Loading

Journal of Neurological Disorders and Stroke

Alteration of Hippocampal Cytokines and Astrocyte Morphology Observed in Rats 24 Hour after Fluid Percussion Injury

Research Article | Open Access | Volume 2 | Issue 3

  • 1. Department of Surgery, Scott & White Hospital, USA
  • 2. Central Texas Veterans Health System, USA
  • 3. Departments of Surgery and Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, USA
+ Show More - Show Less
Corresponding Authors
Lee A. Shapiro, Department of Surgery, College of Medicine, Texas A&M University, 1901 South 1st Street, Building 205, Temple, TX 76504, USA, Tel: 1-254-743-2365
ABSTRACT

Traumatic brain injury (TBI) is one of the few known etiological factors contributing to the development of post-traumatic epilepsy (PTE). An understanding of the mechanisms involved in the development of PTE is vital because PTEs are amongst the most difficult to treat, and are often resistant to first and second line anti-epileptic treatment regimens. TBI-induced inflammation, neuroplasticity and neuropathology in the hippocampus have been observed in animal models of TBI. Components of these alterations have been implicated in TBI-pathogenesis and the epileptogenic development of PTE. However, the early time course of these changes is not fully elucidated. This study was designed to examine inflammation and neuropathology in the hippocampus in a rat fluid percussion injury (FPI) model of PTE. Cytokine analyses in the hippocampus, as well as astrocyte morphology were assessed to determine early inflammatory changes after TBI. To examine early seizure-promoting neuropathology, we performed stereological analysis of parvalbumin-labeled interneurons in the hippocampus at 24 hrs after TBI. The results demonstrate that FPI in rats results in early hippocampal inflammation, but not a loss of parvalbumin-labeled interneurons.

KEYWORDS

•    Fluid percussion injury
•    Cell tracing and counting
•    TBI
•    Multiplex assay
•    Astrocyte

CITATION

Mukherjee S, Bricker PC, Shapiro LA (2014) Alteration of Hippocampal Cytokines and Astrocyte Morphology Observed in Rats 24 Hour after Fluid Percussion Injury. J Neurol Disord Stroke 2(4): 1078.

ABBREVIATIONS

TBI: Traumatic Brain Injury; FPI: Fluid Percussion Injury; Controlled Cortical Impact (CCI); Chemokine (C-X-C Motif) Ligand (CXCL); Chemokine (C-C Motif) Ligand (CCL); Interleukin (IL)

INTRODUCTION

Post-traumatic epilepsy (PTE) can result from a Traumatic brain injury (TBI). In fact, TBI is one of the few known etiological factors contributing to the development of epilepsy. PTE accounts for approximately 5% of all epileptic cases and 20% of symptomatic epilepsies [1-3]. Although it is not known who will develop PTE after a TBI, increased injury severity is associated with an increased incidence of PTE [4-6]. However, even relatively mild TBIs can lead to the development of PTE [2].

Considering that there are approximately 1.7 million reported TBIs in the United States each year, understanding the mechanisms involved in the development of PTE is vital to provide optimal treatment. This is especially important because PTEs are amongst the most difficult types of epilepsy to treat, and are often resistant to first and second line anti-epileptic treatment regimens [7-9].

There are various neuropathological changes that may underlie the development of PTE. Among these, TBI-induced inflammation and neuronal plasticity in the hippocampus is frequently observed. Hilar interneuron loss and dysfunction was reported at 7 days after TBI in rodents [10] and humans [11-13]. TBI-induced dysfunction and/or loss of hippocampal interneurons may alter inhibition within the hippocampus, resulting in hyperexcitability and a pro-epileptogenic state [14,15]. Thus, determining the precise times when neuroplastic changes begin to affect this population of neurons is vital to the mechanistic understanding of TBI and the post-traumatic consequences and developing optimal treatments at the appropriate time points.

In addition to interneuron loss, mounting evidence shows that neuroinflammatory cascades are initiated in the hippocampus following TBI ([16,17], and these effects may not be entirely unrelated. For example, Raghupathi and colleagues observed increased IL-1β and TNFα in the hippocampus and cortex following TBI [18]. Moreover, others have reported elevated levels of IL1α, IL-6, IL1β and TNFα in the hippocampus following TBI [19] and such inflammation may contribute to neuronal dysfunction and/or loss. It has been suggested that blocking inflammation can improve post-traumatic outcomes [20]. For example, treatment with a

TNFα protein synthesis inhibitor prevents cognitive impairment after TBI [21]. Such a finding was supported in a clinical TBI study, where cohorts carrying the homozygous TNFα-308 single nucleotide polymorphism had significantly worse Glasgow outcomes Score [22]. Other studies also support the idea that some components of the neuroinflammatory response may be detrimental outcomes of TBI, because inhibiting neuroinflammation can improve outcome measures [19,23]. However, other studies that inhibited the action of particular cytokine(s) either failed, or provided limited protection from TBI [24,25]. Considering these somewhat conflicting data, it is likely that the positive and negative effects of the neuroinflammatory response are dependent on spatial and temporal release, of the cytokine milieu, as well as the interacting and opposing, postTBI cytokine milieu. Thus, understanding the full spatial and temporal extent of TBI-induced neuroinflammation is vital to understanding the mechanisms involved in increased seizure susceptibility, the development of PTE and treating the many symptomology associated with brain trauma.

Therefore, the present study was designed to determine the alterations to 23 different cytokines in the hippocampus at 24 hrs after a fluid percussion TBI. To further define the cellular components of the TBI-induced inflammatory response in the hippocampus, astrocyte morphology was assessed in the hilus. Astrocytosis occurs rapidly in the neocortex after an FPI [26], but the early effects of TBI on astrocytes in the hippocampus has not been defined in this model. Considering that astrogliosis can be indicative of neuronal degeneration, it is possible that early hippocampal inflammation is associated with hilar interneuron loss. The observation that hilar interneuron loss is implicated in the development of epilepsy in other injury models [27-29] suggests a potential avenue for neuroprotection after TBI. Thus, this study also examined the number of parvalbumin-expressing interneurons in the hilus at 24 hrs after a fluid percussion TBI.

MATERIALS AND METHODS

All experimental protocols were approved by the Institutional Animal Care Committee (IACUC) of Texas A&M University, Health Science Center and Scott &White hospital. Animals were housed singly after FPI with a 12-h light–dark cycle (light on 6:00 and light off 18:00). All animals had continuous access to food and water. Twenty male Sprague-Dawley rats (Charles River), 9-10 weeks of age and weighing between 275-310 g were used in this study for cytokine estimation in the hippocampus. Rats were randomly assigned to either the experimental group or the sham group, so that each group had 10 rats. For immunohistochemistry, we used an additional 12 male Sprague-Dawley rats that were the same ages and weights as those used for cytokine analysis.

