Expression and role of TNIP2 in multiple organ dysfunction syndrome following severe trauma

  • Authors:
    • Hui Gong
    • Xiaomin Sheng
    • Jianhua Xue
    • Dongbo Zhu
  • View Affiliations

  • Published online on: January 24, 2019     https://doi.org/10.3892/mmr.2019.9893
  • Pages: 2906-2912
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Severe trauma can result in secondary multiple organ dysfunction syndrome (MODS) and death. Inflammation response and oxidative stress promote the occurrence and development of MODS. TNFAIP3‑interacting protein 2 (TNIP2), which can repress the activation of nuclear factor‑κB (NF‑κB) and may be involved in MODS progression, has not been studied in regards to MODS. The present study aimed to investigate the expression, role and mechanism of TNIP2 in MODS following severe trauma. The expression level of TNIP2 was initially detected in the blood of patients with MODS using reverse transcription‑quantitative polymerase chain reaction and western blot assay. Then, to investigate the role of TNIP2 in MODS, a MODS rat model was conducted by trauma and the model rats were treated with TNIP2‑plasmid (intraperitoneal injection). Blood levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST), lactate dehydrogenase (LDH), blood urea nitrogen (BUN), creatine (Cr) and creatine kinase (CK); and tumor necrosis factor α (TNF‑α), high‑mobility group box 1 (HMGB‑1), malondialdehyde (MDA) and total antioxidant capacity (TAC) in the different groups were assessed. In addition, activation of NF‑κB was assessed by detecting the level of phospho‑p65. The results showed that TNIP2 was significantly decreased in the blood of patients with MODS. TNIP2 was also significantly downregulated in the blood and the pulmonary, renal and hepatic tissues of MODS rats. The levels of ALT, AST, LDH, BUN, Cr and CK were markedly increased in the blood of MODS rats, and these increases were inhibited by TNIP2‑plasmid administration. Moreover, blood levels of TNF‑α, HMGB‑1 and MDA were significantly increased in MODS rats, while TAC was notably decreased, and these changes were prevented by TNIP2‑plasmid administration. Furthermore, it was found that activation of NF‑κB induced by MODS was eliminated by TNIP2‑plasmid. In conclusion, the data indicated that TNIP2 is significantly decreased in MODS following severe trauma, and it plays a protective role in MODS development by inhibiting the inflammation response and oxidative stress by preventing NF‑κB activation.

Introduction

Severe trauma can cause local tissue damage, systemic inflammatory response syndrome, as well as multiple organ dysfunction syndrome (MODS), shock and even death (1,2). MODS is one of the major, serious complications following severe trauma and is the progressive dysfunction of one or more organ systems resulting from an exaggerated and long-term inflammatory response to severe illness and/or injury. Multiple factors including severe shock, infection, burns, trauma and severe pancreatitis are involved in the pathologic process of MODS (37). In recent years, the mechanism underlying MODS caused by trauma is under investigation, and there are a number of hypotheses to explain the pathological mechanism. However, the mechanisms underlying MODS caused by trauma have not yet been fully elucidated. At present, due to the lack of effective treatment, MODS is still a major cause of morbidity and mortality in intensive care units (8,9).

Nuclear factor (NF)-κB is a protein complex that controls transcription of DNA, cytokine production and cell survival. NF-κB is found in almost all animal cell types and is involved in cellular responses to stimuli such as stress, cytokines and free radicals (1012). Abnormal expression of NF-κB has been reported to be linked to cancer, inflammatory and autoimmune diseases, septic shock, viral infection and improper immune development (13,14). Recent studies have reported that NF-κB is activated during the progression of MODS (15,16). TNIP2 (also termed ABIN2) interacts with several components of the NF-κB signaling pathway and can both positively and negatively regulate NF-κB-dependent transcription of target genes (17). The TNIP2 gene encodes a protein that represses the activation of NF-κB, and it is generally accepted that TNIP2 has an essential role in the NF-κB signaling pathway (18). Moreover, studies have indicated that TNIP2 plays an important role in acute pancreatitis-induced myocardial injury and lupus nephritis via regulating the NF-κB pathway (19,20).

