraumatic brain injury
TNFαtumor necrosis factor alpha
VGCCvoltage-gated calcium channels

References

  1. Capizzi, A.; Woo, J.; Verduzco-Gutierrez, M. Traumatic Brain Injury. Med. Clin. N. Am. 2020, 104, 213–238. [Google Scholar] [CrossRef] [PubMed]
  2. Roozenbeek, B.; Maas, A.I.R.; Menon, D.K. Changing patterns in the epidemiology of traumatic brain injury. Nat. Rev. Neurol. 2013, 9, 231–236. [Google Scholar] [CrossRef] [PubMed]
  3. Lu, J.; Marmarou, A.; Choi, S.; Maas, A.; Murray, G.; Steyerberg, E.W. Mortality from traumatic brain injury. In Acta Neurochirurgica, Supplementum; Springer-Verlag: Wien, Austria, 2005; Volume 95, pp. 281–285. [Google Scholar]
  4. Dawson, S.L.; Hirsch, C.S.; Lucas, F.V.; Sebek, B.A. The contrecoup phenomenon. Hum. Pathol. 1980, 11, 155–166. [Google Scholar] [CrossRef]
  5. Simon, D.W.; McGeachy, M.J.; Bayır, H.; Clark, R.S.B.; Loane, D.J.; Kochanek, P.M. The far-reaching scope of neuroinflammation after traumatic brain injury. Nat. Rev. Neurol. 2017, 13, 171–191. [Google Scholar] [CrossRef] [Green Version]
  6. Tudor, R.M.; Thompson, C.J. Posterior pituitary dysfunction following traumatic brain injury: Review. Pituitary 2019, 22, 296–304. [Google Scholar] [CrossRef]
  7. Agha, A.; Thompson, C.J. Anterior pituitary dysfunction following traumatic brain injury (TBI). Clin. Endocrinol. 2006, 64, 481–488. [Google Scholar] [CrossRef]
  8. Aimaretti, G.; Ghigo, E. Traumatic Brain Injury and Hypopituitarism. Sci. World J. 2005, 5, 777–781. [Google Scholar] [CrossRef]
  9. Webster, J.B.; Bell, K.R. Primary adrenal insufficiency following traumatic brain injury: A case report and review of the literature. Arch. Phys. Med. Rehabil. 1997, 78, 314–318. [Google Scholar] [CrossRef]
  10. Beez, T.; Steiger, H.-J.; Etminan, N. Pharmacological targeting of secondary brain damage following ischemic or hemorrhagic stroke, traumatic brain injury, and bacterial meningitis—A systematic review and meta-analysis. BMC Neurol. 2017, 17, 209. [Google Scholar] [CrossRef]
  11. Dewall, J. The ABCs of TBI. Evidence-based guidelines for adult traumatic brain injury care. JEMS 2010, 35, 54–61. [Google Scholar]
  12. Segatore, M. Corticosteroids and Traumatic Brain Injury: Status at the End of the Decade of the Brain. J. Neurosci. Nurs. 1999, 31, 239–250. [Google Scholar] [CrossRef] [PubMed]
  13. Daskalakis, N.P.; Provost, A.C.; Hunter, R.G.; Guffanti, G. Noncoding RNAs: Stress, Glucocorticoids, and Posttraumatic Stress Disorder. Biol. Psychiatry 2018, 83, 849–865. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Weis, C.N.; Webb, E.K.; DeRoon-Cassini, T.A.; Larson, C.L. Emotion Dysregulation Following Trauma: Shared Neurocircuitry of Traumatic Brain Injury and Trauma-Related Psychiatric Disorders. Biol. Psychiatry 2022, 91, 470–477. [Google Scholar] [CrossRef] [PubMed]
  15. Vink, R.; Van Den Heuvel, C. Recent advances in the development of multifactorial therapies for the treatment of traumatic brain injury. Expert Opin. Investig. Drugs 2004, 13, 1263–1274. [Google Scholar] [CrossRef]
  16. Xu, C.; Fu, F.; Li, X.; Zhang, S. Mesenchymal stem cells maintain the microenvironment of central nervous system by regulating the polarization of macrophages/microglia after traumatic brain injury. Int. J. Neurosci. 2017, 127, 1124–1135. [Google Scholar] [CrossRef]
  17. Tapp, Z.M.; Godbout, J.P.; Kokiko-Cochran, O.N. A Tilted Axis: Maladaptive Inflammation and HPA Axis Dysfunction Contribute to Consequences of TBI. Front. Neurol. 2019, 10, 345. [Google Scholar] [CrossRef]
  18. Annegers, J.F.; Hauser, W.A.; Coan, S.P.; Rocca, W.A. A population-based study of seizures after traumatic brain injuries. N. Engl. J. Med. 1998, 338, 20–24. [Google Scholar] [CrossRef]
  19. Gupta, P.K.; Sayed, N.; Ding, K.; Agostini, M.A.; Van Ness, P.C.; Yablon, S.; Madden, C.; Mickey, B.; D’Ambrosio, R.; Diaz-Arrastia, R. Subtypes of Post-Traumatic Epilepsy: Clinical, Electrophysiological, and Imaging Features. J. Neurotrauma 2014, 31, 1439–1443. [Google Scholar] [CrossRef]
  20. Malmgren, K.; Thom, M. Hippocampal sclerosis-Origins and imaging. Epilepsia 2012, 53, 19–33. [Google Scholar] [CrossRef]
  21. Englander, J.; Bushnik, T.; Duong, T.T.; Cifu, D.X.; Zafonte, R.; Wright, J.; Hughes, R.; Bergman, W. Analyzing risk factors for late posttraumatic seizures: A prospective, multicenter investigation. Arch. Phys. Med. Rehabil. 2003, 84, 365–373. [Google Scholar] [CrossRef]
  22. Haltiner, A.M.; Temkin, N.R.; Dikmen, S.S. Risk of seizure recurrence after the first late posttraumatic seizure. Arch. Phys. Med. Rehabil. 1997, 78, 835–840. [Google Scholar] [CrossRef]
  23. Temkin, N.R. Risk Factors for Posttraumatic Seizures in Adults. Epilepsia 2003, 44, 18–20. [Google Scholar] [CrossRef] [PubMed]
  24. Pohlmann-Eden, B.; Bruckmeir, J. Predictors and dynamics of posttraumatic epilepsy. Acta Neurol. Scand. 1997, 95, 257–262. [Google Scholar] [CrossRef] [PubMed]
  25. Bombardier, C.H. Rates of Major Depressive Disorder and Clinical Outcomes Following Traumatic Brain Injury. JAMA 2010, 303, 1938. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Gulyaeva, N.V. Biochemical Mechanisms and Translational Relevance of Hippocampal Vulnerability to Distant Focal Brain Injury: The Price of Stress Response. Biochemistry 2019, 84, 1306–1328. [Google Scholar] [CrossRef] [PubMed]
  27. Malykhin, N.V.; Carter, R.; Seres, P.; Coupland, N.J. Structural changes in the hippocampus in major depressive disorder: Contributions of disease and treatment. J. Psychiatry Neurosci. 2010, 35, 337–343. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Hesdorffer, D.C.; Ishihara, L.; Mynepalli, L.; Webb, D.J.; Weil, J.; Hauser, W.A. Epilepsy, suicidality, and psychiatric disorders: A bidirectional association. Ann. Neurol. 2012, 72, 184–191. [Google Scholar] [CrossRef]