Fluid Percussion Injury (FPI) Rats were initially anesthetized with 4% isoflurane and oxygen for anesthesia induction and later to 2% isoflurane for maintenance. Once under anesthesia, the heads of the animals were shaved. Strict sterile technique was maintained during surgical procedures. Animals were placed in a stereotaxic instrument (Stoelting, Illinois). A 2 mm hole was drilled, with dura intact, in the skull over the left parietal cortex at 2 mm posterior and 3 mm lateral from the bregma, as previously described, [10]. The animals were connected to the fluid percussion instrument (custom design and fabrication from the radiology department from VCU, Model 01-B) via the female Luer-Lok. A single 4–4.25 atm pressure pulse was delivered, with a duration of 18–20 ms, as measured by a digital Oscilloscope (Tenma model # 728395), connected to a signal transducer hooked in-line between the fluid percussion cylinder and the syringe 

Sham animals were anesthetized and manipulated in an identical manner, but no pressure pulse from the fluid percussion instrument was delivered to their brain. After 24 hrs, animals used for the cytokine assay (N = 10 sham and 10 FPI) were euthanized under deep anesthesia and hippocampal tissue was collected and flash frozen. For immunohistochemistry, animals (N = 6 sham and 6 FPI) were perfused first by normal saline and then with 4% paraformaldehyde under deep anesthesia. After removing the brain and post-fixing for 24 hrs, the tissue was cut into 50 µm sections using a vibratome (Pelco 102, Ted Pella INC., Redding, CA).

Multiplex assay

The assay was performed as previously described (Mukherjee et al, 2011). The following 23 different cytokines (See Table 1) were assayed: TNFα, CCL2, CCL3, CCL5, IL 1α, IL 1 β, IL 2, IL 4, IL 5, IL 6, IL10, IL12, IL 13, IL17, IL 18, IFNγ, interferon inducible protein 10 (IP 10), Growth-related oncogene (GRO KC), Leptin, Eotaxin, Granulocyte macrophage colony stimulating factor (GMCSF), granulocyte colony stimulating factor (GCSF), vascular endothelial growth factor (VEGF). Frozen tissue was homogenized, as previously described [30] and following the manufacturer’s instructions (Milliplex MAP kit, Millipore). 25 µl of undiluted homogenate was added to 25 µl of assay buffer. The samples were analyzed using the Bio-Rad LS200 (Bio-Rad Instruments). Assays were run in triplicate and analytes were normalized to total protein concentration, estimated using a Bradford assay.

Immunohistochemistry

For GFAP labeling of astrocytes, sections were reacted freefloating as previously described [15,16]. Briefly, sections were incubated overnight in GFAP antibody (1:1000; Sigma G9269), rinsed and incubated with biotinylated goat anti-rabbit IgG antibody (1:200; Vector Labs) for 1.5-2 h. Tissue was again rinsed and incubated in Vector Elite ABC (Vector Labs) for 1 hr, after which tissue was rinsed and reacted in DAB. For parvalbumin immunohistochemistry, the same protocol was used as above, except that the primary antibody was anti-parvalbumin (1:100; Santa Cruz Biotechnology, Inc., Santa Cruz, CA).

Tracing of astrocytes for Sholl analysis: Astrocytes were traced and reconstructed using the Neurolucida (Microbrightfield Inc. V. 6.0) system. After tracing the hilus, a grid was superimposed and random areas were selected. A higher magnification was then used to zoom in on this location and cells that were within this area were selected for tracing. Only cells located in the hilus and showing clear and relatively complete process in the X,Y and Z plane, were chosen for tracing. After tracing, a Sholl analysis was used to examine various parameters of cell morphology using concentric spheres of a specified distance. For the purposes of our study we chose spheres with 10 µm increment diameter (Figure 1). Within each concentric sphere, overall process length, amount of bifurcations, amount of process endings, and amount of intersections through that sphere are analyzed.

Schematic diagram illustrating the method of sholl analysis. Processes traced directly from cell body are considered first order process and those arising from the first order processes are second order process, and so on. Spheres with a diameter of 10 µm increments were chosen for this study. The total amount of bifurcations, process endings and intersections through each sphere are shown.

Figure 1: Schematic diagram illustrating the method of sholl analysis. Processes traced directly from cell body are considered first order process and those arising from the first order processes are second order process, and so on. Spheres with a diameter of 10 µm increments were chosen for this study. The total amount of bifurcations, process endings and intersections through each sphere are shown.

Stereological analysis of parvalbumin-labeled interneurons in the hippocampus: Sections of tissue were stained with parvalbumin to allow for stereological quantification of labeled interneurons using Streoinvestigator (microbrightfield inc. V. 6.0). As part of this technique, hippocampal volume was also stereologically estimated. We performed Stereological estimation in the: pyramidal layer of CA1 and CA3, as well as in the dentate gyrus granule cell layer and hilus. The infra- and suprapyramidal blades were quantified separately and we also provided combined interneuron data.

Data analyzed with T-test and 1 way ANOVA using SPSS software 9.0. A post-hoc Bonferroni multiple comparisons was used to compare the groups.

Table 1: Effect of FPI on the analyte concentration in the hippocampus.

Analytes Sham ipsi FPI ipsi Sham contra FPI contra
IFN γ: 1.9 ± 0.3 1.5 ± 0.4 2.2 ± 0.3 1.3 ± 0.1
IL 13: 8.3 ± 1.2 6.6 ± 1.5 6.9 ± 0.7 5.2 ± 0.4
IL 1α: 3.8 ± 0.5 6.8 ± 1.6 3.9 ± 0.4 3.2 ± 0.6
IL 2: 10.0 ± 1.5 7.9 ± 1.4 9.6 ± 0.9 7.4 ± 0.8
IL 4: 4.4 ± 0.7 3.3 ± 0.7 3.2 ± 0.4 2.5 ± 0.4
IL 5: 2.4 ± 0.3 2.0 ±0.3 1.8 ± 0.2 1.0 ± 0.1
TNF α: 0.8 ± 0.3 2.5 ± 0.6 0.8 ± 0.2 1.1 ± 0.3
GMCSF: 1.3 ± 0.2 1.0 ± 0.07 1.20 ± 0.15 0.5 ± 0.04
IP 10: 3.6 ± 0.6 2.7 ± 0.7 3.1 ± 0.4 2.1 ± 0.17
VEGF: 1.1 ± 0.3 2.2 ± 0.56 1.3 ± 0.17 0.7 ± 0.1
GCSF: 0.5 ± 0.09 0.6 ± 0.07 0.5 ± 0.06 0.3 ± 0.04
GRO KC: 37.5 ± 8.6 149.4 ± 40.1 20.9 ± 3.0 54.5 ± 17.1
IL 18: 76.9 ± 6.4 88.8 ± 10.4 74.1 ± 5.2 62.7 ± 5.3
CCL2: 12.2 ± 2.9 92.64 ± 26.7 21.1 ± 1.8 17.8 ± 2.7
CCL3 3.8 ± 2.1 17.4 ± 4.6 0.3 ± 0.05 6.6 ± 4.8
Leptiin: 15.2 ± 4.6 13.3 ± 3.0 12.4 ± 2.1 8.7 ± 0.8
IL 17: 31.5 ± 125 23.3 ± 6.2 30.7 ± 3.1 21.2 ± 2.1
IL 6: 18.8 ± 2.4 35.3 ± 12.9 21.1 ± 1.9 2.7 ± 0.3
CCL 5: 10.1 ± 0.9 11.4 ± 0.8 7.6 ± 1.1 6.8 ± 0.9
IL12: 26.3 ± 4.0 20.1 ± 4.3 21.8 ± 2.1 16.1 ± 1.4
IL 1β: 11.2 ± 1.3 13.3 ± 2.0 11.9 ± 0.6 9.2 ± 0.7
IL 10: 14.1 ± 2.3 11.7 ± 2.8 18.6 ± 2.6 10.6 ± 1.0
Eotaxin: 4.8 ± 0.6 3.5 ± 0.7 6.1 ± 0.6 4.7 ± 0.4

Analyte concentration is expressed as pico gram per microgram of hippocampal tissue. For each hemisphere, data was analyzed using Ttest with p<0.05.