To the best of our knowledge, no study has been conducted concerning the role of TNIP2 in the development and progression of MODS. Therefore, the aim of the present study was to investigate the role and molecular mechanism of TNIP2 in MODS following severe trauma.

Materials and methods

Clinical specimens

Peripheral blood samples (5 ml per individual) from 16 patients with MODS and peripheral blood samples from 16 healthy individuals were collected at the Affiliated Hospital of Nantong University (Nantong, China) from August 2014 to April 2017. The inclusion criteria were as follows: i) No history of injury; ii) injury severity score (ISS), ≥16; iii) age, ≥18 years; and iv) time interval between trauma and admission, <90 h. The exclusion criteria were as follows: i) Mortality within 24 h after trauma; ii) main diagnosis, cardiac trauma; iii) intracranial hemorrhage; iv) heart, liver, kidney or endocrine history. Following admission, the age, sex, cause of trauma and medical history of the patients were recorded. Blood samples were collected from MODS patients 48 h after sever trauma and stored at −85°C. Informed consent was obtained from every patient enrolled and the present study was approved by the Ethics Committee of the Affiliated Hospital of Nantong University.

Animal model of MODS

A total of 40 male Sprague-Dawley (SD) rats (~200 g) were purchased from Vital River Company (Beijing, China). All rats were fed ad libitim and maintained under standard conditions at 22–30°C and a 12-h light/dark cycle. The experimental protocol was approved by the Animal Care Committee of Nantong University, and experiments were performed under the guidelines of guide for the care and use of laboratory animals (21). The rat model of MODS was designed and established according to a previous study (22). To investigate the role of TNIP2 in MODS, the rats were randomly assigned into 4 groups: Control, MODS model, MODS model + control-plasmid [C; intraperitoneal (i.p.) injection] and MODS model + TNIP2-plasmid (T; i.p. injection). The rats in the control and MODS model groups received saline (0.9% NaCl) solution i.p. instead of the plasmids. All rats were anesthetized with 30 mg/kg pentobarbital and handled at certain time points (6, 12, 24 and 48 h). Following the specific treatment, the subsequent experiments were conducted.

To confirm multiple organ injury/dysfunction, the following biochemical indicators in the plasma were measured. Liver injury was assessed by detecting the enhanced levels of alanine aminotransferase (ALT; a specific marker for hepatic parenchymal injury), aspartate aminotransferase (AST; a nonspecific marker for hepatic injury) and lactate dehydrogenase (LDH) in the plasma. Renal dysfunction was assessed by the increased levels of creatine (Cr) and blood urea nitrogen (BUN) (indicator of reduced glomerular filtration rate and hence renal failure) in the plasma (23).

Serum biochemical analyses

At 6, 12, 24 and 48 h after treatment, 3 ml venous blood was collected from the rats in all groups. Serum was prepared through centrifugation (4°C, 1,000 × g, 15 min) and stored at −80°C. To detect the levels of ALT, AST, LDH, BUN, CK and Cr, a Hitachi Automatic Analyzer 7170 (Hitachi, Ltd., Japan) was utilized.

Enzyme-linked immunosorbent assay (ELISA)

At 6, 12, 24, and 48 h after treatment, the serum of rats were harvested by centrifugation (1,000 × g, 10 min) to determine the secretion of TNF-α, HMGB-1, MDA and TAC using an ELISA kit (Abcam, Cambridge, MA, USA) following the manufacturer's protocols. Every sample was detected at least three times by utilizing a microplate reader (Bio-Rad Laboratories, Inc., Hercules, CA, USA).