  29. de Kloet, E.R.; Karst, H.; Joëls, M. Corticosteroid hormones in the central stress response: Quick-and-slow. Front. Neuroendocrinol. 2008, 29, 268–272. [Google Scholar] [CrossRef]
  30. Maggio, N.; Segal, M. Corticosteroid Regulation of Synaptic Plasticity in the Hippocampus. Sci. World J. 2010, 10, 462–469. [Google Scholar] [CrossRef]
  31. Gulyaeva, N.V. Functional Neurochemistry of the Ventral and Dorsal Hippocampus: Stress, Depression, Dementia and Remote Hippocampal Damage. Neurochem. Res. 2019, 44, 1306–1322. [Google Scholar] [CrossRef]
  32. Kusmenkov, T.; Braunstein, M.; Schneider, H.; Bidlingmaier, M.; Prall, W.; Flatz, W.; Boecker, W.; Bogner, V. Initial free cortisol dynamics following blunt multiple trauma and traumatic brain injury: A clinical study. J. Int. Med. Res. 2019, 47, 1185–1194. [Google Scholar] [CrossRef] [PubMed]
  33. Kakati, A.; Devi, B.I.; Bhadrinarayan, V.; Kalra, P.; Shukla, D. Endocrine dysfunction following traumatic brain injury in acute stage. Indian J. Neurotrauma 2013, 10, 92–96. [Google Scholar] [CrossRef]
  34. Rao, T.P. A study of serum cortisol levels in acute head injury patients. J. Basic Clin. Physiol. Pharmacol. 2020, 32, 20190136. [Google Scholar] [CrossRef] [PubMed]
  35. Agha, A.; Rogers, B.; Mylotte, D.; Taleb, F.; Tormey, W.; Phillips, J.; Thompson, C.J. Neuroendocrine dysfunction in the acute phase of traumatic brain injury. Clin. Endocrinol. 2004, 60, 584–591. [Google Scholar] [CrossRef] [PubMed]
  36. Bensalah, M.; Donaldson, M.; Aribi, Y.; Iabassen, M.; Cherfi, L.; Nebbal, M.; Medjaher, M.; Haffaf, E.; Abdennebi, B.; Guenane, K.; et al. Cortisol evaluation during the acute phase of traumatic brain injury-A prospective study. Clin. Endocrinol. 2018, 88, 627–636. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Bernard, F.; Outtrim, J.; Lynch, A.G.; Menon, D.K.; Matta, B.F. Hemodynamic Steroid Responsiveness is Predictive of Neurological Outcome After Traumatic Brain Injury. Neurocrit. Care 2006, 5, 176–179. [Google Scholar] [CrossRef]
  38. Tanriverdi, F.; Schneider, H.J.; Aimaretti, G.; Masel, B.E.; Casanueva, F.F.; Kelestimur, F. Pituitary Dysfunction After Traumatic Brain Injury: A Clinical and Pathophysiological Approach. Endocr. Rev. 2015, 36, 305–342. [Google Scholar] [CrossRef] [Green Version]
  39. Saichan, X.; Wei, C.; Qinglong, F.; Jun, W.; Lei, X. Plasma cortisol as a noninvasive biomarker to assess severity and prognosis of patients with craniocerebral injury. Eur. Rev. Med. Pharmacol. Sci. 2016, 20, 3835–3838. [Google Scholar]
  40. Sörbo, A.; Eiving, I.; Theodorsson, E.; Rydenhag, B.; Jonsdottir, I.H. Pre-traumatic conditions can influence cortisol levels before and after a brain injury. Acta Neurol. Scand. 2020, 141, 342–350. [Google Scholar] [CrossRef]
  41. Spikman, J.M.; van der Horn, H.J.; Scheenen, M.E.; de Koning, M.E.; Savas, M.; Langerak, T.; van Rossum, E.F.C.; van der Naalt, J. Coping with stress before and after mild traumatic brain injury: A pilot hair cortisol study. Brain Inj. 2021, 35, 871–879. [Google Scholar] [CrossRef]
  42. Bay, E.; Sikorskii, A.; Gao, F. Functional Status, Chronic Stress, and Cortisol Response After Mild-to-Moderate Traumatic Brain Injury. Biol. Res. Nurs. 2009, 10, 213–225. [Google Scholar] [CrossRef] [PubMed]
  43. Bay, E.; Hagerty, B.; Williams, R.A.; Kirsch, N. Chronic Stress, Salivary Cortisol Response, Interpersonal Relatedness, and Depression Among Community-Dwelling Survivors of Traumatic Brain Injury. J. Neurosci. Nurs. 2005, 37, 4–14. [Google Scholar] [CrossRef] [PubMed]
  44. McIntosh, T.K.; Noble, L.; Andrews, B.; Faden, A.I. Traumatic brain injury in the rat: Characterization of a midline fluid-percussion model. Cent. Nerv. Syst. Trauma 1987, 4, 119–134. [Google Scholar] [CrossRef] [PubMed]
  45. Thompson, H.J.; Lifshitz, J.; Marklund, N.; Grady, M.S.; Graham, D.I.; Hovda, D.A.; McIntosh, T.K. Lateral Fluid Percussion Brain Injury: A 15-Year Review and Evaluation. J. Neurotrauma 2005, 22, 42–75. [Google Scholar] [CrossRef] [PubMed]
  46. Marklund, N. Injury Models of the Central Nervous System; Kobeissy, F.H., Dixon, C.E., Hayes, R.L., Mondello, S., Eds.; Methods in Molecular Biology; Springer: New York, NY, USA, 2016; Volume 1462, ISBN 978-1-4939-3814-8. [Google Scholar]
  47. Alder, J.; Fujioka, W.; Lifshitz, J.; Crockett, D.P.; Thakker-Varia, S. Lateral Fluid Percussion: Model of Traumatic Brain Injury in Mice. J. Vis. Exp. 2011, 54, e3063. [Google Scholar] [CrossRef] [Green Version]
  48. O’Connor, W.T.; Smyth, A.; Gilchrist, M.D. Animal models of traumatic brain injury: A critical evaluation. Pharmacol. Ther. 2011, 130, 106–113. [Google Scholar] [CrossRef]
  49. Jarrahi, A.; Braun, M.; Ahluwalia, M.; Gupta, R.V.; Wilson, M.; Munie, S.; Ahluwalia, P.; Vender, J.R.; Vale, F.L.; Dhandapani, K.M.; et al. Revisiting traumatic brain injury: From molecular mechanisms to therapeutic interventions. Biomedicines 2020, 8, 389. [Google Scholar] [CrossRef]
  50. Grady, M.S.; Charleston, J.S.; Maris, D.; Witgen, B.M.; Lifshitz, J. Neuronal and glial cell number in the hippocampus after experimental traumatic brain injury: Analysis by stereological estimation. J. Neurotrauma 2003, 20, 929–941. [Google Scholar] [CrossRef]
  51. Komoltsev, I.G.; Volkova, A.A.; Levshina, I.P.; Novikova, M.R.; Manolova, A.O.; Stepanichev, M.; Gulyaeva, N. The Number of IgG-Positive Neurons in the Rat Hippocampus Increases after Dosed Traumatic Brain Injury. Neurochem. J. 2018, 12, 256–261. [Google Scholar] [CrossRef]
  52. Tran, L.D.; Lifshitz, J.; Witgen, B.M.; Schwarzbach, E.; Cohen, A.S.; Grady, M.S. Response of the Contralateral Hippocampus to Lateral Fluid Percussion Brain Injury. J. Neurotrauma 2006, 23, 1330–1342. [Google Scholar] [CrossRef]