RESULTS

Multiplex cytokine assay

The entirety of the results from the cytokine analysis is shown in Table 1 and Figure 2. In the ipsilateral hemisphere, the following cytokines were significantly increased at 24 hrs after FPI: GRO KC (P = 0.014), CCL2 (P = 0.008), CCL3 (P = 0.02), TNF-α (P = 0.01). In addition, a trend towards increase was observed for IL 1α (p = 0.09) and VEGF (p = 0.08) (Table 1; Figure 2 A,B). In the contralateral hemisphere, the results demonstrated a significant decrease in the following cytokines at 24 hrs after FPI: IL17 (P = 0.028), IL2 (P = 0.045), IL1? (P = 0.011), IL10 (P = 0.015), IFN? (P = 0.018), IL13 (P = 0.05), IL5 (P = 0.043), GMCSF (P = 0.0006), IP10 (P = 0.022), VEGF (P = 0.021), and GCSF (P = 0.005). Although not significant, a trend was observed where Eotaxin appeared to decrease (p = 0.06) and GRO KC appeared to increase (P = 0.069) (Figure 2 C and D, and Table 1).

Figure 2 Analysis of cytokine concentrations after sham or FPI in the ipsilateral cortex (A and B) and contralateral cortex (C and D) at 24 hrs after FPI. A number of cytokines were found to be significantly increased in the ipsilateral hippocampus, whereas in the contralateral hippocampi, there were numerous decreases in cytokine concentration. In addition, a trend toward significance was also observed for various cytokines in the ipsilateral and contralateral hippocampi. The raw data for this analysis can be found in table 1. Taken together, the data illustrate a complex inflammatory response in the hippocampus that greatly varies from one hemisphere to the other. * indicates significant alteration (p < 0.05). # indicates a trend of significant change (p < 0.09 but > 0.05).

Figure 2: Analysis of cytokine concentrations after sham or FPI in the ipsilateral cortex (A and B) and contralateral cortex (C and D) at 24 hrs after FPI. A number of cytokines were found to be significantly increased in the ipsilateral hippocampus, whereas in the contralateral hippocampi, there were numerous decreases in cytokine concentration. In addition, a trend toward significance was also observed for various cytokines in the ipsilateral and contralateral hippocampi. The raw data for this analysis can be found in table 1. Taken together, the data illustrate a complex inflammatory response in the hippocampus that greatly varies from one hemisphere to the other. * indicates significant alteration (p < 0.05).

# indicates a trend of significant change (p < 0.09 but > 0.05).

Astrocyte morphology

Sholl analysis revealed that at the 20 μm distance from the cell body, a significant increase in the length of ipsi and contra lateral processes of astrocytes from FPI rats was observed (P < .001 and respectively; Figure 3A).

Analysis of astrocyte morphology at 20 µm from the cell body (A) and 30 µm from the cell body (B) at 24 hour after FPI. In A, a significant increase is observed in the number of intersections and nodes at the 20 µm sphere, in the ipsilateral hippocampus following FPI. At this same distance from the cell body, bilateral increase in astrocyte process length was observed. The number of astrocytic endings was significantly increased in the contralateral hemisphere at this distance from the cell body. In B, at 30 µm from the cell body, no significant differences were observed for the total number of intersections or nodes. However, a significant increase in astrocyte length and endings was observed in the ipsilateral hippocampus at this distance from the cell body. Taken together, these data indicate bilateral astrocyte activation at 24 hrs after TBI that is more pronounced in the ipsilateral hippocampi.

Figure 3: Analysis of astrocyte morphology at 20 µm from the cell body (A) and 30 µm from the cell body . In A, a significant increase is observed in the number of intersections and nodes at the 20 µm sphere, in the ipsilateral hippocampus following FPI. At this same distance from the cell body, bilateral increase in astrocyte process length was observed. The number of astrocytic endings was significantly increased in the contralateral hemisphere at this distance from the cell body. In B, at 30 µm from the cell body, no significant differences were observed for the total number of intersections or nodes. However, a significant increase in astrocyte length and endings was observed in the ipsilateral hippocampus at this distance from the cell body. Taken together, these data indicate bilateral astrocyte activation at 24 hrs after TBI that is more pronounced in the ipsilateral hippocampi.

In addition, at this distance, astrocytes in the ipsilateral hippocampus had significantly more nodes (P < 0.03) and intersections (p < .002) compared to shams, whereas astrocytes in the contralateral hemisphere had significantly more endings compared to shams (P < .02). At the 30 µm distance, astrocytes from the ipsilateral cortex of FPI rats had significantly more endings (P < .002) and greater length (P < .005) compared to shams (Figure 3 B). The data show that the astrocytic processes from FPI animals have increased length, and branch points compared to sham rats, indicative of relatively rapid astrocyte activation in the hippocampus 24 hrs after FPI. Such morphological data is consistent with our data showing increased expression of many pro-inflammatory proteins at this time point and further demonstrate an early post-traumatic hippocampal inflammatory response.

Figure 3 Analysis of astrocyte morphology at 20 µm from the cell body (A) and 30 µm from the cell body (B) at 24 hour after FPI. In A, a significant increase is observed in the number of intersections and nodes at the 20 µm sphere, in the ipsilateral hippocampus following FPI. At this same distance from the cell body, bilateral increase in astrocyte process length was observed. The number of astrocytic endings was significantly increased in the contralateral hemisphere at this distance from the cell body. In B, at 30 µm from the cell body, no significant differences were observed for the total number of intersections or nodes. However, a significant increase in astrocyte length and endings was observed in the ipsilateral hippocampus at this distance from the cell body. Taken together, these data indicate bilateral astrocyte activation at 24 hrs after TBI that is more pronounced in the ipsilateral hippocampi.

Figure 3: Analysis of astrocyte morphology at 20 µm from the cell body  (B) at 24 hour after FPI. In A, a significant increase is observed in the number of intersections and nodes at the 20 µm sphere, in the ipsilateral hippocampus following FPI. At this same distance from the cell body, bilateral increase in astrocyte process length was observed. The number of astrocytic endings was significantly increased in the contralateral hemisphere at this distance from the cell body. In B, at 30 µm from the cell body, no significant differences were observed for the total number of intersections or nodes. However, a significant increase in astrocyte length and endings was observed in the ipsilateral hippocampus at this distance from the cell body. Taken together, these data indicate bilateral astrocyte activation at 24 hrs after TBI that is more pronounced in the ipsilateral hippocampi.