Western blot analysis

Western blot analysis was carried out as previously described (24). Total proteins were extracted from tissues/blood using radioimmunoprecipitation asay lysis buffer [50 mM Tris (pH 7.4), 150 mM NaCl, 1% NP-40, 0.5% sodium deoxycholate] supplemented with phenylmethylsulfonyl fluoride at a final concentration of 1 mM. Protein concentration was determined using the bicinchoninic acid protein assay (Pierce; Thermo Fisher Scientific, Inc., Waltham, MA, USA). Equal amounts of protein (30 µg/lane) were resolved using SDS-PAGE on 10% gels, and then transferred to a polyvinylidene difluoride membrane (EMD Millipore, Billerica, MA, USA). Following blocking with 5% skimmed milk in TBS-Tween at room temperature for 1.5 h, the samples were probed with antibodies against TNIP2 (cat no. sc-271850; dilution 1:1,000; Santa Cruz Biotechnology, Inc., Dallas, TX, USA), p-p65 (cat no. 3033; dilution 1:1,000; Cell Signaling Technology, Inc., Danvers, MA, USA), p65 (cat no. 8242; dilution 1:1,000; Cell Signaling Technology, Inc., Danvers, MA, USA), and β-actin (cat no. 4970; 1:1,000; Cell Signaling Technology, Inc.). After three times of washing, the blots were then incubated with horseradish peroxidase-conjugated secondary antibody (anti-rabbit IgG; 1:2,000; cat no. 7074; Cell Signaling Technology, Inc.) at room temperature for 2 h. Immunoreactive bands were visualized using the enhanced chemiluminescence detection system (Applygen Technologies, Inc., Beijing, China). ImageJ 1.38X (National Institutes of Health, Bethesda, MD, USA) was used to perform densitometry.

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

RT-qPCR was performed as previously described (24). In brief, total RNA was extracted from tissues/blood samples using RNAiso Plus (Takara Bio, Inc., Otsu, Japan) following the manufacturer's instructions. RT was performed to synthesize cDNAs using the ThermoScript RT-PCR system (Invitrogen; Thermo Fisher Scientific, Inc.). qPCR was performed to analyze the synthesized cDNA using SYBR Green PCR Master Mix (Applied Biosystems; Thermo Fisher Scientific, Inc.) on a 7900 Real-Time PCR System (Applied Biosystems; Thermo Fisher Scientific, Inc.). The conditions of qPCR used for amplification were as follows: 95°C for 5 min, 40 cycles at 95°C for 30 sec, 55°C for 30 sec, and 72°C for 30 sec, then 72°C for 10 min. The primer sequences used for qPCR are as follows: TNIP2, forward 5′-CTAAAGAGGCGGCAGGTCCCTC-3′ and reverse 5′-CAAGATGACCTTCCAGTGAC-3′; GAPDH, forward 5′-CTTTGGTATCGTGGAAGGACTC-3′ and reverse 5′-GTAGAGGCAGGGATGATGTTCT-3′. GAPDH served as an internal control. The relative gene expression was assessed by using the 2−ΔΔCq method (25). The experiment was repeated at least three times.

Statistical analysis

All data analyses were performed using SPSS 17.0 software (SPSS, Inc., Chicago, IL, USA). Results are expressed as mean ± standard deviation. Student's t-tests and one-way analysis of variance followed by Student-Newman-Keuls tests were performed to analyze the differences between groups. P<0.05 was considered to indicate a statistically significant difference.

Results

Basic patient characteristics

The present study included 16 patients. The basic characteristics of the patients are presented in Table I.

Table I.

Basic patient characteristics and clinical data 24 h after admission (n=16).

Table I.

Basic patient characteristics and clinical data 24 h after admission (n=16).

Patient characteristicsValues
Sex (male/female)10/6
Age range (years)34-57
ISS34.9±6.7
APACHE II score25.3±8.1
CRP (mg/l)104.7±13.7
WBCs (×109/l)13.2±9.1
Neutrophils (%)83.9±5.5

[i] ISS, injury severity score; APACHE II, acute physiology and chronic health evaluation II; CRP, Creactive protein; WBCs, white blood cells.

TNIP2 is significantly reduced in MODS patients and rats

To detect the expression level of TNIP2 in the blood of MODS patients and rats, RT-qPCR and western blot assay were used, respectively. As shown in Fig. 1A and B, compared with the healthy control, the protein and mRNA levels of TNIP2 were significantly decreased in the blood samples of patients with MODS. The data indicated that TNIP2 may be involved in the development of MODS.

Then the levels of TNIP2 were detected in the blood and in the pulmonary, renal, and hepatic tissues of MODS rats. The findings revealed that TNIP2 was significantly downregulated in the blood and the pulmonary, renal and hepatic tissues of the MODS rats (Fig. 1C-J).