  53. Aungst, S.L.; Kabadi, S.V.; Thompson, S.M.; Stoica, B.A.; Faden, A.I. Repeated mild traumatic brain injury causes chronic neuroinflammation, changes in hippocampal synaptic plasticity, and associated cognitive deficits. J. Cereb. Blood Flow Metab. 2014, 34, 1223–1232. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Komoltsev, I.G.; Tret’yakova, L.V.; Frankevich, S.O.; Shirobokova, N.I.; Volkova, A.A.; Butuzov, A.V.; Novikova, M.R.; Kvichansky, A.A.; Moiseeva, Y.V.; Onufriev, M.V.; et al. Neuroinflammatory Cytokine Response, Neuronal Death, and Microglial Proliferation in the Hippocampus of Rats During the Early Period After Lateral Fluid Percussion-Induced Traumatic Injury of the Neocortex. Mol. Neurobiol. 2021, 59, 1151–1167. [Google Scholar] [CrossRef] [PubMed]
  55. Komoltsev, I.G.; Frankevich, S.O.; Shirobokova, N.I.; Volkova, A.A.; Onufriev, M.V.; Moiseeva, J.V.; Novikova, M.R.; Gulyaeva, N.V. Neuroinflammation and Neuronal Loss in the Hippocampus Are Associated with Immediate Posttraumatic Seizures and Corticosterone Elevation in Rats. Int. J. Mol. Sci. 2021, 22, 5883. [Google Scholar] [CrossRef] [PubMed]
  56. Ruth, R.E. Kainic-acid lesions of hippocampus produced iontophoretically: The problem of distant damage. Exp. Neurol. 1982, 76, 508–527. [Google Scholar] [CrossRef]
  57. Bagetta, G.; Iannone, M.; Palma, E.; Nisticò, G.; Dolly, J.O. N-methyl-D-aspartate and non-N-methyl-D-aspartate receptors mediate seizures and CA1 hippocampal damage induced by dendrotoxin-K in rats. Neuroscience 1996, 71, 613–624. [Google Scholar] [CrossRef]
  58. Becker, A.; Tiedge, A.; Grecksch, G.A. Diazepam—Its effects on the development of pentylenetetrazol kindling, related learning impairments, and neuronal cell loss. Pharmacol. Res. 1997, 35, 27–32. [Google Scholar] [CrossRef]
  59. Aniol, V.A.; Stepanichev, M.; Lazareva, N.A.; Gulyaeva, N.V. An early decrease in cell proliferation after pentylenetetrazole-induced seizures. Epilepsy Behav. 2011, 22, 433–441. [Google Scholar] [CrossRef]
  60. Huusko, N.; Römer, C.; Ndode-Ekane, X.E.; Lukasiuk, K.; Pitkänen, A. Loss of hippocampal interneurons and epileptogenesis: A comparison of two animal models of acquired epilepsy. Brain Struct. Funct. 2015, 220, 153–191. [Google Scholar] [CrossRef]
  61. Ben-Ari, Y.; Lagowska, Y.; Le Gal La Salle, G.; Tremblay, E.; Ottersen, O.P.; Naquet, R. Diazepam pretreatment reduces distant hippocampal damage induced by intra-amygdaloid injections of kainic acid. Eur. J. Pharmacol. 1978, 52, 419–420. [Google Scholar] [CrossRef]
  62. Bernert, H.; Turski, L. Traumatic brain damage prevented by the non-N-methyl-D-aspartate antagonist 2,3-dihydroxy-6-nitro-7-sulfamoylbenzo[f] quinoxaline. Proc. Natl. Acad. Sci. USA 1996, 93, 5235–5240. [Google Scholar] [CrossRef] [Green Version]
  63. Klein, P.; Dingledine, R.; Aronica, E.; Bernard, C.; Blümcke, I.; Boison, D.; Brodie, M.J.; Brooks-Kayal, A.R.; Engel, J.; Forcelli, P.A.; et al. Commonalities in epileptogenic processes from different acute brain insults: Do they translate? Epilepsia 2018, 59, 37–66. [Google Scholar] [CrossRef] [PubMed]
  64. Lerner-Natoli, M.; Rondouin, G.; Belaidi, M.; Baldy-Moulinier, M.; Kamenka, J.M. N-[1-(2-Thienyl)cyclohexyl]-piperidine (TCP) does not block kainic acid-induced status epilepticus but reduces secondary hippocampal damage. Neurosci. Lett. 1991, 122, 174–178. [Google Scholar] [CrossRef]
  65. Ben-Ari, Y.; Tremblay, E.; Ottersen, O.P.; Meldrum, B.S. The role of epileptic activity in hippocampal and “remote” cerebral lesions induced by kainic acid. Brain Res. 1980, 191, 79–97. [Google Scholar] [CrossRef]
  66. Dubreuil, C.I.; Marklund, N.; Deschamps, K.; McIntosh, T.K.; McKerracher, L. Activation of Rho after traumatic brain injury and seizure in rats. Exp. Neurol. 2006, 198, 361–369. [Google Scholar] [CrossRef]
  67. Rink, A.; Fung, K.; Trojanowski, J.Q.; Lee, V.M.; Neugebauer, E.; Mcintosh, T.K. Evidence of Apoptotic Cell Death after Experimental Traumatic Brain Injury in the Rat. Am. J. Pathol. 1995, 63, 305–309. [Google Scholar] [CrossRef]
  68. Raghupathi, R. Cell Death Mechanisms Following Traumatic Brain Injury. Brain Pathol. 2004, 14, 215–222. [Google Scholar] [CrossRef] [PubMed]
  69. Newton, R. Molecular mechanisms of glucocorticoid action: What is important? Thorax 2000, 55, 603–613. [Google Scholar] [CrossRef] [Green Version]
  70. Sorrells, S.F.; Caso, J.R.; Munhoz, C.D.; Sapolsky, R.M. The Stressed CNS: When Glucocorticoids Aggravate Inflammation. Neuron 2009, 64, 33–39. [Google Scholar] [CrossRef] [Green Version]
  71. Licht, T.; Sasson, E.; Bell, B.; Grunewald, M.; Kumar, S.; Kreisel, T.; Ben-Zvi, A.; Keshet, E. Hippocampal neural stem cells facilitate access from circulation via apical cytoplasmic processes. Elife 2020, 9, e52134. [Google Scholar] [CrossRef]
  72. Prager, E.M.; Johnson, L.R. Stress at the Synapse: Signal Transduction Mechanisms of Adrenal Steroids at Neuronal Membranes. Sci. Signal. 2009, 2, re5. [Google Scholar] [CrossRef]
  73. Karst, H.; Berger, S.; Turiault, M.; Tronche, F.; Schütz, G.; Joëls, M. Mineralocorticoid receptors are indispensable for nongenomic modulation of hippocampal glutamate transmission by corticosterone. Proc. Natl. Acad. Sci. USA 2005, 102, 19204–19207. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Mifsud, K.R.; Kennedy, C.L.M.; Salatino, S.; Sharma, E.; Price, E.M.; Haque, S.N.; Gialeli, A.; Goss, H.M.; Panchenko, P.E.; Broxholme, J.; et al. Distinct regulation of hippocampal neuroplasticity and ciliary genes by corticosteroid receptors. Nat. Commun. 2021, 12, 4737. [Google Scholar] [CrossRef] [PubMed]