Parvalbumin-labeled interneurons quantification

Stereological analysis of parvalbumin-labeled interneurons revealed no significant differences in the ipsilateral or contralateral hippocampus following TBI, in any of the regions examined (Figure 4).

Figure 4: Sterological analysis of parvalbumin-labeled cells in the hippocampus. Several parameters are illustrated: number of parvalbuminlabeled cells (A), surface area or regions of interest (B) and volume or region of interest (C). No significant differences were observed for any of the parameters examined.

DISCUSSION

The results from the current study revealed an increase in several different cytokines in the ipsilateral hippocampus and a concomitant decrease in various cytokines in the contralateral hippocampus at 24 hrs after a FPI. In addition, the data revealed rapid post-traumatic astrocyte activation in hippocampal CA4 (hilus), that is more robust in the ipsilateral hemisphere at the 24 hr post-TBI time point. Despite the rapid appearance of molecular and anatomical inflammation in the hippocampus, no loss of parvalbumin-labeled interneurons in the hippocampus was observed at 24 hrs after FPI.

The findings from the current study showing a rapid inflammatory response in the archicortex, following an FPI with a neocortical focus, suggests that the TBI-induced neuroinflammatory response extends into the hippocampus within 24 hrs after injury. It is possible that early inflammation can promote a pro-epileptogenic environment because inflammation has been implicated in the development of PTE [31,32]. Moreover, previous studies using chemoconvusant-induced epilepsy have linked neuroinflammation to pro-epileptigenic changes in the hippocampus [33,34,28]. The hippocampal cytokine data from the current study are consistent with, and expand upon, previous studies. For example, Fan and colleagues observed increased IL-1β mRNA and TNFα mRNA in the hippocampus in rats at 6 hrs after FPI [35,36]. Another study showed acute elevation of IL1α, IL-6, IL-1β and TNF-α in the hippocampus of rats following TBI [19]. Therefore, several lines of evidence demonstrate a rapid increase in hippocampal cytokine expression following FPI. However, the present study expands on these data by providing analysis of many more cytokines and chemokines than those previously reported (Table 1) and adds the dynamic of bilateral astrocyte activation in the hippocampus that is more robust in the ipsilateral hippocampi.

Studies have examined the neuropathological and behavioral consequences of altering neuroinflammation after injury. Administration of IL18 binding protein, a specific inhibitor of IL18, promotes an improved neurological score on measures of alertness, motor and physiological behavior after TBI [37]. The administration of IL10, an anti-inflammatory cytokine, promotes enhanced motor outcome after FPI [38]. Studies manipulating TNFα have yielded mixed outcomes. One study showed that increased expression of 

TNFα exacerbates pathology after TBI [39], whereas other studies have observed a protective role for TNFα ?after TBI [40,41]. These latter studies demonstrated that TNFα and TNFαreceptor knockout mice have greater deficiencies after TBI [40,41]. These conflicting results suggest both pro- and antiinflammatory effects of TNFα [42] that are likely related to when, where and in what concentrations TNFα is expressed following injury. Therefore, the spatial and temporal gradient of this (and all) cytokine(s) after TBI needs further elucidation.

TNFα exacerbates pathology after TBI [39], whereas other studies have observed a protective role for TNFα ?after TBI [40,41]. These latter studies demonstrated that TNFα and TNFαreceptor knockout mice have greater deficiencies after TBI [40,41]. These conflicting results suggest both pro- and antiinflammatory effects of TNFα [42] that are likely related to when, where and in what concentrations TNFα is expressed following injury. Therefore, the spatial and temporal gradient of this (and all) cytokine(s) after TBI needs further elucidation.

Chemokine and cytokine alterations are not the only indices of the neuroinflammatory response following TBI. GFAP-labeled astrocyte activation and its related elevation of GFAP protein is also a hallmark of TBI. Studies in adults and children have found that injury severity predicted the extent of elevated levels GFAP in serum [44-47]. Considering that GFAP is expressed exclusively in astrocytes, astrocyte activation and dysfunction may mediate disease pathogenesis, including seizure disorders [48-52]. For example, ablation of astrocytes resulted in an increased tissue loss after controlled cortical impact (CCI) TBI [53]. Moreover, astrocyte activation in the hippocampus may contribute to a hyperexcitable circuit. Shapiro and Ribak [54], and Shapiro et al. [55], showed that following pilocarpine-induced seizures, newly born dentate granule cells extend hilar basal dendrites along the processes of hypertrophied GFAP-expressing astrocytes [54,55]. These hilar basal dendrites were targeted for aberrant synaptogenesis by mossy fibers, leading to the development of a so-called, “recurrent excitatory circuitry” [55,56]. Thus, there are several mechanisms whereby TBI-induced changes to hippocampal astrocytes may promote a pro-epileptogenic environment.

In a previous study, Mukherjee et al., showed the spatial and temporal astrocytic response to FPI in mouse cortex [26]. They showed that at 1 day after injury, GFAP-labeled astrocytes were migrating towards the injury site and that by 3 days after injury, a robust astrocyte reaction was observed that was concentrated in the peri-injury area. In the current study, the data show that as early as 1 day after FPI in the rat, astrocyte activation in the hippocampus is observed. Thus, it would seem as though the signal for astrocyte activation in cortex, including archicortex is signaled within 1 day of an FPI.

Considering that the pro-inflammatory proteins are more robustly increased in the ipsilateral hippocampus at 24 hrs after injury, it is not surprising that a more robust activated morphology was observed in the ipsilateral hippocampus relative to the contralateral hemisphere. Interestingly, a previous study reported a significant decrease in the number of astrocytes in the hippocampus at 24 h after FPI with minimal changes in morphology [57]. However, that study did not examine astrocytes in the hilus, which is where astrocytes were examined in the current study. Moreover, Zhao et al. [57] used a more lateral placement for their injury (4.8 mm diameter hole on the right parietal cortex at 4.5 mm behind and 3.0 mm lateral to the bregma) relative to the coordinates used in the current study, which could potentially influence the pattern of astrocytosis.

Astrocyte activation can be indicative of neuronal damage and loss [58- 60]. Interneuron loss in the hippocampus has been implicated in epileptogenesis in numerous models [61- 63]. Because interneuron loss is known to occur within 1 week following TBI [ 63-68], we hypothesized that the astrocyte activation observed in the hippocampus by 1 day post-TBI would be indicative of a loss of hilar interneurons. Such a finding has not been previously reported at such an early timepoint after TBI and evidence was lacking from the present study. In support of this null finding, Toth and colleagues [66] showed that Parvalbuminpositive interneurons are relatively resilient following TBI. Thus, it is possible that other populations of interneurons are effected following TBI. It is also possible that despite the fact that no changes to the number of Parvalbumin-labeled interneurons was observed in the current study, plasticity of their connectome cannot be ruled out with regards to the contributions of these cells to hyper-excitability following TBI [69]. It is also possible that, as has been previously reported, this population of hilar interneurons become affected at later timepoints after FPI. More studies are needed to better define the timing of interneuron loss and plasticity in different models of TBI.

