• No results found

Original Article Low molecular weight heparin prevents lipopolysaccharide induced-hippocampus-dependent cognitive impairments in mice

N/A
N/A
Protected

Academic year: 2020

Share "Original Article Low molecular weight heparin prevents lipopolysaccharide induced-hippocampus-dependent cognitive impairments in mice"

Copied!
11
0
0

Loading.... (view fulltext now)

Full text

(1)

Original Article

Low molecular weight heparin prevents

lipopolysaccharide induced-hippocampus-dependent

cognitive impairments in mice

Renqi Li, Jianhua Tong, Yuanhui Tan, Sihai Zhu, Jianjun Yang, Muhuo Ji

Department of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China Received June 12, 2015; Accepted July 24, 2015; Epub August 1, 2015; Published August 15, 2015

Abstract: Sepsis-associated encephalopathy (SAE) is a common complication after sepsis development, which is associated with the poor prognosis. However, no effective agent is currently available to treat this complication. The objective of the present study was to investigate whether low-molecular-weight heparin (LMWH) has protective effects against sepsis-induced cognitive impairments. Male mice were randomly divided into the control + vehicle, control + LMWH, lipopolysaccharide (LPS) + vehicle, or LPS + LMWH group. LMWH was administrated 30 min after the LPS administration (5 mg/kg) and daily afterward for 2 days. The survival rate was estimated by the Kaplan-Meier method. Behavioral tests were performed by open field and fear conditioning tests at day 7 after LPS admin-istration. The levels of tumor necrosis factor alpha, interleukin (IL)-1β, IL-6, IL-10, malondialdehyde, and superoxide dismutase, Toll-like receptor 4, nuclear factor kappa B p65, inducible nitric oxide synthase, cyclooxygenase-2, oc-cluding, high mobility group box-1, brain derived neurotrophic factor, and IBA1 positive cells were assessed at the indicated time points. LMWH attenuated LPS-induced hippocampus-dependent cognitive impairments, which was accompanied by decreased hippocampal IL-1β, malondialdehyde, Toll-like receptor 4, nuclear factor kappa B p65, inducible nitric oxide synthase, cyclooxygenase-2, high mobility group box-1 protein, and IBA1 positive cells, and increased occluding and brain derived neurotrophic factor levels. In conclusion, LMWH treatment protects against sepsis-induced cognitive impairments by attenuating hippocampal microglial activation, cytokine and oxidative stress production, disruption of blood-brain barrier, and the loss of synaptic plasticity related proteins.

Keywords: Sepsis, cognitive impairments, hippocampus, LMWH

Introduction

Sepsis-associated encephalopathy (SAE) is a well described complication in critical ill patients, which is associated with increased mortality and poor prognosis [1-4]. SAE repre-sents one brain dysfunction caused by sys- temic inflammation evoked by the immune response to bacterial lipopolysaccharide (LPS) or other endotoxic bacterial cell wall compo-nents in the absence of direct brain infection [1-3]. The leading mechanisms thought to explain the pathogenesis of this acute brain dysfunction, such as inflammation, abnormal cerebral blood flow, and increased blood-brain barrier permeability, each involve the endothe-lium, a dynamic component of the blood-brain barrier (BBB) with vasomotor and biochemical properties [5]. These pathological processes are thought to arise from cytokine-mediated

effects, either directly or indirectly by acting on the microvasculature, coagulation system, or the organs themselves [4, 5]. Therefore, drug that targets at some of these pathological pro-cesses may represent one option to treat sep-sis-induced cognitive impairments.

(2)

Surviving Sepsis Campaign guidelines [9]. In particular, accumulating evidence has suggest-ed that, apart from their anticoagulant capaci-ty, LMWH can regulate a wide array of inflam-matory diseases such as sepsis [10-12]. These findings have led to the suggestion that LMWH may decrease inflammatory responses and preserve the integrity of endothelium, which consequently improves LPS-induced cognitive impairments.