Blood levels of ALT, AST, LDH, BUN, Cr and CK

To assess changes in the cellular integrity and functionality in the organs at 6, 12, 24 and 48 h after trauma, blood tests were performed. As shown in Fig. 2, the levels of ALT, AST, LDH, BUN, Cr and CK in the blood of the MODS rats were significantly higher at different time points after injury than those in the rats of the control group. TNIP2-plasmid administration significantly reduced the levels of ALT, AST, LDH, BUN, Cr and CK in the blood of the MODS rats.

Blood levels of TNF-α and HMGB-1

As shown in Fig. 3, the production of TNF-α and HMGB-1 in the blood of the MODS rats was markedly higher at different time points after injury than those in the rats of the control group. TNIP2-plasmid administration significantly reduced the levels of TNF-α and HMGB-1 in the blood of the MODS rats.

Blood levels of MDA and TAC

As shown in Fig. 4, the production of MDA in the blood of the MODS rats was higher, and TAC was lower, at different time points after injury than those in the rats of the control group. However, TNIP2-plasmid administration significantly decreased the MDA level and increased TAC in the blood of the MODS rats.

Effect of TNIP2 on the activation of NF-κB in MODS rats

To investigate the mechanism of the effect of TNIP2 on MODS rats, the NF-κB pathway was detected (Fig. 5). Forty-eight hours after treatment, total protein and RNA were extracted from tissues/blood of rats in the different groups and measured by western blot assay and qRT-PCR. Our results showed that compared with the control group, the mRNA and protein level of TNIP2 was significantly decreased in the MODS model group, and compared with the MODS model group, TNIP2-plasmid notably increased TNIP2 expression. Phosphorylation of p65 was significantly enhanced in the MODS rats compared to the control rats, and this enhancement was eliminated by TNIP2-plasmid administration (Fig. 5).

Discussion

In the present study, it was determined that TNIP2 is significantly decreased in MODS following severe trauma. Our findings also showed that TNIP2 plays a protective role in MODS development by the inhibiting inflammation response and oxidative stress through regulation of NF-κB activation. We revealed that TNIP2 may be a promising therapeutic target for the treatment of MODS.

Severe post-traumatic complications, such as sepsis and systemic inflammatory response syndrome (SIRS), are the leading cause of mortality in hospitals with a mortality rate of 30–50%. SIRS can eventually lead to (26). Multiple organ failure (MOF) is common in trauma patients with the most severe injuries, and 29% of trauma patients present with one or more systemic organ failure (27,28). MOF is the leading cause of morbidity and delayed mortality in survivors immediately after injury. The mortality rate of trauma patients with failure of three organ systems is approximately 67%, and the mortality rate of patients with failure of four organ systems is close to 100%. There is a distinct difference between MODS and MOF. The function of MODS organs is not completely lost and will not fail, while the organs of MOF patients have sequential failure of function. Early detection of MODS and interventions can improve organ function. At present, the therapeutic effect of MODS remains unsatisfactory. Therefore, it is urgent to find new effective diagnostic markers and therapeutic targets for MODS.

Severe trauma can lead to the release of various inflammatory mediators and inflammatory cytokines in the body and cause systemic inflammatory response syndrome and oxidative stress (29,30). Inflammation response and oxidative stress promote the occurrence and development of MODS (31,32). NF-κB is an important transcription factor involved in the regulation of proliferation, survival, apoptosis, immune, oxidative stress, and inflammatory reaction (3336). In addition, studies have revealed the important role of NF-κB in the progression of MODS (15,37). Over-activated NF-κB may significantly enhance the inflammatory response and oxidative stress in MODS patient and cause tissue injury, thus leading to patient death. TNIP2 gene encodes a protein that can repress the activation of NF-κB, thus we hypothesized that it may be involved in the development of MODS.