CONCLUSION

This study demonstrates a rapid inflammatory and astrocytic response in the ispi and contralateral hippocampus at 24 hrs after FPI in the rat, but no change in parvalbumin-expressing neuronal numbers. It is possible that inflammation, as well as neuroplasticity to hippocampal interneurons may contribute to subsequent pro-epileptogenic alterations to hippocampal circuitry function following TBI. Future studies are needed to fully define the epileptogenic contributions of the inflammatory and neuropathological changes in the hippocampus following TBI.

ACKNOWLEDGEMENT

We are grateful for the support from Scott &White Hospital (RGP# 90347). We are thankful for the technical assistance and support from Megan Ruch. This material is the result of work supported with resources and the use of facilities at the Central Texas Veterans Health Care System, Temple, Texas.

REFERENCES

1. Agrawal A, Timothy J, Pandit L, Manju M. Post-traumatic epilepsy: an overview. Clin Neurol Neurosurg. 2006; 108: 433-439.

2. Temkin NR. Preventing and treating posttraumatic seizures: the human experience. Epilepsia. 2009; 50 Suppl 2: 10-13.

3. Christensen J. Traumatic brain injury: risks of epilepsy and implications for medicolegal assessment. Epilepsia. 2012; 53 Suppl 4: 43-47.

4. Weiss GH, Feeney DM, Caveness WF, Dillon D, Kistler JP, Mohr JP, et al. Prognostic factors for the occurrence of posttraumatic epilepsy. Arch Neurol. 1983; 40: 7-10.

5. Weiss GH, Salazar AM, Vance SC, Grafman JH, Jabbari B. Predicting posttraumatic epilepsy in penetrating head injury. Arch Neurol. 1986; 43: 771-773.

6. Curia G, Levitt M, Fender JS, Miller JW, Ojemann J, D’Ambrosio R. Impact of injury location and severity on posttraumatic epilepsy in the rat: role of frontal neocortex. Cereb Cortex. 2011; 21: 1574-1592.

7. Hitiris N, Mohanraj R, Norrie J, Sills GJ, Brodie MJ. Predictors of pharmacoresistant epilepsy. Epilepsy Res. 2007; 75: 192-196.

8. Schneiderman JH. Monotherapy versus polytherapy in epilepsy: a framework for patient management. Can J Neurol Sci. 1998; 25: S9-13.

9. Brodie MJ, Dichter MA. Antiepileptic drugs. N Engl J Med. 1996; 334: 168-175.

10. Zhang BL, Chen X, Tan T, Yang Z, Carlos D, Jiang RC, et al. Traumatic brain injury impairs synaptic plasticity in hippocampus in rats. Chin Med J (Engl). 2011; 124: 740-745.

11. Golarai G, Greenwood AC, Feeney DM, Connor JA. Physiological and structural evidence for hippocampal involvement in persistent seizure susceptibility after traumatic brain injury. J Neurosci. 2001; 21: 8523-8537.

12. Swartz BE, Houser CR, Tomiyasu U, Walsh GO, DeSalles A, Rich JR, et al. Hippocampal cell loss in posttraumatic human epilepsy. Epilepsia. 2006; 47: 1373-1382.

13. Buriticá E, Villamil L, Guzmán F, Escobar MI, García-Cairasco N, Pimienta HJ. Changes in calcium-binding protein expression in human cortical contusion tissue. J Neurotrauma. 2009; 26: 2145-2155.

14. Dudek FE, Spitz M. Hypothetical mechanisms for the cellular and neurophysiologic basis of secondary epileptogenesis: proposed role of synaptic reorganization. J Clin Neurophysiol. 1997; 14: 90-101.

15. Dudek FE, Sutula TP. Epileptogenesis in the dentate gyrus: a critical perspective. Prog Brain Res. 2007; 163: 755-773.

16. Ziebell JM, Morganti-Kossmann MC. Involvement of pro- and anti-inflammatory cytokines and chemokines in the pathophysiology of traumatic brain injury. Neurotherapeutics. 2010; 7: 22-30.

17. Frugier T, Morganti-Kossmann MC, O’Reilly D, McLean CA. In situ detection of inflammatory mediators in post mortem human brain tissue after traumatic injury. J Neurotrauma. 2010; 27: 497-507.

18. Raghupathi R, McIntosh TK, Smith DH. Cellular responses to experimental brain injury. Brain Pathol. 1995; 5: 437-442.

19. Harting MT, Jimenez F, Adams SD, Mercer DW, Cox CS Jr. Acute, regional inflammatory response after traumatic brain injury: Implications for cellular therapy. Surgery. 2008; 144: 803-813.

20. Vezzani A, French J, Bartfai T, Baram TZ. The role of inflammation in epilepsy. Nat Rev Neurol. 2011; 7: 31-40.

21. Belarbi K, Jopson T, Tweedie D, Arellano C, Luo W, Greig NH, et al. TNF-α protein synthesis inhibitor restores neuronal function and reverses cognitive deficits induced by chronic neuroinflammation. J Neuroinflammation. 2012; 9: 23.

22. Waters RJ, Murray GD, Teasdale GM, Stewart J, Day I, Lee RJ, et al. Cytokine gene polymorphisms and outcome after traumatic brain injury. J Neurotrauma. 2013; 30: 1710-1716.

23. Chen G, Shi J, Hu Z, Hang C. Inhibitory Effect on Cerebral Inflammatory Response following Traumatic Brain Injury in Rats: A Potential Neuroprotective Mechanism of N-Acetylcysteine. Mediators Inflamm. 2008; 2008: 716458.

24. Knoblach SM, Faden AI. Cortical interleukin-1 beta elevation after traumatic brain injury in the rat: no effect of two selective antagonists on motor recovery. Neurosci Lett. 2000; 289: 5-8.

25. Marklund N, Keck C, Hoover R, Soltesz K, Millard M, LeBold D, et al. Administration of monoclonal antibodies neutralizing the inflammatory mediators tumor necrosis factor alpha and interleukin -6 does not attenuate acute behavioral deficits following experimental traumatic brain injury in the rat. Restor Neurol Neurosci. 2005; 23: 31-42.

26. Mukherjee S, Zeitouni S, Cavarsan CF, Shapiro LA. Increased seizure susceptibility in mice 30 days after fluid percussion injury. Front Neurol. 2013; 4: 28.

27. Sun C, Mtchedlishvili Z, Bertram EH, Erisir A, Kapur J. Selective Loss of Dentate Hilar Interneurons Contributes to Reduced Synaptic Inhibition of Granule Cells in an Electrical Stimulation- Based Animal Model of Temporal Lobe Epilepsy. J Comp Neurol. 2007; 500: 876– 893.

28. Somera-Molina KC, Nair S, Van Eldik LJ, Watterson DM, Wainwright MS. Enhanced microglial activation and proinflammatory cytokine Central Shapiro et al. (2014) Email: J Neurol Disord Stroke 2(3): 1058 (2014) 8/9 upregulation are linked to increased susceptibility to seizures and neurologic injury in a ‘two-hit’ seizure model, Brain Res. 2009; 1282: 162–172.