In the present study, we tested whether LMWH administration has protective effects against LPS-induced cognitive impairments. Since the hippocampus plays a key role in cognitive func-tion and is vulnerable to inflammafunc-tion, we test-ed whether LMWH administration would help modulate inflammation, preserve levels of neu-rotrophic regulatory factors, and attenuate cog-nitive impairments in an animal model of SAE. Materials and methods

Animals

Two hundred and sixty C57BL/6 male mice (3-4 months, 25-32 g) were purchased from the Animal Center of Jinling Hospital, Nanjing, China. The study protocol was approved by the Ethics Committee of Jinling Hospital, Nanjing University and all procedures were performed in accordance with the Guideline for the Care and Use of Laboratory Animals from the National Institutes of Health, USA. The animals were housed under a 12-h light/dark cycle in a temperature-controlled room at 24 ± 1°C with free access to food and water.

Experimental protocol

LPS derived from Escherichia Coli endotoxin (0111:B5, Sigma, CA, USA, 5 mg/kg) was dis-solved in normal saline and injected intraperito-neally as described previously [13]. Control ani-mals were injected with equivalent volumes (0.1 ml) of normal saline. The animals were ran-domly divided into the following four groups: Control + vehicle (normal saline) group; Control + LMWH group; LPS + vehicle group; or LPS + LMWH group. The flow chart of the experimen-tal protocol is presented in Figure 1A.

Drug administration

For the dose-response study, LMWH (Sigma, St. Louis, MO, USA) at 100 IU/kg, 200 IU/kg, or

400 IU/kg was given subcutaneously 30 min after LPS administration and daily afterward for 2 days to determine the optimal dose for the prevention of LPS-induced cognitive impair-ments. Our data suggested that 200 IU/kg was the optimal dose effective in increasing the freezing behavior (Figure 1B). Therefore, 200 IU/kg of LMWH was used in the control + LMWH group and the LPS + LMWH group. For the con-trol group, the same dose of normal saline was injected subcutaneously.

Behavioral and cognitive tests

All behavioral tests were performed at 14:00 p.m.-17:00 p.m. in a sound-isolated room by the instrument of XR-XZ301 (Xinruan Corpora- tion, Shanghai Soft maze Information Tech- nology Co. Ltd., China). All behavioral data were recorded by the same investigator who was blinded to the animal groups as described in our previous studies [14, 15].

Open field test

The open field apparatus consisted of a square, opaque acrylic container (60 cm × 60 cm × 40 cm). A video camera fixed 60 cm above the arena tracked the animals’ movements. Mice were gently placed in the center of a white plas-tic chambers for 5 min while exploratory behav-ior was automatically recorded by a video track-ing system (XR-XZ301, Shanghai Soft maze Information Technology Co, Ltd). Travel speed lower than 0.25 mm/s was defined as immobil-ity. The total distance and total time traveled in the open field were recorded. After each test, the arena was cleaned with 75% alcohol to avoid the presence of olfactory cues.

Fear conditioning test

(3)

mem-ory was tested 48 h after training and the rats were placed in a novel chamber with a con-tinuous 3 min training tone presentation to monitor freezing behavior. Freezing behavior was defined as cessation of all movement except for respiration-related and slow pendu-lum movements. An observer blind to the treat-ment conditions scored the rats’ responses as freezing or no freezing every 10 s. The percent-age freezing was calculated using the formula (number of freezing responses/total number of responses) multiplied by 100.

Western blotting analysis

[image:3.612.96.518.71.461.2]
(4)

buffer (10 mM Tris-HCl, pH 7.4, 150 mM NaCl, 2 mM EDTA, and 0.5% Nonidet P-40) plus pro-tease inhibitors (1 μg/ml aprotinin, 1 μg/ml leu-peptin, and 1 μg/ml pepstatin A). The lysates were collected, centrifuged at 10,000 g at 4°C for 10 min. The supernatant was removed, and protein concentration was determined using the Pierce bicinchoninic acid Protein Assay kit (Pierce Technology Co., Iselin, NJ) with a bovine serum albumin standard. Equal amounts of protein were electrophoretically separated on 4-12% NuPAGE Novex Bis-Tris gradient gels (Invitrogen, NY, USA) and transferred to the nitrocellulose membranes. After blocking with 5% non-fat milk for 1 h at room temperat- ure, membranes were incubated with mouse anti-TLR4 (1:500; Santa Cruz Biotechnology), mouse anti-NF-κB (1:500; Santa Cruz Biotech- nology), mouse anti-iNOS (1:500; Santa Cruz Biotechnology), mouse anti-Cox2 (1:500; Santa Cruz Biotechnology), mouse anti-HMGB1 (1: 500; Shino-Test corporation, Kanagawa, Japan), rabbit anti-BDNF (1:500; Santa Cruz Biotechnology), and mouse anti-β-actin (1: 5000; Sigma-Aldrich) overnight at 4°C room, followed by horseradish peroxidase-conjugated secondary antibodies (GE Healthcare, Pittsbur- gh, PA) for 2 h at room temperature. The protein bands were detected by enhanced chemilumi-nescence and the quantitation of bands was undertaken using the Image J software (NIH Image, USA).