We firstly investigated the expression level of TNIP2 in MODS patients and MODS rats, and the MODS rat model was conducted. The findings indicated that TNIP2 was significantly decreased in the blood of MODS patients, and significant downregulation of TNIP2 was also observed in the blood, pulmonary, renal, and hepatic tissues of MODS rats, indicating the downregulation of TNIP2 in MODS. Then, to study the effect of TNIP2 on MODS rats, the rats were treated with or without the TNIP2-plasmid. It was found that the increased expression levels of markers of cellular integrity and organ function including ALT, AST, LDH, BUN, Cr and CK in the blood of rats induced by MODS were inhibited by TNIP2-plasmid administration. Moreover, it was found that blood levels of TNF-α, HMGB-1 and MDA were significantly enhanced in MODS rats, while TAC was notably decreased, and these changes were notably reversed by TNIP2-plasmid treatment, indicating that TNIP2-plasmid administration prevented inflammation response and oxidative stress in MODS rats. Finally, to investigate the mechanism of the effect of TNIP2 on MODS rats, the NF-κB pathway was detected. The results revealed that the phosphorylation of p65 was significantly enhanced in MODS rats compared to the control rats, and these enhancements were eliminated by TNIP2-plasmid administration.

Taken together, it was found for the first time that TNIP2 is decreased in MODS, and it plays a protective role in MODS development by inhibiting inflammation response and oxidative stress via repressing the activation of the NF-κB pathway. TNIP2 may be a potential diagnostic marker and therapeutic target for MODS treatment.

Acknowledgements

Thanks to Dr Dongbo Zhu, chief physician of Nantong University Affiliated Hospital (Nantong, China) for his help and guidance.

Funding

No funding was recieved.

Availability of data and materials

The data sets used during the present study are available from the corresponding author upon reasonable request.

Authors' contributions

HG contributed to study design; HG, XS and JX contributed to data collection, statistical analysis and data interpretation; DZ contributed to data collection, manuscript preparation and literature searching; All authors contributed to the development of the manuscript.

Ethics approval and consent to participate

Written informed consent was obtained from every patient enrolled, and the present study was approved by the Ethics Committee of the Affiliated Hospital of Nantong University. The experimental protocol was approved by the Animal Care Committee of Nantong University, and experiments were performed following with the guidance of guide for the care and use of laboratory animals (21).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Uzzan B, Cohen R, Nicolas P, Cucherat M and Perret GY: Procalcitonin as a diagnostic test for sepsis in critically ill adults and after surgery or trauma: A systematic review and meta-analysis. Crit Care Med. 34:1996–2003. 2006. View Article : Google Scholar : PubMed/NCBI

2 

Fabiano G, Pezzolla A, Filograna MA and Ferrarese F: Traumatic shock-physiopathologic aspects. G Chir. 29:51–57. 2008.PubMed/NCBI

3 

Hanisch E, Brause R, Paetz J and Arlt B: Review of a large clinical series: Predicting death for patients with abdominal septic shock. J Intensive Care Med. 26:27–33. 2011. View Article : Google Scholar : PubMed/NCBI

4 

Mera S, Tatulescu D, Cismaru C, Bondor C, Slavcovici A, Zanc V, Carstina D and Oltean M: Multiplex cytokine profiling in patients with sepsis. APMIS. 119:155–163. 2011. View Article : Google Scholar : PubMed/NCBI

5 

Nguyen LN and Nguyen TG: Characteristics and outcomes of multiple organ dysfunction syndrome among severe-burn patients. Burns. 35:937–941. 2009. View Article : Google Scholar : PubMed/NCBI

6 

Walsh CR: Multiple organ dysfunction syndrome after multiple trauma. Orthop Nurs. 24:324–333. 2005. View Article : Google Scholar : PubMed/NCBI

7 

Bhatia M: Inflammatory response on the pancreatic acinar cell injury. Scand J Surg. 94:97–102. 2005. View Article : Google Scholar : PubMed/NCBI

8 

Liu F, Yang S, Liu W, Tang B, Zhang W, Zhang C, Liao W and Hu A: Early thrombolytic failure in a patient with massive pulmonary embolism combined with multiple organ dysfunction syndrome: What next? J Int Med Res. 46:3440–3445. 2018. View Article : Google Scholar : PubMed/NCBI

9 

Huster D, Härtel F, Nuding S, Schroeder J, Zhang Y, Werdan K and Ebelt H: Prognostic relevance of tissue oxygen saturation in patients in the early stage of multiple organ dysfunction syndrome. Med Klin Intensivmed Notfmed. 2018.PubMed/NCBI

10 

Gilmore TD: Introduction to NF-kappaB: Players, pathways, perspectives. Oncogene. 25:6680–6684. 2006. View Article : Google Scholar : PubMed/NCBI