  75. Maggio, N.; Krugers, H.J.; Segal, M. Stress and steroid regulation of synaptic transmission: From physiology to pathophysiology. Front. Cell. Neurosci. 2013, 6, 69. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Arnsten, A.F.T. Stress signalling pathways that impair prefrontal cortex structure and function. Nat. Rev. Neurosci. 2009, 10, 410–422. [Google Scholar] [CrossRef]
  77. Marin, M.-F.; Raymond, C.; Lupien, S.J. Memory and Stress. In Stress: Physiology, Biochemistry, and Pathology; Elsevier: Amsterdam, The Netherlands, 2019; pp. 69–78. ISBN 9780128131466. [Google Scholar]
  78. Narla, C.; Jung, P.S.; Bautista Cruz, F.; Everest, M.; Martinez-Trujillo, J.; Poulter, M.O. CRF Mediates Stress-Induced Pathophysiological High-Frequency Oscillations in Traumatic Brain Injury. eneuro 2019, 6, ENEURO.0334-18.2019. [Google Scholar] [CrossRef] [PubMed]
  79. Sapolsky, R.M. Stress, Glucocorticoids, and Damage to the Nervous System: The Current State of Confusion. Stress 1996, 1, 1–19. [Google Scholar] [CrossRef] [PubMed]
  80. Sapolsky, R.M. Stress and the brain: Individual variability and the inverted-U. Nat. Neurosci. 2015, 18, 1344–1346. [Google Scholar] [CrossRef]
  81. Mattson, M.P. Excitotoxicity. In Stress: Physiology, Biochemistry, and Pathology; Elsevier: Amsterdam, The Netherlands, 2019; pp. 125–134. [Google Scholar]
  82. Virgin, C.E., Jr.; Ha, T.P.; Packan, D.R.; Tombaugh, G.C.; Yang, S.H.; Homer, H.C.; Sapolsky, R.M. Glucocorticoids Inhibit Glucose Transport and Glutamate Uptake in Hippocampal Astrocytes: Implications for Glucocorticoid Neurotoxicity. J. Neurochem. 1991, 57, 1422–1428. [Google Scholar] [CrossRef]
  83. Joëls, M. Corticosteroid effects in the brain: U-shape it. Trends Pharmacol. Sci. 2006, 27, 244–250. [Google Scholar] [CrossRef]
  84. Gulyaeva, N.V. Glucocorticoid regulation of the glutamatergic synapse: Mechanisms of stress-dependent neuroplasticity. Ross. Fiziol. Zhurnal Im. I. M. Sechenova Sechenov Physiol. J. 2021, 107, 518–523. [Google Scholar] [CrossRef]
  85. Joëls, M.; Karst, H.; Sarabdjitsingh, R.A. The stressed brain of humans and rodents. Acta Physiol. 2018, 223, e13066. [Google Scholar] [CrossRef] [PubMed]
  86. Karst, H.; Joëls, M. Corticosterone Slowly Enhances Miniature Excitatory Postsynaptic Current Amplitude in Mice CA1 Hippocampal Cells. J. Neurophysiol. 2005, 94, 3479–3486. [Google Scholar] [CrossRef] [PubMed]
  87. Kole, M.H.P.; Swan, L.; Fuchs, E. The antidepressant tianeptine persistently modulates glutamate receptor currents of the hippocampal CA3 commissural associational synapse in chronically stressed rats. Eur. J. Neurosci. 2002, 16, 807–816. [Google Scholar] [CrossRef] [PubMed]
  88. Joëls, M.; Stienstra, C.; Karten, Y. Effect of Adrenalectomy on Membrane Properties and Synaptic Potentials in Rat Dentate Granule Cells. J. Neurophysiol. 2001, 85, 699–707. [Google Scholar] [CrossRef] [Green Version]
  89. Karst, H.; Joëls, M. Effect of Chronic Stress on Synaptic Currents in Rat Hippocampal Dentate Gyrus Neurons. J. Neurophysiol. 2003, 89, 625–633. [Google Scholar] [CrossRef] [Green Version]
  90. Joëls, M. Corticosteroid Actions in the Hippocampus. J. Neuroendocrinol. 2001, 13, 657–669. [Google Scholar] [CrossRef]
  91. Karst, H.; Joëls, M. Brief RU 38486 Treatment Normalizes the Effects of Chronic Stress on Calcium Currents in Rat Hippocampal CA1 Neurons. Neuropsychopharmacology 2007, 32, 1830–1839. [Google Scholar] [CrossRef]
  92. van Gemert, N.G.; Joëls, M. Effect of Chronic Stress and Mifepristone Treatment on Voltage-Dependent Ca2+ Currents in Rat Hippocampal Dentate Gyrus. J. Neuroendocrinol. 2006, 18, 732–741. [Google Scholar] [CrossRef]
  93. Maggio, N.; Segal, M. Differential Corticosteroid Modulation of Inhibitory Synaptic Currents in the Dorsal and Ventral Hippocampus. J. Neurosci. 2009, 29, 2857–2866. [Google Scholar] [CrossRef]
  94. Hu, W.; Zhang, M.; Czéh, B.; Flügge, G.; Zhang, W. Stress Impairs GABAergic Network Function in the Hippocampus by Activating Nongenomic Glucocorticoid Receptors and Affecting the Integrity of the Parvalbumin-Expressing Neuronal Network. Neuropsychopharmacology 2010, 35, 1693–1707. [Google Scholar] [CrossRef]
  95. de Kloet, E.R.; Meijer, O.C.; de Nicola, A.F.; de Rijk, R.H.; Joëls, M. Importance of the brain corticosteroid receptor balance in metaplasticity, cognitive performance and neuro-inflammation. Front. Neuroendocrinol. 2018, 49, 124–145. [Google Scholar] [CrossRef] [PubMed]
  96. Sorrells, S.F.; Munhoz, C.D.; Manley, N.C.; Yen, S.; Sapolsky, R.M. Glucocorticoids Increase Excitotoxic Injury and Inflammation in the Hippocampus of Adult Male Rats. Neuroendocrinology 2014, 100, 129–140. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Hunt, R.F.; Haselhorst, L.A.; Schoch, K.M.; Bach, E.C.; Rios-Pilier, J.; Scheff, S.W.; Saatman, K.E.; Smith, B.N. Posttraumatic mossy fiber sprouting is related to the degree of cortical damage in three mouse strains. Epilepsy Res. 2012, 99, 167–170. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Lowenstein, D.H.; Thomas, M.J.; Smith, D.H.; McIntosh, T.K. Selective vulnerability of dentate hilar neurons following traumatic brain injury: A potential mechanistic link between head trauma and disorders of the hippocampus. J. Neurosci. 1992, 12, 4846–4853. [Google Scholar] [CrossRef] [PubMed]
  99. DiSabato, D.J.; Quan, N.; Godbout, J.P. Neuroinflammation: The devil is in the details. J. Neurochem. 2016, 139, 136–153. [Google Scholar] [CrossRef] [Green Version]
  100. Vezzani, A.; Auvin, S.; Ravizza, T.; Aronica, E. Glia-Neuronal Interactions in Ictogenesis and Epileptogenesis: Role of Inflammatory Mediators. In Jasper’s Basic Mechanisms of the Epilepsies [Internet], 4th ed.; Noebels, J.L., Avoli, M., Rogawski, M., Olsen, R., Delgado-Escueta, A., Eds.; Oxford University Press: Oxford, UK, 2012; ISBN 9780199746545. [Google Scholar]