Enzyme-linked immunosorbent assay (ELISA)

The mice were killed by decapitation and the brain tissues were isolated and washed with ice-cold physiological saline to remove the sur-face blood at 24 h, day 3, day 5, and day 7 after LPS administration. The hippocampus was then separated, weighed and placed in a homogenizer. The tissue was homogenized with 1 ml ice-cold physiological saline per 100 mg brain tissue. Hypothermal centrifugation was performed at 10,000 × g for 10 min and the supernatant was obtained. Standard curves including all cytokines (in duplicates) were gen-erated using the reference cytokine concentra-tions supplied. The quantificaconcentra-tions of tumor necrosis factor alpha (TNF-α), interleukin (IL)-1β, IL-6, and IL-10 were done by the instruc-tions provided by Santa Cruz Biotechnology (Duoset kit; R&D Systems).

Measurement for malondialdehyde (MDA) and superoxide dismutase (SOD) concentrations

MDA and SOD levels were determined at 24 h, day 3, day 5, and day 7 after LPS administra-tion. The level of MDA in the hippocampus, a measure of lipid peroxidation, was assayed in the form of thiobarbituric acid-reactive sub-stances (Jiancheng Bioengineering Institute, Nanjing, China) as described in our previous study [17]. The malondialdehyde concentration is expressed as nmol/mg protein. SOD activity in the hippocampus was determined using a SOD assay kit (Jiancheng Bioengineering Institute, Nanjing, China) as described in our previous study [17]. The SOD concentration is expressed as U/mg protein.

Immunohistochemistry

The brains were histologically analyzed using paraffin-embedded sections (4 µm thick). The sections were incubated in PBS-Triton (PBS-T) containing 0.1% H2O2 for 30 min to block the endogenous peroxidase, followed by rinsing for 5 min in PBST and incubated with 1.5% normal goat serum for 30 min. The sections were then incubated overnight (4°C) using anti-active cas-pase-3 (Santa Cruz Biotechnology, Santa Cruz, CA, USA) primary antibodies, diluted 1:100 with PBS. Following extensive rinsing steps in PBS, sections were reincubated with peroxidase con-jugated secondary antibody (Dako EnVision+ system; Dako) for 1 h at room temperature. IBA1, a marker of microglia activation, was per-formed on fresh frozen tissue (10 µm coronal sections) at the level of the hippocampus. Sections were labeled with goat anti-IBA1 (1:300, Dako, Glostrup, Denmark), after pre-treatment with 1% H2O2/methanol (20 min), microwaving in citrate buffer (pH = 6) for 2~5 min and preblocking with normal rabbit serum. IBA1 sections were pretreated with 0.04% pep-sin for 20 min before blocking. The BDNF expression was determined by rabbit anti-BDNF (1:500; Santa Cruz Biotechnology).

Statistical analysis

(5)

assessed by one-way or repeated-measures of analysis of variance (ANOVA) followed by a Tukey test as appropriate. The survival rate was estimated by the Kaplan-Meier method and compared by log-rank test. A P value < 0.05 was regarded as statistically significant difference.

Results

Survival rate

Since our previous study indicated that sepsis mice showed a high mortality rate in the first 7 days, the effect of LMWH on the survival rate of sepsis mice was evaluated within the first 7 days in the present study. As shown in Figure 1C, LMWH treatment did not affect the survival rate of sepsis mice (P > 0.05).

Chronic treatment of LMWH attenuated cogni-tive impairments induced by sepsis

[image:5.612.96.520.72.405.2]

The locomotor activity was evaluated by the open field test at 7 d after LPS administration to determine whether LPS or LMWH treatment influence the locomotor activity. There was no difference in the total distance (P = 0.537) and the time spent in the center of the arena (P = 0.247) among the four groups. In the contextu-al fear conditioning test, LPS treated mice dis-played less freezing time compared with those in the control group, which was reversed by treatment of LMWH (P = 0.042). However, there was no significant difference in posttone freez-ing time in the auditory-cued fear conditionfreez-ing test among the four groups (P = 0.453, Figure 2).