11 

Brasier AR: The NF-kappaB regulatory network. Cardiovas Toxicol. 6:111–130. 2006. View Article : Google Scholar

12 

Perkins ND: Integrating cell-signalling pathways with NF-kappaB and IKK function. Nat Rev Mol Cell Biol. 8:49–62. 2007. View Article : Google Scholar : PubMed/NCBI

13 

Pasparakis M: Regulation of tissue homeostasis by NF-κB signalling: Implications for inflammatory diseases. Nat Rev Immunol. 9:778–788. 2009. View Article : Google Scholar : PubMed/NCBI

14 

Baker RG, Hayden MS and Ghosh S: NF-κB, inflammation, and metabolic disease. Cell metabolism. 13:11–22. 2011. View Article : Google Scholar : PubMed/NCBI

15 

Wang YT, Fu JJ, Li XL, Li YR, Li CF and Zhou CY: Effects of hemodialysis and hemoperfusion on inflammatory factors and nuclear transcriptionfactors in peripheral blood cell of multiple organ dysfunction syndrome. Eur Rev Med Pharmacol Sci. 20:745–750. 2016.PubMed/NCBI

16 

Wang YL, Shen HH, Cheng PY, Chu YJ, Hwang HR, Lam KK and Lee YM: 17-DMAG, an HSP90 inhibitor, ameliorates multiple organ dysfunction syndrome via induction of HSP70 in endotoxemic rats. PLoS One. 11:e01555832016. View Article : Google Scholar : PubMed/NCBI

17 

Banks CAS, Boanca G, Lee ZT, Eubanks CG, Hattem GL, Peak A, Weems LE, Conkright JJ, Florens L and Washburn MP: TNIP2 is a hub protein in the NF-κB network with both protein and RNA mediated interactions. Mol Cell Proteomics. 15:3435–3449. 2016. View Article : Google Scholar : PubMed/NCBI

18 

Verstrepen L, Carpentier I, Verhelst K and Beyaert R: ABINs: A20 binding inhibitors of NF-kappa B and apoptosis signaling. Biochem Pharmacol. 78:105–114. 2009. View Article : Google Scholar : PubMed/NCBI

19 

Xie H, Yang M, Zhang B, Liu M and Han S: Protective role of TNIP2 in myocardial injury induced by acute pancreatitis and its mechanism. Med Sci Monit. 23:5650–5656. 2017. View Article : Google Scholar : PubMed/NCBI

20 

Wang W, Gao J and Wang F: MiR-663a/MiR-423-5p are involved in the pathogenesis of lupus nephritis via modulating the activation of NF-κB by targeting TNIP2. Am J Transl Res. 9:3796–3803. 2017.PubMed/NCBI

21 

Bayne K: Revised Guide for the Care and Use of Laboratory Animals available. American Physiological Society. Physiologist. 39:208, 199–211. 1996.

22 

Teng L, Yu M, Li JM, Tang H, Yu J, Mo LH, Jin J and Liu XZ: Matrix metalloproteinase-9 as new biomarkers of severity in multiple organ dysfunction syndrome caused by trauma and infection. Mol Cell Biochem. 360:271–277. 2012. View Article : Google Scholar : PubMed/NCBI

23 

Cuzzocrea S, Mazzon E, Di Paola R, Genovese T, Serraino I, Dugo L, Cuzzocrea E, Fulia F, Caputi AP and Salvemini D: Protective effect of M40401, selective superoxide dismutase mimetic, on zymosan-induced nonseptic shock. Crit Care Med. 32:157–167. 2004. View Article : Google Scholar : PubMed/NCBI

24 

Liu C, Wang L, Chen W, Zhao S, Yin C, Lin Y, Jiang A and Zhang P: USP35 activated by miR let-7a inhibits cell proliferation and NF-κB activation through stabilization of ABIN-2. Oncotarget. 6:27891–27906. 2015.PubMed/NCBI

25 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using realtime quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

26 

Brongel L: Guidelines for severe multiple and multiorgan traumatic injuries. Przegl Lek. 60:56–62. 2003.PubMed/NCBI