  101. Vezzani, A.; Balosso, S.; Ravizza, T. Inflammation and epilepsy. In Handbook of Clinical Neurology; Elsevier: Amsterdam, The Netherlands, 2012; Volume 107, pp. 163–175. ISBN 9780444528988. [Google Scholar]
  102. Donat, C.K.; Scott, G.; Gentleman, S.M.; Sastre, M. Microglial Activation in Traumatic Brain Injury. Front. Aging Neurosci. 2017, 9, 208. [Google Scholar] [CrossRef] [Green Version]
  103. Tobin, R.P.; Mukherjee, S.; Kain, J.M.; Rogers, S.K.; Henderson, S.K.; Motal, H.L.; Rogers, M.K.N.; Shapiro, L.A. Traumatic brain injury causes selective, CD74-dependent peripheral lymphocyte activation that exacerbates neurodegeneration. Acta Neuropathol. Commun. 2014, 2, 143. [Google Scholar] [CrossRef] [Green Version]
  104. Hicks, R.; Soares, H.; Smith, D.; McIntosh, T. Temporal and spatial characterization of neuronal injury following lateral fluid-percussion brain injury in the rat. Acta Neuropathol. 1996, 91, 236–246. [Google Scholar] [CrossRef]
  105. Cortez, S.C.; McIntosh, T.K.; Noble, L.J. Experimental fluid percussion brain injury: Vascular disruption and neuronal and glial alterations. Brain Res. 1989, 482, 271–282. [Google Scholar] [CrossRef]
  106. Lescot, T.; Fulla-Oller, L.; Po, C.; Chen, X.R.; Puybasset, L.; Gillet, B.; Plotkine, M.; Meric, P.; Marchand-Leroux, C. Temporal and Regional Changes after Focal Traumatic Brain Injury. J. Neurotrauma 2010, 27, 85–94. [Google Scholar] [CrossRef]
  107. Xu, S.; Sun, Q.; Fan, J.; Jiang, Y.; Yang, W.; Cui, Y.; Yu, Z.; Jiang, H.; Li, B. Role of Astrocytes in Post-traumatic Epilepsy. Front. Neurol. 2019, 10, 1149. [Google Scholar] [CrossRef] [PubMed]
  108. Thrane, A.S.; Rangroo Thrane, V.; Nedergaard, M. Drowning stars: Reassessing the role of astrocytes in brain edema. Trends Neurosci. 2014, 37, 620–628. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. D’amico, R.; Salinaro, A.T.; Fusco, R.; Cordaro, M.; Impellizzeri, D.; Scuto, M.; Ontario, M.L.; Dico, G.L.; Cuzzocrea, S.; Di Paola, R.; et al. Hericium erinaceus and coriolus versicolor modulate molecular and biochemical changes after traumatic brain injury. Antioxidants 2021, 10, 898. [Google Scholar] [CrossRef] [PubMed]
  110. Chiu, C.-C.; Liao, Y.-E.; Yang, L.-Y.; Wang, J.-Y.; Tweedie, D.; Karnati, H.K.; Greig, N.H.; Wang, J.-Y. Neuroinflammation in animal models of traumatic brain injury. J. Neurosci. Methods 2016, 272, 38–49. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. Bolshakov, A.P.; Tret’yakova, L.V.; Kvichansky, A.A.; Gulyaeva, N.V. Glucocorticoids: Dr. Jekyll and Mr. Hyde of Hippocampal Neuroinflammation. Biochemistry 2021, 86, 156–167. [Google Scholar] [CrossRef] [PubMed]
  112. Cain, D.W.; Cidlowski, J.A. Immune regulation by glucocorticoids. Nat. Rev. Immunol. 2017, 17, 233–247. [Google Scholar] [CrossRef] [PubMed]
  113. Frank, M.G.; Miguel, Z.D.; Watkins, L.R.; Maier, S.F. Prior exposure to glucocorticoids sensitizes the neuroinflammatory and peripheral inflammatory responses to E. coli lipopolysaccharide. Brain. Behav. Immun. 2010, 24, 19–30. [Google Scholar] [CrossRef]
  114. Espinosa-Oliva, A.M.; de Pablos, R.M.; Villarán, R.F.; Argüelles, S.; Venero, J.L.; Machado, A.; Cano, J. Stress is critical for LPS-induced activation of microglia and damage in the rat hippocampus. Neurobiol. Aging 2011, 32, 85–102. [Google Scholar] [CrossRef]
  115. Munhoz, C.D. Chronic Unpredictable Stress Exacerbates Lipopolysaccharide-Induced Activation of Nuclear Factor- B in the Frontal Cortex and Hippocampus via Glucocorticoid Secretion. J. Neurosci. 2006, 26, 3813–3820. [Google Scholar] [CrossRef]
  116. Munhoz, C.D.; Sorrells, S.F.; Caso, J.R.; Scavone, C.; Sapolsky, R.M. Glucocorticoids Exacerbate Lipopolysaccharide-Induced Signaling in the Frontal Cortex and Hippocampus in a Dose-Dependent Manner. J. Neurosci. 2010, 30, 13690–13698. [Google Scholar] [CrossRef] [Green Version]
  117. Tretyakova, L.V.; Kvichansky, A.A.; Bolshakov, A.P.; Gulyaeva, N.V. Dexamethasone Modulates Lipopolysaccharide-Induced Expression of Proinflammatory Cytokines in Rat Hippocampus. Neurochem. J. 2021, 15, 302–307. [Google Scholar] [CrossRef]
  118. Skupio, U.; Tertil, M.; Sikora, M.; Golda, S.; Wawrzczak-Bargiela, A.; Przewlocki, R. Behavioral and molecular alterations in mice resulting from chronic treatment with dexamethasone: Relevance to depression. Neuroscience 2015, 286, 141–150. [Google Scholar] [CrossRef] [PubMed]
  119. Frank, M.G.; Hershman, S.A.; Weber, M.D.; Watkins, L.R.; Maier, S.F. Chronic exposure to exogenous glucocorticoids primes microglia to pro-inflammatory stimuli and induces NLRP3 mRNA in the hippocampus. Psychoneuroendocrinology 2014, 40, 191–200. [Google Scholar] [CrossRef] [Green Version]
  120. Fenn, A.M.; Gensel, J.C.; Huang, Y.; Popovich, P.G.; Lifshitz, J.; Godbout, J.P. Immune Activation Promotes Depression 1 Month After Diffuse Brain Injury: A Role for Primed Microglia. Biol. Psychiatry 2014, 76, 575–584. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  121. Dong, T.; Zhi, L.; Bhayana, B.; Wu, M.X. Cortisol-induced immune suppression by a blockade of lymphocyte egress in traumatic brain injury. J. Neuroinflamm. 2016, 13, 197. [Google Scholar] [CrossRef] [Green Version]
  122. Raefsky, S.M.; Mattson, M.P. Adaptive responses of neuronal mitochondria to bioenergetic challenges: Roles in neuroplasticity and disease resistance. Free Radic. Biol. Med. 2017, 102, 203–216. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Gulyaeva, N.V.; Onufriev, M.V.; Moiseeva, Y.V. Ischemic Stroke, Glucocorticoids, and Remote Hippocampal Damage: A Translational Outlook and Implications for Modeling. Front. Neurosci. 2021, 15, 1–10. [Google Scholar] [CrossRef]
  124. Onufriev, M.V.; Moiseeva, Y.V.; Zhanina, M.Y.; Lazareva, N.A.; Gulyaeva, N.V. A Comparative Study of Koizumi and Longa Methods of Intraluminal Filament Middle Cerebral Artery Occlusion in Rats: Early Corticosterone and Inflammatory Response in the Hippocampus and Frontal Cortex. Int. J. Mol. Sci. 2021, 22, 13544. [Google Scholar] [CrossRef]