(6)

LMWH treatment decreased inflammatory response, and MDA levels in the hippocampus induced by sepsis

We measured the lipid peroxidation, as indicat-ed by MDA levels. A significant increase in MDA levels was observed in the hippocampus at day 3 after sepsis development, while the treat-ment of LMWH prevented the increase in the

[image:6.612.92.520.74.528.2]

MDA levels (P = 0.037). However, no differen- ce in anti-inflammatory IL-10 and antioxidant enzyme SOD was observed among groups (P > 0.05, Figure 3). We measured TNF-α, IL-1β, IL-6, and IL-10 expressions in the hippocam-pus. As shown in Figures 3-5, sepsis up-regu-lated the inflammatory responses, as evi-denced by significantly increased IL-1β at 24 h after sepsis development when compared with Figure 3. Sepsis-induced enhanced neuroninflammation and oxidative stress were attenuated by LMWH treatment. Compared with the control + vehicle group, sepsis significantly up-regulated IL-1β and MDA expressions in the hip-pocampus, while LMWH treatment decreased IL-1β and MDA expressions induced by LPS administration. Data are shown as mean ± SEM (n = 6 per group). #P < 0.05 vs. the control + vehicle group; *P < 0.05 vs. the LPS + vehicle

(7)

the control and LMWH-treated mice (all P < 0.05). On the other hand, HMGB1, a late inflam-matory mediator, was significantly increased at day 7 after LPS administration. All these chang-es were reversed by LMWH administration.

LMWH treatment inhibited microglia activation in the hippocampus induced by sepsis

We measured IBA1 expressions in the hippo-campus. As shown in Figure 4, sepsis activated microglia, as demonstrated by IBA1-positive cells in the hippocampus when compared with the control and LMWH-treated mice (P < 0.05).

LMWH treatment decreased inflammation re-lated proteins in the hippocampus induced by sepsis

To investigate the underling mechanism, we tested whether LMWH treatment affects inflam-matory markers such as TLR4, NF-κb p65, iNOS, and Cox2 in the hippocampus. As shown in Figure 4, the TLR4, NF-κB p65, iNOS, and

LMWH treatment rescued the decreased BDNF expression in the hippocampus induced by sepsis

To investigate whether LPS induced changes of synaptic plasticity-related proteins, we exam-ined the levels of BDNF in the hippocampus. The levels of BDNF were decreased at 7 day after LPS administration compared with the control group. LMWH treatment restored the LPS-induced down-regulation of BDNF com-pared with the LPS + vehicle group (P = 0.012, Figure 5). In addition, immunohistochemical study also suggested that LMWH treatment reversed the LPS-induced down-regulation of BDNF induced by LPS administration (P = 0.026, Figure 6).

Discussion

With the increase in patients surviving a critical illness, sepsis survivors frequently experience significant neurological morbidity, including cognitive impairments [3]. This study demon-Figure 4. Representative western blotting and quantitative analysis of TLR-4,

NF-κB p65, iNOS, Cox2, occludin, HMGB1, and BDNF in the hippocampus. The TLR-4, NF-κB p65, iNOS, Cox2, and HMGB1 expressions were significantly up-regulated while occludin and BDNF were decreased after LPS administration, which were reversed by LMWH treatment. Data are shown as mean ± SEM (n = 4 per group). #P < 0.05 vs. the control + vehicle group; *P < 0.05 vs. the LPS

+ vehicle group.

Cox2 levels increased sig-nificantly at 3 h after LPS administration compared with the control group (all

P < 0.05). However, LM- WH treatment reversed the LPS-induced all these changes compared with the LPS + vehicle group (all

P < 0.05, Figure 5).

LMWH treatment in-creased occludin level in the hippocampus induced by sepsis

[image:7.612.92.381.70.336.2]
(8)

strates that sepsis survivors following LPS administration developed significant hippo-campus-dependent cognitive impairments, with concomitant enhanced hippocampal infl- ammatory responses and oxidative stress, BBB disruption, and loss of synaptic plasticity relat-ed proteins. Notably, LMWH significantly re- versed all these abnormal changes.