27 

Frohlich M, Lefering R, Probst C, Paffrath T, Schneider MM, Maegele M, Sakka SG, Bouillon B and Wafaisade A; Committee on Emergency Medicine, Intensive Care, Trauma Management of the German Trauma Society Sektion NIS, : Epidemiology and risk factors of multiple-organ failure after multiple trauma: An analysis of 31,154 patients from the TraumaRegister DGU. J Trauma Acute Care Surg. 76:921–927. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Vogel JA, Liao MM, Hopkins E, Seleno N, Byyny RL, Moore EE, Gravitz C and Haukoos JS: Prediction of postinjury multiple-organ failure in the emergency department: Development of the denver emergency department trauma organ failure score. J Trauma Acute Care Surg. 76:140–145. 2014. View Article : Google Scholar : PubMed/NCBI

29 

Liu MH, Tian J, Su YP, Wang T, Xiang Q and Wen L: Cervical sympathetic block regulates early systemic inflammatory response in severe trauma patients. Med Sci Monit. 19:194–201. 2013. View Article : Google Scholar : PubMed/NCBI

30 

Pierce A and Pittet JF: Inflammatory response to trauma: Implications for coagulation and resuscitation. Curr Opin Anaesthesiol. 27:246–252. 2014. View Article : Google Scholar : PubMed/NCBI

31 

Wang Z, Chen R, Zhu Z, Zhang X and Wang S: Effects of insulin combined with ethyl pyruvate on inflammatory response and oxidative stress in multiple-organ dysfunction syndrome rats with severe burns. Am J Emerg Med. 34:2154–2158. 2016. View Article : Google Scholar : PubMed/NCBI

32 

Osterbur K, Mann FA, Kuroki K and DeClue A: Multiple organ dysfunction syndrome in humans and animals. J Vet Intern Med. 28:1141–1151. 2014. View Article : Google Scholar : PubMed/NCBI

33 

Leotoing L, Chereau F, Baron S, Hube F, Valencia HJ, Bordereaux D, Demmers JA, Strouboulis J and Baud V: A20-binding inhibitor of nuclear factor-κB (NF-κB)-2 (ABIN-2) is an activator of inhibitor of NF-κB (IκB) kinase α (IKKα)-mediated NF-κB transcriptional activity. J Biol Chem. 286:32277–32288. 2011. View Article : Google Scholar : PubMed/NCBI

34 

Liang Y, Zhou Y and Shen P: NF-kappaB and its regulation on the immune system. Cell Mol Immunol. 1:343–350. 2004.PubMed/NCBI

35 

Vallabhapurapu S and Karin M: Regulation and function of NF-κB transcription factors in the immune system. Annu Rev Immunol. 27:693–733. 2009. View Article : Google Scholar : PubMed/NCBI

36 

Mariappan N, Elks CM, Sriramula S, Guggilam A, Liu Z, Borkhsenious O and Francis J: NF-kappaB-induced oxidative stress contributes to mitochondrial and cardiac dysfunction in type II diabetes. Cardiovasc Res. 85:473–483. 2010. View Article : Google Scholar : PubMed/NCBI

37 

Liu S, Zhang J, Pang Q, Song S, Miao R, Chen W, Zhou Y and Liu C: The protective role of curcumin in zymosan-induced multiple organ dysfunction syndrome in mice. Shock. 45:209–219. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

April-2019
Volume 19 Issue 4

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Gong H, Sheng X, Xue J and Zhu D: Expression and role of TNIP2 in multiple organ dysfunction syndrome following severe trauma. Mol Med Rep 19: 2906-2912, 2019
APA
Gong, H., Sheng, X., Xue, J., & Zhu, D. (2019). Expression and role of TNIP2 in multiple organ dysfunction syndrome following severe trauma. Molecular Medicine Reports, 19, 2906-2912. https://doi.org/10.3892/mmr.2019.9893
MLA
Gong, H., Sheng, X., Xue, J., Zhu, D."Expression and role of TNIP2 in multiple organ dysfunction syndrome following severe trauma". Molecular Medicine Reports 19.4 (2019): 2906-2912.
Chicago
Gong, H., Sheng, X., Xue, J., Zhu, D."Expression and role of TNIP2 in multiple organ dysfunction syndrome following severe trauma". Molecular Medicine Reports 19, no. 4 (2019): 2906-2912. https://doi.org/10.3892/mmr.2019.9893