29. Gao F, Liu Y, Li X, Wang Y, Wei D, Jiang W. Fingolimod (FTY720) inhibits neuroinflammation and attenuates spontaneous convulsions in lithium-pilocarpine induced status epilepticus in rat model. Pharmacol Biochem Behav. 2012; 103: 187-196.

30. Mukherjee S, Katki K, Arisi GM, Foresti ML, Shapiro LA. Early TBIInduced Cytokine Alterations are Similarly Detected by Two Distinct Methods of Multiplex Assay. Front Mol Neurosci. 2011; 4: 21.

31. Lloyd E, Somera-Molina K, VanEldik LJ, Watterson DM, Wain-wright MS. Suppression of acute proinflammatory cytokine and chemokine upregulation by post-injury administration of a novel small molecule improves long-term neuro-logic outcome in a mouse model of traumatic brain injury. J Neuroin-flammation. 2008; 5: 28.

32. Chraszcz M, Venkatesan C, Dragisic T, Watterson DM, and Wainwright MS. Minozac treatment prevents increased seizure susceptibility in a mouse “two-hit” model of closed skull traumatic brain injury and electroconvulsive shock-induced seizures. J Neurotrauma. 2010; 27: 1283–1295.

33. Galic MA, Riazi K, Heida JG, Mouihate A, Fournier NM, Spencer SJ, et al. Postnatal inflammation increases seizure susceptibility in adult rats. J Neurosci. 2008; 28: 6904-6913.

34. Galic MA, Riazi K, Henderson AK, Tsutsui S, Pittman QJ. Viral-like brain inflammation during development causes increased seizure susceptibility in adult rats. Neurobiol Dis. 2009; 36: 343-351.

35. Fan L, Young PR, Barone FC, Feuerstein GZ, Smith DH, McIntosh TK. Experimental brain injury induces expression of interleukin-1 beta mRNA in the rat brain. Brain Res Mol Brain Res. 1995; 30: 125-130.

36. Fan L, Young PR, Barone FC, Feuerstein GZ, Smith DH, McIntosh TK. Experimental brain injury induces differential expression of tumor necrosis factor-alpha mRNA in the CNS. Brain Res Mol Brain Res. 1996; 36: 287-291.

37. Yatsiv I, Morganti-Kossmann MC, Perez D, Dinarello CA, Novick D, Rubinstein M, et al. Elevated Intracranial IL-18 in Humans and Mice After Traumatic Brain Injury and Evidence of Neuroprotective Effects of IL-18–Binding Protein After Experimental Closed Head Injury. Journal of Cerebral Blood Flow & Metabolism. 2002; 22: 971–978.

38. Knoblach SM, Faden AI. Interleukin-10 improves outcome and alters proinflammatory cytokine expression after experimental traumatic brain injury. Exp Neurol. 1998; 153: 143–151.

39. Ramilo O, Sáez-Llorens X, Mertsola J, Jafari H, Olsen KD, Hansen EJ, et al. Tumor necrosis factor alpha/cachectin and interleukin 1 beta initiate meningeal inflammation. J Exp Med. 1990; 172: 497-507.

40. Scherbel U, Raghupathi R, Nakamura M, Saatman KE, Trojanowski JQ, Neugebauer E, et al. Differential acute and chronic responses of tumor necrosis factor-deficient mice to experimental brain injury. Proc Natl Acad Sci U S A. 1999; 96: 8721-8726.

41. Sullivan PG, Bruce-Keller AJ, Rabchevsky AG, Christakos S, Clair DK, Mattson MP, et al. Exacerbation of damage and altered NF-kappaB activation in mice lacking tumor necrosis factor receptors after traumatic brain injury. J Neurosci.1999; 19: 6248–6256.

42. Shohami E, Ginis I, Hallenbeck JM. Dual role of tumor necrosis factor alpha in brain injury. Cytokine Growth Factor Rev. 1999; 10: 119-130.

43. Semple BD, Bye N, Rancan M, Ziebell JM, Morganti-Kossmann MC. Role of CCL2 (MCP-1) in traumatic brain injury (TBI): evidence from severe TBI patients and CCL2-/- mice. J Cereb Blood Flow Metab. 2010; 30: 769-782.

 44. Pelinka LE, Kroepfl A, Leixnering M, Buchinger W, Raabe A, Redl H. GFAP versus S100B in serum after traumatic brain injury: relationship to brain damage and outcome. J Neurotrauma. 2004; 21: 1553-1561.

45. Lumpkins KM, Bochicchio GV, Keledjian K, Simard JM, McCunn M, Scalea T. Glial fibrillary acidic protein is highly correlated with brain injury. J Trauma. 2008; 65: 778-782. 46.Zurek J, Fedora M. Dynamics of glial fibrillary acidic protein during traumatic brain injury in children. J Trauma. 2011; 71: 854-859.

47. Fraser DD, Close TE, Rose KL, Ward R, Mehl M, Farrell C, et al. Severe traumatic brain injury in children elevates glial fibrillary acidic protein in cerebrospinal fluid and serum. Pediatr Crit Care Med. 2011; 12: 319-324.

48. Shapiro LA, Wang L, Ribak CE. Rapid astrocyte and microglial activation following pilocarpine-induced seizures in rats. Epilepsia. 2008; 49 Suppl 2: 33-41.

49. Das A, Wallace GC 4th, Holmes C, McDowell ML, Smith JA, Marshall JD, Bonilha L. Hippocampal tissue of patients with refractory temporal lobe epilepsy is associated with astrocyte activation, inflammation, and altered expression of channels and receptors. Neuroscience. 2012; 220: 237-246.

50. Pekny M, Nilsson M. Astrocyte activation and reactive gliosis. Glia. 2005; 50: 427-434.

51. Foresti ML, Arisi GM, Katki K, Montañez A, Sanchez RM, Shapiro LA,. Chemokine CCL2 and its receptor CCR2 are increased in the hippocampus following pilocarpine-induced status epilepticus. J Neuroinflammation. 2009; 6: 40.

52. Steinhäuser C, Seifert G. Astrocyte dysfunction in epilepsy. Jaspers Basic Mechanims of Epilepsy. Glia Special Issue: Astrocytes and Epilepsy, 4th edn. 2012; 60: 1191.

53. Myer DJ, Gurkoff GG, Lee SM, Hovda DA, Sofroniew MV. Essential protective roles of reactive astrocytes in traumatic brain injury. Brain. 2006; 129: 2761-2772.

54. Shapiro LA, Ribak CE. Newly born dentate granule neurons after pilocarpine-induced epilepsy have hilar basal dendrites with immature synapses. Epilepsy Res. 2006; 69: 53-66.

55. Shapiro LA, Wang L, Ribak CE. Rapid astrocyte and microglial activation following pilocarpine-induced seizures in rats. Epilepsia. 2008; 49 Suppl 2: 33-41.

56. Dashtipour K, Tran PH, Okazaki MM, Nadler JV, Ribak CE. Ultrastructural features and synaptic connections of hilar ectopic granule cells in the rat dentate gyrus are different from those of granule cells in the granule cell layer. Brain Res. 2001; 890: 261-71.

57. Zhao X, Ahram A, Berman RF, Muizelaar JP, Lyeth BG. Early loss of astrocytes after experimental traumatic brain injury. Glia. 2003; 44: 140-152.

58. Zhou BY, Liu Y, Kim Bo, Xiao Y, He JJ. Astrocyte activation and dysfunction and neuron death by HIV-1 Tat expression in astrocytes. Mol Cell Neurosci. 2004; 27: 296-305.

59. Deshpande M, Zheng J, Borgmann K, Persidsky R, Wu L, Schellpeper C, Ghorpade A. Role of activated astrocytes in neuronal damage: potential links to HIV-1-associated dementia. Neurotox Res. 2005; 7: 183-192.

60. Ouyang YB, Voloboueva LA, Xu LJ, Giffard RG. Selective dysfunction of hippocampal CA1 astrocytes contributes to delayed neuronal damage after transient forebrain ischemia. J Neurosci. 2007; 27: 4253-4260.

61. Sloviter RS. Decreased hippocampal inhibition and a selective loss of interneurons in experimental epilepsy. Science. 1987; 235: 73-76.

62. Kuruba R, Hattiangady B, Parihar VK, Shuai B, Shetty AK. Differential Susceptibility of Interneurons Expressing Neuropeptide Y or Parvalbumin in the Aged Hippocampus to Acute Seizure Activity (Aging, Seizures and Interneuron Loss). PLoS ONE. 2011; 6: e24493.

63. Pavlov I, Huusko N, Drexel M, Kirchmair E, Sperk G, Pitkänen A, et al. Progressive loss of phasic, but not tonic, GABAA receptor-mediated inhibition in dentate granule cells in a model of post-traumatic epilepsy in rats. Neuroscience. 2011; 194: 208-219.

64. Hunt RF, Haselhorst LA, Schoch KM, Bach EC, Rios-Pilier J, Scheff SW, et al. Posttraumatic mossy fiber sprouting is related to the degree of cortical damage in three mouse strains. Epilepsy Res. 2012; 99: 167- 170.

65. Lowenstein DH, Thomas MJ, Smith DH, McIntosh TK. 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.

66. Toth Z, Hollrigel GS, Gorcs T, Soltesz I. Instantaneous perturbation of dentate interneuronal networks by a pressure wave-transient delivered to the neocortex. J Neurosci. 1997; 17: 8106-8117.

67. Ross ST, Soltesz I. Selective depolarization of interneurons in the early posttraumatic dentate gyrus: involvement of the Na(+)/K(+)-ATPase. J Neurophysiol. 2000; 83: 2916-2930.

68. Santhakumar V, Bender R, Frotscher M, Ross ST, Hollrigel GS, Toth Z, Soltesz I. Granule cell hyperexcitability in the early post-traumatic rat dentate gyrus: the ‘irritable mossy cell’ hypothesis. J Physiol. 2000; 524 Pt 1: 117-134.

69. Gupta A, Elgammal FS, Proddutur A, Shah S, Santhakumar V. Decrease in tonic inhibition contributes to increase in dentate semilunar granule cell excitability after brain injury. J Neurosci. 2012; 32: 2523-2537.

 

Mukherjee S, Bricker PC, Shapiro LA (2014) Alteration of Hippocampal Cytokines and Astrocyte Morphology Observed in Rats 24 Hour after Fluid Percussion Injury. J Neurol Disord Stroke 2(3): 1058.