The bi-directional communication between the immune system and the brain is necessary for the appropriate immunological, physiological, and behavioral responses to immune challenge [16]. Under pathological situations, proinflam-matory mediators including IL-1β can be relayed from the periphery to the brain by the humoral pathways [4, 13]. We have selected a well-defined stimulus for the innate immunity by using LPS, which has enabled us to better iden-tify key molecules and pathways in LPS-induced cognitive dysfunction [13]. Microglia are the resident macrophage-like cells and play an important role in the brain’s innate immunity

[image:8.612.89.518.71.375.2]

and inflammatory responses [18]. Although microglia have essential protective roles, acti-vated microglia can contribute to neuronal cell death through the production of cytotoxic factors, including nitric oxide, TNF-α, IL-1β, and reactive oxygen species (ROS) [18]. Accu- mulating evidence has demonstrated that brain dysfunction during an immune challenge are mediated, in part by actions in the brain of pro-inflammatory cytokines [19-21]. In particular, it is reported that IL-1β levels are increased in vitro and are associated with hippocampal dys-function after sepsis development, whereas anti-IL-1 therapy appears to be able to amelio-rate the associated cognitive dysfunction [20]. In addition, increased IL-6 level in the brain has been linked with severe cognitive impairments and is likely to contribute to the cognitive and neuroanatomical alternations after sepsis development [21]. On the other hand, HMGB1 is a late onset biomarker that has been shown to mediate cognitive decline in sepsis survivors [22]. TLR4, the major signaling receptor for Figure 5. Representative micrographs of IBA1 immunolabeling in the dentate gyrus region of the hippocampus at 7 d after LPS injection.Sepsis significantly up-regulated IBA1 positive cells in the dentate gyrus region of the hippo-campus, where LMWH treatment decreased IBA1 positive cells. Scale bar: 30 µm. #P < 0.05 vs. the control + vehicle

(9)

LPS, plays a vital role in LPS-induced inflamma-tory responses [23]. TLR4 shares the capacity to bind the intracellular myeloid differentiation factor 88 [23], which further lead to the activa-tion of NF-κB that is responsible for the induc-tion of various inflammatory molecules includ-ing COX-2, iNOS, TNF-a, IL-1β, and IL-6 [24]. In the present study, we demonstrated that hippo-campal-dependent memory impairment was evident after 7 days after LPS administration, which was supported by previous studies that peripheral administration of LPS either as sin-gle injection or given repeatedly affects learn-ing and memory functions [25]. Importantly, LMWH treatment was able to decrease the markers of neuroinflammation, which might account for the protective effect of LMWH against LPS-induced cognitive impairments. In normal situations, the endothelial layer pro-vides for an anticoagulant surface to prevent the blood from clotting, which plays a vital role in the poor prognosis of sepsis as first-line responders against invading pathogens [26]. When infectious bacteria invade the

blood-ently, we showed the LMWH ameliorated the breakdown of BBB, as evidenced by increased occludin expression after sepsis development. Our data were supported by previous study demonstrating that LMWH significantly reduces the brain edema, and improves the functional outcomes in focal cerebral ischemia in rats [32]. Collectively, the beneficial effects of LMWH observed in the present study might be attributed to the direct inhibition of cytokine production or a decrease in the permeability of the BBB to systemic-produced cytokines. Although it is reported that LMWH does not cross the blood brain barrier under normal con-ditions, we can not exclude this possibility in the context of increased BBB permeability such as sepsis. The anti-inflammatory property of LMWH is likely attributed to its anticoagulant effects, by which LMWH can improve micro- circulation of organs and prevent organ dys- function.

Synaptic transmission is essential for nervous system function, and its dysfunction is a known major contributing factor to cognitive impair-Figure 6. Representative micrographs of BDNF immunolabeling in the CA1

re-gion of the hippocampus at 7 d after LPS injection.Sepsis significantly reduced BDNF expression in the CA1, where LMWH treatment increased BDNF expres-sion. Scale bar: 30 µm. #P < 0.05 vs. the control + vehicle group; *P < 0.05 vs.

the LPS + vehicle group.