  125. Ben Assayag, E.; Tene, O.; Korczyn, A.D.; Shopin, L.; Auriel, E.; Molad, J.; Hallevi, H.; Kirschbaum, C.; Bornstein, N.M.; Shenhar-Tsarfaty, S.; et al. High hair cortisol concentrations predict worse cognitive outcome after stroke: Results from the TABASCO prospective cohort study. Psychoneuroendocrinology 2017, 82, 133–139. [Google Scholar] [CrossRef]
  126. Kwon, S.K.C.; Kovesdi, E.; Gyorgy, A.B.; Wingo, D.; Kamnaksh, A.; Walker, J.; Long, J.B.; Agoston, D.V. Stress and traumatic brain injury: A behavioral, proteomics, and histological study. Front. Neurol. 2011, 2, 12. [Google Scholar] [CrossRef] [Green Version]
  127. Kamnaksh, A.; Kovesdi, E.; Kwon, S.-K.; Wingo, D.; Ahmed, F.; Grunberg, N.E.; Long, J.; Agoston, D.V. Factors Affecting Blast Traumatic Brain Injury. J. Neurotrauma 2011, 28, 2145–2153. [Google Scholar] [CrossRef] [PubMed]
  128. Russell, A.L.; Richardson, M.R.; Bauman, B.M.; Hernandez, I.M.; Saperstein, S.; Handa, R.J.; Wu, T.J. Differential Responses of the HPA Axis to Mild Blast Traumatic Brain Injury in Male and Female Mice. Endocrinology 2018, 159, 2363–2375. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Zhang, B.; Xu, X.; Niu, F.; Mao, X.; Dong, J.; Yang, M.; Gao, F.; Liu, B. Corticosterone Replacement Alleviates Hippocampal Neuronal Apoptosis and Spatial Memory Impairment Induced by Dexamethasone via Promoting Brain Corticosteroid Receptor Rebalance after Traumatic Brain Injury. J. Neurotrauma 2020, 37, 262–272. [Google Scholar] [CrossRef]
  130. Zhang, B.; Yang, M.; Yan, Q.; Xu, X.; Niu, F.; Dong, J.; Zhuang, Y.; Lu, S.; Ge, Q.; Liu, B. The Dual Dose-Dependent Effects of Corticosterone on Hippocampal Cell Apoptosis After Traumatic Brain Injury Depend on the Activation Ratio of Mineralocorticoid Receptors to Glucocorticoid Receptors. Front. Pharmacol. 2021, 12, 713715. [Google Scholar] [CrossRef]
  131. Geddes, R.I.; Kapoor, A.; Hayashi, K.; Rauh, R.; Wehber, M.; Bongers, Q.; Jansen, A.D.; Anderson, I.M.; Farquhar, G.; Vadakkadath-Meethal, S.; et al. Hypogonadism induced by surgical stress and brain trauma is reversed by human chorionic gonadotropin in male rats: A potential therapy for surgical and TBI-induced hypogonadism? Endocrinol. Diabetes Metab. 2021, 4, e00239. [Google Scholar] [CrossRef] [PubMed]
  132. Zhang, B.; Zhu, X.; Wang, L.; Hou, Z.; Hao, S.; Yang, M.; Gao, F.; Liu, B. Inadequate Expression and Activation of Mineralocorticoid Receptor Aggravates Spatial Memory Impairment after Traumatic Brain Injury. Neuroscience 2020, 424, 1–11. [Google Scholar] [CrossRef] [PubMed]
  133. Zhang, B.; Bai, M.; Xu, X.; Yang, M.; Niu, F.; Gao, F.; Liu, B. Corticosteroid receptor rebalancing alleviates critical illness-related corticosteroid insufficiency after traumatic brain injury by promoting paraventricular nuclear cell survival via Akt/CREB/BDNF signaling. J. Neuroinflammat. 2020, 17, 318. [Google Scholar] [CrossRef]
  134. Gottesfeld, Z.; Moore, A.N.; Dash, P.K. Acute Ethanol Intake Attenuates Inflammatory Cytokines after Brain Injury in Rats: A Possible Role for Corticosterone. J. Neurotrauma 2002, 19, 317–326. [Google Scholar] [CrossRef]
  135. Taylor, A.N.; Rahman, S.U.; Tio, D.L.; Sanders, M.J.; Bando, J.K.; Truong, A.H.; Prolo, P. Lasting Neuroendocrine-Immune Effects of Traumatic Brain Injury in Rats. J. Neurotrauma 2006, 23, 1802–1813. [Google Scholar] [CrossRef]
  136. Niesman, P.J.; Wei, J.; LaPorte, M.J.; Carlson, L.J.; Nassau, K.L.; Bao, G.C.; Cheng, J.P.; de la Tremblaye, P.; Lajud, N.; Bondi, C.O.; et al. Albeit nocturnal, rats subjected to traumatic brain injury do not differ in neurobehavioral performance whether tested during the day or night. Neurosci. Lett. 2018, 665, 212–216. [Google Scholar] [CrossRef]
  137. Meffre, D.; Pianos, A.; Liere, P.; Eychenne, B.; Cambourg, A.; Schumacher, M.; Stein, D.G.; Guennoun, R. Steroid Profiling in Brain and Plasma of Male and Pseudopregnant Female Rats after Traumatic Brain Injury: Analysis by Gas Chromatography/Mass Spectrometry. Endocrinology 2007, 148, 2505–2517. [Google Scholar] [CrossRef] [PubMed]
  138. Taylor, A.N.; Tio, D.L.; Paydar, A.; Sutton, R.L. Sex Differences in Thermal, Stress, and Inflammatory Responses to Minocycline Administration in Rats with Traumatic Brain Injury. J. Neurotrauma 2018, 35, 630–638. [Google Scholar] [CrossRef] [PubMed]
  139. Geddes, R.I.; Hayashi, K.; Bongers, Q.; Wehber, M.; Anderson, I.M.; Jansen, A.D.; Nier, C.; Fares, E.; Farquhar, G.; Kapoor, A.; et al. Conjugated Linoleic Acid Administration Induces Amnesia in Male Sprague Dawley Rats and Exacerbates Recovery from Functional Deficits Induced by a Controlled Cortical Impact Injury. PLoS ONE 2017, 12, e0169494. [Google Scholar] [CrossRef] [PubMed]
  140. McCullers, D.L.; Sullivan, P.G.; Scheff, S.W.; Herman, J.P. Traumatic brain injury regulates adrenocorticosteroid receptor mRNA levels in rat hippocampus. Brain Res. 2002, 947, 41–49. [Google Scholar] [CrossRef]
  141. Taylor, A.N.; Rahman, S.U.; Sanders, N.C.; Tio, D.L.; Prolo, P.; Sutton, R.L. Injury Severity Differentially Affects Short- and Long-Term Neuroendocrine Outcomes of Traumatic Brain Injury. J. Neurotrauma 2008, 25, 311–323. [Google Scholar] [CrossRef]
  142. Taylor, A.N.; Tio, D.L.; Sutton, R.L. Restoration of Neuroendocrine Stress Response by Glucocorticoid Receptor or GABA A Receptor Antagonists after Experimental Traumatic Brain Injury. J. Neurotrauma 2013, 30, 1250–1256. [Google Scholar] [CrossRef] [Green Version]
  143. Kim, D.-H.; Ko, I.-G.; Kim, B.-K.; Kim, T.-W.; Kim, S.-E.; Shin, M.-S.; Kim, C.-J.; Kim, H.; Kim, K.-M.; Baek, S.-S. Treadmill exercise inhibits traumatic brain injury-induced hippocampal apoptosis. Physiol. Behav. 2010, 101, 660–665. [Google Scholar] [CrossRef]