Received : 02 Apr 2014
Accepted : 17 Sep 2014
Published : 19 Sep 2014
Journals
Annals of Otolaryngology and Rhinology
ISSN : 2379-948X
Launched : 2014
JSM Schizophrenia
Launched : 2016
Journal of Nausea
Launched : 2020
JSM Internal Medicine
Launched : 2016
JSM Hepatitis
Launched : 2016
JSM Oro Facial Surgeries
ISSN : 2578-3211
Launched : 2016
Journal of Human Nutrition and Food Science
ISSN : 2333-6706
Launched : 2013
JSM Regenerative Medicine and Bioengineering
ISSN : 2379-0490
Launched : 2013
JSM Spine
ISSN : 2578-3181
Launched : 2016
Archives of Palliative Care
ISSN : 2573-1165
Launched : 2016
JSM Nutritional Disorders
ISSN : 2578-3203
Launched : 2017
Annals of Neurodegenerative Disorders
ISSN : 2476-2032
Launched : 2016
Journal of Fever
ISSN : 2641-7782
Launched : 2017
JSM Bone Marrow Research
ISSN : 2578-3351
Launched : 2016
JSM Mathematics and Statistics
ISSN : 2578-3173
Launched : 2014
Journal of Autoimmunity and Research
ISSN : 2573-1173
Launched : 2014
JSM Arthritis
ISSN : 2475-9155
Launched : 2016
JSM Head and Neck Cancer-Cases and Reviews
ISSN : 2573-1610
Launched : 2016
JSM General Surgery Cases and Images
ISSN : 2573-1564
Launched : 2016
JSM Anatomy and Physiology
ISSN : 2573-1262
Launched : 2016
JSM Dental Surgery
ISSN : 2573-1548
Launched : 2016
Annals of Emergency Surgery
ISSN : 2573-1017
Launched : 2016
Annals of Mens Health and Wellness
ISSN : 2641-7707
Launched : 2017
Journal of Preventive Medicine and Health Care
ISSN : 2576-0084
Launched : 2018
Journal of Chronic Diseases and Management
ISSN : 2573-1300
Launched : 2016
Annals of Vaccines and Immunization
ISSN : 2378-9379
Launched : 2014
JSM Heart Surgery Cases and Images
ISSN : 2578-3157
Launched : 2016
Annals of Reproductive Medicine and Treatment
ISSN : 2573-1092
Launched : 2016
JSM Brain Science
ISSN : 2573-1289
Launched : 2016
JSM Biomarkers
ISSN : 2578-3815
Launched : 2014
JSM Biology
ISSN : 2475-9392
Launched : 2016
Archives of Stem Cell and Research
ISSN : 2578-3580
Launched : 2014
Annals of Clinical and Medical Microbiology
ISSN : 2578-3629
Launched : 2014
JSM Pediatric Surgery
ISSN : 2578-3149
Launched : 2017
Journal of Memory Disorder and Rehabilitation
ISSN : 2578-319X
Launched : 2016
JSM Tropical Medicine and Research
ISSN : 2578-3165
Launched : 2016
JSM Head and Face Medicine
ISSN : 2578-3793
Launched : 2016
JSM Cardiothoracic Surgery
ISSN : 2573-1297
Launched : 2016
JSM Bone and Joint Diseases
ISSN : 2578-3351
Launched : 2017
JSM Bioavailability and Bioequivalence
ISSN : 2641-7812
Launched : 2017
JSM Atherosclerosis
ISSN : 2573-1270
Launched : 2016
Journal of Genitourinary Disorders
ISSN : 2641-7790
Launched : 2017
Journal of Fractures and Sprains
ISSN : 2578-3831
Launched : 2016
Journal of Autism and Epilepsy
ISSN : 2641-7774
Launched : 2016
Annals of Marine Biology and Research
ISSN : 2573-105X
Launched : 2014
JSM Health Education & Primary Health Care
ISSN : 2578-3777
Launched : 2016
JSM Communication Disorders
ISSN : 2578-3807
Launched : 2016
Annals of Musculoskeletal Disorders
ISSN : 2578-3599
Launched : 2016
Annals of Virology and Research
ISSN : 2573-1122
Launched : 2014
JSM Renal Medicine
ISSN : 2573-1637
Launched : 2016
Journal of Muscle Health
ISSN : 2578-3823
Launched : 2016
JSM Genetics and Genomics
ISSN : 2334-1823
Launched : 2013
JSM Anxiety and Depression
ISSN : 2475-9139
Launched : 2016
Clinical Journal of Heart Diseases
ISSN : 2641-7766
Launched : 2016
Annals of Medicinal Chemistry and Research
ISSN : 2378-9336
Launched : 2014
JSM Pain and Management
ISSN : 2578-3378
Launched : 2016
JSM Women's Health
ISSN : 2578-3696
Launched : 2016
Clinical Research in HIV or AIDS
ISSN : 2374-0094
Launched : 2013
Journal of Endocrinology, Diabetes and Obesity
ISSN : 2333-6692
Launched : 2013
Journal of Substance Abuse and Alcoholism
ISSN : 2373-9363
Launched : 2013
JSM Neurosurgery and Spine
ISSN : 2373-9479
Launched : 2013
Journal of Liver and Clinical Research
ISSN : 2379-0830
Launched : 2014
Journal of Drug Design and Research
ISSN : 2379-089X
Launched : 2014
JSM Clinical Oncology and Research
ISSN : 2373-938X
Launched : 2013
JSM Bioinformatics, Genomics and Proteomics
ISSN : 2576-1102
Launched : 2014
JSM Chemistry
ISSN : 2334-1831
Launched : 2013
Journal of Trauma and Care
ISSN : 2573-1246
Launched : 2014
JSM Surgical Oncology and Research
ISSN : 2578-3688
Launched : 2016
Annals of Food Processing and Preservation
ISSN : 2573-1033
Launched : 2016
Journal of Radiology and Radiation Therapy
ISSN : 2333-7095
Launched : 2013
JSM Physical Medicine and Rehabilitation
ISSN : 2578-3572
Launched : 2016
Annals of Clinical Pathology
ISSN : 2373-9282
Launched : 2013
Annals of Cardiovascular Diseases
ISSN : 2641-7731
Launched : 2016
Journal of Behavior
ISSN : 2576-0076
Launched : 2016
Annals of Clinical and Experimental Metabolism
ISSN : 2572-2492
Launched : 2016
Clinical Research in Infectious Diseases
ISSN : 2379-0636
Launched : 2013
JSM Microbiology
ISSN : 2333-6455
Launched : 2013
Journal of Urology and Research
ISSN : 2379-951X
Launched : 2014
Journal of Family Medicine and Community Health
ISSN : 2379-0547
Launched : 2013
Annals of Pregnancy and Care
ISSN : 2578-336X
Launched : 2017
JSM Cell and Developmental Biology
ISSN : 2379-061X
Launched : 2013
Annals of Aquaculture and Research
ISSN : 2379-0881
Launched : 2014
Clinical Research in Pulmonology
ISSN : 2333-6625
Launched : 2013
Journal of Immunology and Clinical Research
ISSN : 2333-6714
Launched : 2013
Annals of Forensic Research and Analysis
ISSN : 2378-9476
Launched : 2014
JSM Biochemistry and Molecular Biology
ISSN : 2333-7109
Launched : 2013
Annals of Breast Cancer Research
ISSN : 2641-7685
Launched : 2016
Annals of Gerontology and Geriatric Research
ISSN : 2378-9409
Launched : 2014
Journal of Sleep Medicine and Disorders
ISSN : 2379-0822
Launched : 2014
JSM Burns and Trauma
ISSN : 2475-9406
Launched : 2016
Chemical Engineering and Process Techniques
ISSN : 2333-6633
Launched : 2013
Annals of Clinical Cytology and Pathology
ISSN : 2475-9430
Launched : 2014
JSM Allergy and Asthma
ISSN : 2573-1254
Launched : 2016
Annals of Sports Medicine and Research
ISSN : 2379-0571
Launched : 2014
JSM Sexual Medicine
ISSN : 2578-3718
Launched : 2016
Annals of Vascular Medicine and Research
ISSN : 2378-9344
Launched : 2014
JSM Biotechnology and Biomedical Engineering
ISSN : 2333-7117
Launched : 2013
Journal of Hematology and Transfusion
ISSN : 2333-6684
Launched : 2013
JSM Environmental Science and Ecology
ISSN : 2333-7141
Launched : 2013
Journal of Cardiology and Clinical Research
ISSN : 2333-6676
Launched : 2013
JSM Nanotechnology and Nanomedicine
ISSN : 2334-1815
Launched : 2013
Journal of Ear, Nose and Throat Disorders
ISSN : 2475-9473
Launched : 2016
JSM Ophthalmology
ISSN : 2333-6447
Launched : 2013
Journal of Pharmacology and Clinical Toxicology
ISSN : 2333-7079
Launched : 2013
Annals of Psychiatry and Mental Health
ISSN : 2374-0124
Launched : 2013
Medical Journal of Obstetrics and Gynecology
ISSN : 2333-6439
Launched : 2013
Annals of Pediatrics and Child Health
ISSN : 2373-9312
Launched : 2013
JSM Clinical Pharmaceutics
ISSN : 2379-9498
Launched : 2014
JSM Foot and Ankle
ISSN : 2475-9112
Launched : 2016
JSM Alzheimer's Disease and Related Dementia
ISSN : 2378-9565
Launched : 2014
Journal of Addiction Medicine and Therapy
ISSN : 2333-665X
Launched : 2013
Journal of Veterinary Medicine and Research
ISSN : 2378-931X
Launched : 2013
Annals of Public Health and Research
ISSN : 2378-9328
Launched : 2014
Annals of Orthopedics and Rheumatology
ISSN : 2373-9290
Launched : 2013
Journal of Clinical Nephrology and Research
ISSN : 2379-0652
Launched : 2014
Annals of Community Medicine and Practice
ISSN : 2475-9465
Launched : 2014
Annals of Biometrics and Biostatistics
ISSN : 2374-0116
Launched : 2013
JSM Clinical Case Reports
ISSN : 2373-9819
Launched : 2013
Journal of Cancer Biology and Research
ISSN : 2373-9436
Launched : 2013
Journal of Surgery and Transplantation Science
ISSN : 2379-0911
Launched : 2013
Journal of Dermatology and Clinical Research
ISSN : 2373-9371
Launched : 2013
JSM Gastroenterology and Hepatology
ISSN : 2373-9487
Launched : 2013
Annals of Nursing and Practice
ISSN : 2379-9501
Launched : 2014
JSM Dentistry
ISSN : 2333-7133
Launched : 2013
Author Information X