[image:9.612.91.381.75.343.2]
(10)

ments [33]. In support with this hypothesis, LPS has been demonstrated to impair long-term potentiation, a key cellular process of learning and memory consolidation [31]. It has been previously demonstrated that LPS reduc-es hippocampal levels of BDNF, a neurotrophin that plays an important role in learning and memory processes [34]. Furthermore, it has been demonstrated that inhibition of IL-1β sig-naling through the addition of a soluble IL-1β receptor antagonist fully prevented the synap-tic deficit induced by LPS administration [35]. In line with these findings, we found that LMWH treatment rescued the reduction of BDNF lev-els, providing a potential mechanism by which hippocampal neuroinflammation can induce cognitive impairments induced by sepsis. Conclusion

The present study demonstrated that LMWH attenuates LPS-induced enhanced hippocam-pal inflammatory responses and oxidative stress, and BBB disruption and loss of synapt- ic plasticity related proteins, and ultimately improved cognitive impairments. Our results support the beneficial effects of anticoagulant agents on sepsis-induced cognitive impair-ments and provide relevant insights into the mechanisms involved in the cognitive impair-ments associated with sepsis and into pharma-cologic treatment to SAE.

Acknowledgements

This work was supported by the grants from the National Natural Science Foundation of China (No. 81271216, No. 81300946).

Disclosure of conflict of interest

None.

Address correspondence to: Dr. Muhuo Ji, Depart- ment of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, 305 East Zhongshan Road, Nanjing 210002, China. Tel: +86-25-5232- 3834; E-mail: jimuhuo2009@sina.com

References

[1] Iwashyna TJ, Ely EW, Smith DM, Langa KM. Long-term cognitive impairment and function-al disability among survivors of severe sepsis. JAMA 2010; 304: 1787-1794.

[2] Gilmore EJ, Gaspard N, Choi HA, Cohen E, Burkart KM, Chong DH, Claassen J, Hirsch LJ. Acute brain failure in severe sepsis: a

prospec-tive study in the medical intensive care unit utilizing continuous EEG monitoring. Intensive Care Med 2015; 41: 686-94.

[3] Shah FA, Pike F, Alvarez K, Angus D, Newman AB, Lopez O, Tate J, Kapur V, Wilsdon A, Krishnan JA, Hansel N, Au D, Avdalovic M, Fan VS, Barr RG, Yende S. Bidirectional relation-ship between cognitive function and pneumo-nia. Am J Respir Crit Care Med 2013; 188: 586-592.

[4] Widmann CN, Heneka MT. Long-term cerebral consequences of sepsis. Lancet Neurol 2014; 13: 630-636.

[5] Hughes CG, Morandi A, Girard TD, Riedel B, Thompson JL, Shintani AK, Pun BT, Ely EW, Pandharipande PP. Association between endo-thelial dysfunction and acute brain dysfunc-tion during critical illness. Anesthesiology 2013; 118: 631-639.

[6] Li LF, Yang CT, Huang CC, Liu YY, Kao KC, Lin HC. Low-molecular-weight heparin reduces hyperoxia-augmented ventilator-induced lung injury via serine/threonine kinase-protein ki-nase B. Respir Res 2011; 12: 90.

[7] Slofstra SH, van’t Veer C, Buurman WA, Reitsma PH, ten Cate H, Spek CA. Low molecu-lar weight heparin attenuates multiple organ failure in a murine model of disseminated in-travascular coagulation. Crit Care Med 2005; 33: 1365-1370.

[8] Li LF, Huang CC, Lin HC, Tsai YH, Quinn DA, Liao SK. Unfractionated heparin and enoxapa-rin reduce high-stretch ventilation augmented lung injury: a prospective, controlled animal experiment. Crit Care 2009; 13: R108. [9] Trzeciak S, Dellinger RP. Other supportive

ther-apies in sepsis: an evidence-based review. Crit Care Med 2004; 32: S571-577.

[10] Wahl F, Grosjean-Piot O, Bareyre F, Uzan A, Stutzmann JM. Enoxaparin reduces brain ede-ma, cerebral lesions, and improves motor and cognitive impairments induced by a traumatic brain injury in rats. J Neurotrauma 2000; 17: 1055-1065.

[11] Wildhagen KC, García de Frutos P, Reuteling- sperger CP, Schrijver R, Aresté C, Ortega-Gómez A, Deckers NM, Hemker HC, Soehnlein O, Nicolaes GA. Nonanticoagulant heparin pre-vents histone-mediated cytotoxicity in vitro and improves survival in sepsis. Blood 2014; 123: 1098-1101.

[12] Timmer NM, van Dijk L, van der Zee CE, Kiliaan A, de Waal RM, Verbeek MM. Enoxaparin treat-ment administered at both early and late stag-es of amyloid β deposition improvstag-es cognition of APPswe/PS1dE9 mice with differential ef-fects on brain Aβ levels. Neurobiol Dis 2010; 40: 340-347.

(11)

sep-sis induces long-lasting affective changes in the mouse. Brain Behav Immun 2015; 43: 98-109.

[14] Wu J, Dong L, Zhang M, Jia M, Zhang G, Qiu L, Ji M, Yang J. Class I histone deacetylase inhibi-tor valproic acid reverses cognitive deficits in a mouse model of septic encephalopathy. Neurochem Res 2013; 38: 2440-2449. [15] Ji M, Dong L, Jia M, Liu W, Zhang M, Ju L, Yang

J, Xie Z, Yang J. Epigenetic enhancement of brain-derived neurotrophic factor signaling pathway improves cognitive impairments in-duced by isoflurane exposure in aged rats. Mol Neurobiol 2014; 50: 937-944.

[16] Czerniawski J, Guzowski JF. Acute neuroinflam-mation impairs context discrimination memory and disrupts pattern separation processes in hippocampus. J Neurosci 2014; 34: 12470-12480.

[17] Li GM, Ji MH, Sun XJ, Zeng QT, Tian M, Fan YX, Li WY, Li N, Yang JJ. Effects of hydrogen-rich saline treatment on polymicrobial sepsis. J Surg Res 2013; 181: 279-286.

[18] Hernandes MS, D’Avila JC, Trevelin SC, Reis PA, Kinjo ER, Lopes LR, Castro-Faria-Neto HC, Cunha FQ, Britto LR, Bozza FA. The role of Nox2-derived ROS in the development of cog-nitive impairment after sepsis. J Neuroin- flammation 2014; 11: 36.

[19] Mina F, Comim CM, Dominguini D, Cassol-Jr OJ, Dall Igna DM, Ferreira GK, Silva MC, Galant LS, Streck EL, Quevedo J, Dal-Pizzol F. Il1-β in-volvement in cognitive impairment after sep-sis. Mol Neurobiol 2014; 49: 1069-1076. [20] Terrando N, Rei Fidalgo A, Vizcaychipi M, Cibelli

M, Ma D, Monaco C, Feldmann M, Maze M. The impact of IL-1 modulation on the develop-ment of lipopolysaccharide-induced cognitive dysfunction. Crit Care 2010; 14: R88.

[21] Beurel E, Jope RS. Lipopolysaccharide-induced interleukin-6 production is controlled by glyco-gen synthase kinase-3 and STAT3 in the brain. J Neuroinflammation 2009; 6: 9.

[22] Chavan SS, Huerta PT, Robbiati S, Valdes-Ferrer SI, Ochani M, Dancho M, Frankfurt M, Volpe BT, Tracey KJ, Diamond B. HMGB1 medi-ates cognitive impairment in sepsis survivors. Mol Med 2012; 18: 930-937.

[23] Sparkman NL, Kohman RA, Garcia AK, Boehm GW. Peripheral lipopolysaccharide administra-tion impairs two-way active avoidance condi-tioning in C57BL/6J mice. Physiol Behav 2005; 85: 278-288.

[24] Wang YJ, Lu J, Wu DM, Zheng ZH, Zheng YL, Wang XH, Ruan J, Sun X, Shan Q, Zhang ZF. Ursolic acid attenuates lipopolysaccharide- induced cognitive deficits in mouse brain through suppressing p38/NF-κB mediated in-flammatory pathways. Neurobiol Learn Mem. 2011; 96: 156-165.

[25] Hong ZY, Shi XR, Zhu K, Wu TT, Zhu YZ. SCM-198 inhibits microglial overactivation and at-tenuates Aβ (1-40)-induced cognitive impair-ments in rats via JNK and NF-кB pathways. J Neuroinflammation 2014; 11: 147.

[26] Clark PR, Kim RK, Pober JS, Kluger MS. Tumor Necrosis Factor Disrupts Claudin-5 Endothelial Tight Junction Barriers in Two Distinct NF-κB-Dependent Phases. PLoS One 2015; 10: e0120075.

[27] Coletta C, Módis K, Oláh G, Brunyánszki A, Herzig DS, Sherwood ER, Ungvári Z, Szabo C. Endothelial dysfunction is a potential contribu-tor to multiple organ failure and mortality in aged mice subjected to septic shock: preclini-cal studies in a murine model of cepreclini-cal ligation and puncture. Crit Care 2014; 18: 511. [28] Vassiliou AG, Mastora Z, Orfanos SE, Jahaj E,

Maniatis NA, Koutsoukou A, Armaganidis A, Kotanidou A. Elevated biomarkers of endothe-lial dysfunction/activation at ICU admission are associated with sepsis development. Cytokine 2014; 69: 240-247.

[29] Fox ED, Heffernan DS, Cioffi WG, Reichner JS. Neutrophils from critically ill septic patients mediate profound loss of endothelial barrier integrity. Crit Care 2013; 17: R226.

[30] Wang Y, Braun OÖ, Zhang S, Norström E, Thorlacius H. Monocytes regulate systemic co-agulation and inflammation in abdominal sep-sis. Am J Physiol Heart Circ Physiol 2015; 308: H540-547.

[31] Imamura Y, Wang H, Matsumoto N, Muroya T, Shimazaki J, Ogura H, Shimazu T. Interleukin-1β causes long-term potentiation deficiency in a mouse model of septic encephalopathy. Neuroscience 2011; 187: 63-69.

[32] Flierl MA, Stahel PF, Rittirsch D, Huber-Lang M, Niederbichler AD, Hoesel LM, Touban BM, Morgan SJ, Smith WR, Ward PA, Ipaktchi K. Inhibition of complement C5a prevents break-down of the blood-brain barrier and pituitary dysfunction in experimental sepsis. Crit Care 2009; 13: R12.

[33] Weberpals M, Hermes M, Hermann S, Kummer MP, Terwel D, Semmler A, Berger M, Schäfers M, Heneka MT. NOS2 gene deficiency protects from sepsis-induced long-term cognitive defi-cits. J Neurosci 2009; 29: 14177-14184. [34] Vasconcelos AR, Yshii LM, Viel TA, Buck HS,

Mattson MP, Scavone C, Kawamoto EM. Intermittent fasting attenuates lipopolysaccha-ride-induced neuroinflammation and memory impairment. J Neuroinflammation 2014; 11: 85.

Figure

Figure 1. Schematic timeline of the experimental procedure, dose-response of LMWH on fear conditioning test in mice, and effects of LMWH treatment on the survival rate within 7 days after LPS administration
Figure 2. Sepsis-induced cognitive impairments were reversed by LMWH treatment. A, B. No significant difference was observed in the total distance traveled in the open field test on day 7 after LPS administration among the four groups
Figure 3. Sepsis-induced enhanced neuroninflammation and oxidative stress were attenuated by LMWH treatment
Figure 4. Representative western blotting and quantitative analysis of TLR-4, NF-κB p65, iNOS, Cox2, occludin, HMGB1, and BDNF in the hippocampus
+3

References

Related documents

It is a matter of interest that not only in Dublin University but also in the remote districts of Ireland a spirit of progress existed, as may be gathered from the following: In

To successfully and reliably extract these excited-state energies, we need better resolution in temporal direction (large superfine isotropic lattices or anisotropic lat-

• To explore the lived experience of informal caregivers of people with PDOC and the impact caregiving has on their occupational engagement, roles and routines. • To explore

In the second intervention, we found clear adaptations of AT stiffness, which was a result of significant tendon hypertrophy and changes of the tendon material properties (i.e.

sence of exigent circumstances, a search warrant is required before law officers may enter the home of a third party to execute an arrest warrant. See Government

However, I have overcome my reticence because I am committed to constitutional government, and I believe that national level legal review is crit- ical to military operations, not

INTERNATIONAL REVIEW OF THE RED CROSS (1991), p. 167; Francoise Krill, The International Fact-Finding Commission: The ICRC's Role, ibid., p. CLAUDE PILLOUD, JEAN DE PREUX,