  144. Lajud, N.; Roque, A.; Cheng, J.P.; Bondi, C.O.; Kline, A.E. Early Life Stress Preceding Mild Pediatric Traumatic Brain Injury Increases Neuroinflammation but Does Not Exacerbate Impairment of Cognitive Flexibility during Adolescence. J. Neurotrauma 2021, 38, 411–421. [Google Scholar] [CrossRef]
  145. Taylor, A.N.; Rahman, S.U.; Tio, D.L.; Gardner, S.M.; Kim, C.J.; Sutton, R.L. Injury Severity Differentially Alters Sensitivity to Dexamethasone after Traumatic Brain Injury. J. Neurotrauma 2010, 27, 1081–1089. [Google Scholar] [CrossRef]
  146. Baykara, B.; Aksu, I.; Buyuk, E.; Kiray, M.; Sisman, A.; Baykara, B.; Dayi, A.; Tas, A.; Ozdemir, D.; Arda, M.; et al. Progesterone treatment decreases traumatic brain injury induced anxiety and is correlated with increased serum IGF-1 levels; prefrontal cortex, amygdala, hippocampus neuron density; and reduced serum corticosterone levels in immature rats. Biotech. Histochem. 2013, 88, 250–257. [Google Scholar] [CrossRef]
  147. Baykara, B.; Cetin, F.; Baykara, B.; Aksu, I.; Dayi, A.; Kiray, M.; Sisman, A.R.; Ozdemir, D.; Arda, M.N.; Uysal, N. Anxiety caused by traumatic brain injury correlates to decreased prefrontal cortex vegf immunoreactivity and neuron density in immature rats. Turk. Neurosurg. 2012, 22, 604–610. [Google Scholar] [CrossRef] [PubMed]
  148. Stetter, C.; Lopez-Caperuchipi, S.; Hopp-Krämer, S.; Bieber, M.; Kleinschnitz, C.; Sirén, A.-L.; Albert-Weißenberger, C. Amelioration of Cognitive and Behavioral Deficits after Traumatic Brain Injury in Coagulation Factor XII Deficient Mice. Int. J. Mol. Sci. 2021, 22, 4855. [Google Scholar] [CrossRef] [PubMed]
  149. Macolino, C.M.; Daiutolo, B.V.; Albertson, B.K.; Elliott, M.B. Mechanical allodynia induced by traumatic brain injury is independent of restraint stress. J. Neurosci. Methods 2014, 226, 139–146. [Google Scholar] [CrossRef] [PubMed]
  150. Kosari-Nasab, M.; Sadeghi, T.; Bashiri, H.; Shokouhi, G.; Salari, A.-A. The blockade of corticotropin-releasing factor 1 receptor attenuates anxiety-related symptoms and hypothalamus–pituitary–adrenal axis reactivity in mice with mild traumatic brain injury. Behav. Pharmacol. 2019, 30, 220–228. [Google Scholar] [CrossRef]
  151. Sherman, M.; Liu, M.-M.; Birnbaum, S.; Wolf, S.E.; Minei, J.P.; Gatson, J.W. Adult obese mice suffer from chronic secondary brain injury after mild TBI. J. Neuroinflammat. 2016, 13, 171. [Google Scholar] [CrossRef] [Green Version]
  152. Sanchez, C.M.; Titus, D.J.; Wilson, N.M.; Freund, J.E.; Atkins, C.M. Early Life Stress Exacerbates Outcome after Traumatic Brain Injury. J. Neurotrauma 2021, 38, 555–565. [Google Scholar] [CrossRef]
  153. WHITE-GBADEBO, D.; HAMM, R.J. Chronic Corticosterone Treatment Potentiates Deficits Following Traumatic Brain Injury in Rats: Implications for Aging. J. Neurotrauma 1993, 10, 297–306. [Google Scholar] [CrossRef]
  154. Grundy, P.L.; Patel, N.; Harbuz, M.S.; Lightman, S.L.; Sharples, P.M. Adrenalectomy further suppresses the NT-3 mRNA response to traumatic brain injury but this effect is not reversed with corticosterone. Mol. Brain Res. 2004, 120, 188–192. [Google Scholar] [CrossRef]
  155. Chen, X.; Zhao, Z.; Chai, Y.; Luo, L.; Jiang, R.; Zhang, J. The incidence of critical-illness-related-corticosteroid-insufficiency is associated with severity of traumatic brain injury in adult rats. J. Neurol. Sci. 2014, 342, 93–100. [Google Scholar] [CrossRef]
  156. Grundy, P.L.; Patel, N.; Harbuz, M.S.; Lightman, S.L.; Sharples, P.M. Glucocorticoids modulate BDNF mRNA expression in the rat hippocampus after traumatic brain injury. Neuroreport 2000, 11, 3381–3384. [Google Scholar] [CrossRef]
  157. Chen, X.; Zhang, B.; Chai, Y.; Dong, B.; Lei, P.; Jiang, R.; Zhang, J. Methylprednisolone exacerbates acute critical illness-related corticosteroid insufficiency associated with traumatic brain injury in rats. Brain Res. 2011, 1382, 298–307. [Google Scholar] [CrossRef] [PubMed]
  158. Griesbach, G.S.; Tio, D.L.; Nair, S.; Hovda, D.A. Recovery of Stress Response Coincides with Responsiveness to Voluntary Exercise after Traumatic Brain Injury. J. Neurotrauma 2014, 31, 674–682. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  159. Griesbach, G.S.; Tio, D.L.; Vincelli, J.; McArthur, D.L.; Taylor, A.N. Differential Effects of Voluntary and Forced Exercise on Stress Responses after Traumatic Brain Injury. J. Neurotrauma 2012, 29, 1426–1433. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  160. Zhu, H.; Zhao, Z.; Zhou, Y.; Chen, X.; Li, Y.; Liu, X.; Lu, H.; Zhang, Y.; Zhang, J. High-dose glucocorticoid aggravates TBI-associated corticosteroid insufficiency by inducing hypothalamic neuronal apoptosis. Brain Res. 2013, 1541, 69–80. [Google Scholar] [CrossRef] [PubMed]
  161. Tapp, Z.M.; Kumar, J.E.; Witcher, K.G.; Atluri, R.R.; Velasquez, J.A.; O’Neil, S.M.; Dziabis, J.E.; Bray, C.E.; Sheridan, J.F.; Godbout, J.P.; et al. Sleep Disruption Exacerbates and Prolongs the Inflammatory Response to Traumatic Brain Injury. J. Neurotrauma 2020, 37, 1829–1843. [Google Scholar] [CrossRef] [PubMed]
  162. Rowe, R.K.; Ortiz, J.B.; Thomas, T.C. Mild and Moderate Traumatic Brain Injury and Repeated Stress Affect Corticosterone in the Rat. Neurotrauma Rep. 2020, 1, 113–124. [Google Scholar] [CrossRef]
  163. Rowe, R.K.; Rumney, B.M.; May, H.G.; Permana, P.; Adelson, P.D.; Harman, S.M.; Lifshitz, J.; Thomas, T.C. Diffuse traumatic brain injury affects chronic corticosterone function in the rat. Endocr. Connect. 2016, 5, 152–166. [Google Scholar] [CrossRef] [Green Version]
  164. Thomas, T.C.; Stockhausen, E.M.; Law, L.M.; Khodadad, A.; Lifshitz, J. Rehabilitation modality and onset differentially influence whisker sensory hypersensitivity after diffuse traumatic brain injury in the rat. Restor. Neurol. Neurosci. 2017, 35, 611–629. [Google Scholar] [CrossRef] [Green Version]
  165. McNamara, K.C.S.; Lisembee, A.M.; Lifshitz, J. The Whisker Nuisance Task Identifies a Late-Onset, Persistent Sensory Sensitivity in Diffuse Brain-Injured Rats. J. Neurotrauma 2010, 27, 695–706. [Google Scholar] [CrossRef]
  166. Rowe, R.K.; Harrison, J.L.; O’Hara, B.F.; Lifshitz, J. Recovery of Neurological Function Despite Immediate Sleep Disruption Following Diffuse Brain Injury in the Mouse: Clinical Relevance to Medically Untreated Concussion. Sleep 2014, 37, 743–752. [Google Scholar] [CrossRef] [Green Version]
  167. Arnaud, F.; Pappas, G.; Chen, Y.; Maudlin-Jeronimo, E.; McCarron, R. Effect of acute restraint stress in a polytrauma rat model. Neurosci. Lett. 2018, 684, 91–97. [Google Scholar] [CrossRef] [PubMed]
  168. Taguchi, S.; Choudhury, M.E.; Miyanishi, K.; Nakanishi, Y.; Kameda, K.; Abe, N.; Yano, H.; Yorozuya, T.; Tanaka, J. Aggravating effects of treadmill exercises during the early-onset period in a rat traumatic brain injury model: When should rehabilitation exercises be initiated? IBRO Rep. 2019, 7, 82–89. [Google Scholar] [CrossRef] [PubMed]
  169. Chakraborty, N.; Hammamieh, R.; Gautam, A.; Miller, S.-A.; Condlin, M.L.; Jett, M.; Scrimgeour, A.G. TBI weight-drop model with variable impact heights differentially perturbs hippocampus-cerebellum specific transcriptomic profile. Exp. Neurol. 2021, 335, 113516. [Google Scholar] [CrossRef] [PubMed]
  170. Jindal, A.; Mahesh, R.; Bhatt, S.; Pandey, D. Molecular modifications by regulating cAMP signaling and oxidant-antioxidant defence mechanisms, produce antidepressant-like effect: A possible mechanism of etazolate aftermaths of impact accelerated traumatic brain injury in rat model. Neurochem. Int. 2017, 111, 3–11. [Google Scholar] [CrossRef]
  171. Doulames, V.M.; Vilcans, M.; Lee, S.; Shea, T.B. Social interaction attenuates the extent of secondary neuronal damage following closed head injury in mice. Front. Behav. Neurosci. 2015, 9, 275. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Hypothalamic–pituitary–adrenal (HPA) axis. Neuroendocrine response to stress includes the reaction of HPA axis: the release of hypothalamic corticotropin-releasing hormone (CRH), which stimulates the release of adrenocorticotropic hormone (ACTH) from the pituitary gland and, finally, the release of glucocorticoids (GCs) from the adrenal glands (corticosterone in most rodents; cortisol in humans). GCs enter the blood circulation, implementing both peripheral and central action via specific receptors in almost all organs and tissues, including the brain. The prefrontal cortex, hippocampus and amygdala control the activity of the hypothalamus, thus regulating the HPA axis [29,30,31].
Figure 1. Hypothalamic–pituitary–adrenal (HPA) axis. Neuroendocrine response to stress includes the reaction of HPA axis: the release of hypothalamic corticotropin-releasing hormone (CRH), which stimulates the release of adrenocorticotropic hormone (ACTH) from the pituitary gland and, finally, the release of glucocorticoids (GCs) from the adrenal glands (corticosterone in most rodents; cortisol in humans). GCs enter the blood circulation, implementing both peripheral and central action via specific receptors in almost all organs and tissues, including the brain. The prefrontal cortex, hippocampus and amygdala control the activity of the hypothalamus, thus regulating the HPA axis [29,30,31].
Biomedicines 10 01139 g001
Figure 5. Effects of GCs on neuroinflammation depend on time of damage [54,55,111,113]. Timing of GCs exposure is critical for its pro- or anti-inflammatory action in the brain.
Figure 5. Effects of GCs on neuroinflammation depend on time of damage [54,55,111,113]. Timing of GCs exposure is critical for its pro- or anti-inflammatory action in the brain.
Biomedicines 10 01139 g005
Figure 6. Local and systemic effects of TBI ([54,55]). Based on (1) selectivity and distant character of hippocampal damage, (2) lack of specificity to the type of primary impact leading to distant hippocampal damage and (3) involvement of both ipsilateral and contralateral hippocampus in models of unilateral primary neocortical injury, it can be assumed that there are common CS-dependent mechanisms underlying selective death of hippocampal neurons and chronic neuroinflammation.
Figure 6. Local and systemic effects of TBI ([54,55]). Based on (1) selectivity and distant character of hippocampal damage, (2) lack of specificity to the type of primary impact leading to distant hippocampal damage and (3) involvement of both ipsilateral and contralateral hippocampus in models of unilateral primary neocortical injury, it can be assumed that there are common CS-dependent mechanisms underlying selective death of hippocampal neurons and chronic neuroinflammation.
Biomedicines 10 01139 g006
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Komoltsev, I.G.; Gulyaeva, N.V. Brain Trauma, Glucocorticoids and Neuroinflammation: Dangerous Liaisons for the Hippocampus. Biomedicines 2022, 10, 1139. https://0-doi-org.brum.beds.ac.uk/10.3390/biomedicines10051139

AMA Style

Komoltsev IG, Gulyaeva NV. Brain Trauma, Glucocorticoids and Neuroinflammation: Dangerous Liaisons for the Hippocampus. Biomedicines. 2022; 10(5):1139. https://0-doi-org.brum.beds.ac.uk/10.3390/biomedicines10051139

Chicago/Turabian Style

Komoltsev, Ilia G., and Natalia V. Gulyaeva. 2022. "Brain Trauma, Glucocorticoids and Neuroinflammation: Dangerous Liaisons for the Hippocampus" Biomedicines 10, no. 5: 1139. https://0-doi-org.brum.beds.ac.uk/10.3390/biomedicines10051139

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop