Abstract
Fulminant hepatitis remains a critical health problem owing to its high mortality rate and the lack of effective therapies. An increasing number of studies have shown that glutamine supplementation provides protective benefits in inflammation-related disorders, but the pharmacological significance of glutamine in lipopolysaccharide (LPS)/D-galactosamine (D-Gal)-induced fulminant hepatitis remains unclear. In the present study, the potential effects of glutamine on LPS/D-Gal-induced fulminant hepatitis were investigated. Pretreatment with glutamine decreased plasma activities of alanine and aspartate aminotransferases, and ameliorated hepatic morphological abnormalities in LPS/D-Gal-exposed mice. Glutamine pretreatment also inhibited LPS/D-Gal-induced tumor necrosis factor alpha (TNF-α) and interleukin-6 (IL-6) production. In addition, glutamine pretreatment decreased the level of cleaved cysteinyl aspartate–specific proteinase 3 (caspase-3), suppressed the activities of caspase-3, caspase-8, and caspase-9, and reduced the number of cells positive for TdT-mediated dUTP nick-end labeling in LPS/D-Gal-challenged mice. Interestingly, post-treatment with glutamine also provided protective benefits against LPS/D-Gal-induced acute liver injury, although these effects were less robust than those of glutamine pre-treatment. Thus, glutamine may have potential value as a pharmacological intervention in fulminant hepatitis.
Keywords: Glutamine, fulminant hepatitis, inflammation, apoptosis, lipopolysaccharide, D-galactosamine
Impact Statement
Fulminant hepatitis is a serious clinical syndrome with high morbidity and mortality rates. Glutamine is the most abundant free amino acid in mammals. Accumulating evidence indicates that glutamine supplementation provides protection against inflammation-related disorders. The current results showed that glutamine supplementation alleviated lipopolysaccharide (LPS)/D-galactosamine (D-Gal)-induced fulminant liver injury, suppressed the release of inflammatory factors, and reduced apoptosis. Importantly, the therapeutic administration of glutamine post-LPS/D-Gal also provided appreciable protection against fulminant hepatitis. These results indicate that glutamine may have potential value as a pharmacological intervention for fulminant hepatitis.
Introduction
Fulminant hepatitis remains a critical health problem due to its high mortality, which is caused by several factors, including viruses, bacteria, drugs, alcohol, and so on. 1 It is widely accepted that uncontrolled inflammation and excessive apoptosis are the main underlying mechanisms.2–4 Lipopolysaccharide (LPS), a key pathogenic component of Gram-negative bacteria, is a crucial pro-inflammatory factor that triggers the release of inflammatory cytokines and initiates an inflammatory response.5,6 LPS can lead to acute liver injury with severe inflammation and massive hepatocyte apoptosis in D-galactosamine (D-Gal)-sensitized mice, which has been extensively used for the experimental investigation of fulminant hepatitis.7–9
Glutamine is the most abundant free amino acid in mammals.10,11 A growing body of evidence indicates that glutamine provides protective benefits against inflammation-related disorders.12,13 It has been reported that glutamine supplementation suppresses the production of the pro-inflammatory cytokine tumor necrosis factor alpha (TNF-α) and attenuates the degree of colon injury in dextran sodium sulfate (DSS)-induced colitis. 14 In addition, glutamine supplementation was shown to inhibit inflammatory responses, relieve organ injury, and improve the survival rate of mice with systemic inflammation.12,15–17 Therefore, glutamine may be a promising reagent for the treatment of inflammation-related disorders, but it remains unclear whether glutamine supplementation also results in beneficial outcomes in fulminant hepatitis.
To investigate the potential effects of glutamine in fulminant hepatitis, an LPS/D-Gal-induced acute liver injury model was employed in the current study. Glutamine was injected into mice with LPS/D-Gal-induced acute liver injury and the degree of liver injury, production of inflammatory cytokines, and induction of hepatocyte apoptosis were evaluated.
Materials and methods
Materials and regents
Regents were obtained as follows: LPS, D-Gal were obtained from Sigma-Aldrich (St. Louis, MO, USA). Glutamine was obtained from Adamas-beta (Shanghai, China). The alanine aminotransferase (ALT) and aspartate aminotransferase (AST) assay kits were produced by Nanjing Jiancheng Bioengineering Institute (Nanjing, China). The ELISA kits for mouse TNF-α (EMC102a) and mouse interleukin-6 (IL-6) (EMC004) were from Neo Bioscience Technology Company (Shenzhen, China). Assay kits for caspases 3 (C1116), 8 (C1152) and 9 (C1158) were obtained from Beyotime Institute of Biotechnology (Jiangsu, China). Antibodies against cleaved caspase-3, the HRP-conjugated anti-rabbit IgG, and the HRP-conjugated anti-mouse IgG were provided by Cell Signaling Technology (Ann Arbor, MI, USA). Antibody against β-actin were purchased from 4A BIOTECH (Beijing, China). The BCA protein assay kit was purchased from Pierce Biotechnology (Rockford, IL, USA). The enhanced chemiluminescence (ECL) reagent was obtained from Advansta (Menlo Park, CA, USA).
Animals
Male C57BL/6 J mice (18–22 g) with an average age of 6–8 weeks were provided by the Experimental Animal Center of Chongqing Medical University (Chongqing, China). The animals were housed under controlled temperatures (22°C–25°C), relative humidity (50% ± 5%), and light conditions (12 h:12 h light/dark cycle), and were allowed ad libitum access to water and diet. The Ethics Committee of Chongqing Medical University approved all the mouse experiments.
Experimental protocol
To induce fulminant hepatitis, the mice were intraperitoneally injected with LPS (10 μg/kg, dissolved in normal saline) combined with D-Gal (700 mg/kg, dissolved in normal saline). Mice were randomly divided into the following four groups (n = 8 per group): (1) the control group: mice were treated with the same volume of solvent; (2) the glutamine group: mice were injected intraperitoneally with glutamine (50 mg/kg, dissolved in normal saline); (3) the LPS/D-Gal group: mice received intraperitoneal injection of LPS combined with D-Gal; and (4) the glutamine + LPS/D-Gal group: glutamine was intraperitoneally injected 30 min before LPS/D-Gal exposure. The mice were anesthetized and sacrificed 1.5 h after the LPS/D-Gal challenge. Blood samples were collected and added to tubes, followed by centrifugation to collect plasma to measure the level of TNF-α. In addition, another 32 mice underwent the same procedure as described above, and were sacrificed 6 h after LPS/D-Gal injection. Blood and liver tissues were collected and stored at −80°C for protein extraction, histopathological analysis, and other tests.
Since pretreatment is not practical in clinical practice, an additional 24 mice were selected to investigate the therapeutic effect of glutamine post-treatment on acute liver injury. Mice were randomly divided into three groups (n = 8 per group): (1) the control group: mice were treated with the same volume of solvent; (2) the LPS/D-Gal group: mice were intraperitoneally injected with LPS and D-Gal; and (3) the glutamine + LPS/D-Gal group: glutamine was injected 1.5 h after LPS/D-Gal exposure. Finally, the mice were anesthetized and killed 6 h after LPS/D-Gal injection. Blood and liver tissues were collected and stored at −80°C.
Histological analysis
Liver tissues of mice were fixed in 4% paraformaldehyde and embedded in paraffin. The samples were then sectioned (4 μm) and stained with hematoxylin and eosin (H&E) to evaluate pathological changes in the liver. Finally, the sections were examined under a light microscope (Olympus, Japan).
Evaluation of plasma AST and ALT
Blood samples were separated by centrifugation to obtain plasma samples. The biochemical indices in plasma, including ALT and AST, were determined according to the manufacturer’s instructions to determine the degree of liver injury. First, plasma samples and an ALT (or AST) matrix solution were added to 96-well plates and incubated for 30 min at 37°C. Second, 2-4-dinitrophenylhydrazine was added to each well and incubated for 20 min at 37°C. Finally, NaOH solution was added, and the levels of ALT or AST were calculated according to the absorbance measured at 490 nm based on the standard curve.
Analysis of inflammatory cytokines
TNF-α levels were determined 1.5 h after LPS/D-Gal injection, whereas IL-6 levels were determined 6 h after LPS/D-Gal injection. Both values were determined using ELISA kits according to the manufacturer’s instructions. TNF-α and IL-6 levels were calculated using a standard curve.
Determination of caspase activity
To evaluate the activities of caspases 3, 8 and 9, liver tissue was homogenized in lysis buffer, and then centrifuged at 4°C for 15 min at 16,000 × g. Appropriate amounts of supernatant were transferred to a clean test tube, and the protein concentration of the supernatant was determined using the Bradford protein determination kit. According to the instructions of the caspase activity assay kit, 40 μL detection buffer, 50 μL supernatant, and 10 μL of substrate for caspase (AC-DevD-ρNA for caspase-3, AC-IETD-ρNA for caspase-8, and AC-LEhd-ρNA for caspase-9) were mixed. After incubation at 37ºC for 1.5 h, absorbance was measured at 405 nm. One unit (U) of caspase activity is defined as the amount of caspase cleaving 1 nmol of colorimetric substrate within 1 h at 37ºC when the substrate is saturated. Caspase activities were expressed as units per mg total protein.
Western blot analysis
Total protein was prepared from the liver tissues. Protein concentration was determined using the BCA reagent. Protein was separated using SDS-PAGE and then transferred to nitrocellulose membrane. After blocking with 5% nonfat milk in TBST (TBS + 0.1% Tween-20) for 2 h, the blots were incubated overnight with a primary antibody against cleaved caspase-3, followed by three washes in TBST for 10 min each. The membranes were then incubated with the secondary antibody for 2 h at room temperature, followed by three washes in TBST for 10 min each. Finally, Ab binding was visualized with ECL reagents and the ChemiDoc Touch Imaging System (Bio-Rad), and the intensity of the bands was quantified using the ImageJ software.
TUNEL assay
Paraffin-embedded liver tissue sections were prepared for the TUNEL assay, which was performed by using the In Situ Cell Death Detection Kit (Roche), according to the manufacturer’s instructions. A dark-brown precipitate in the nuclei was produced in terminal transferase reactions, which represent apoptotic cells. Images were observed under a light microscope (Olympus).
Statistical analysis
Statistical analyses were performed using GraphPad Prism (version 8.0.1). All data are presented as means ± SD. Statistical significance between multiple groups was analyzed using one-way analysis of variance (ANOVA) with Tukey’s post hoc test. Statistical significance was set at a p-value < 0.05.
Results
Pretreatment with glutamine alleviates LPS/D-Gal-induced liver injury
Damaged hepatocytes release ALT and AST into the circulation. Thus, elevated plasma transaminase activities indicate the degree of liver injury. 18 In this study, compared to the control group, plasma ALT and AST activities significantly increased after LPS/D-Gal challenge. However, glutamine pretreatment suppressed LPS/D-Gal-induced elevation of plasma ALT and AST (Figure 1). LPS/D-Gal exposure also produced disorganized hepatic lobules, hepatocyte necrosis and inflammatory cell infiltration, but glutamine pretreatment attenuated these hallmarks of ultrastructural damage (Figure 2).
Figure 1.
Pretreatment with glutamine reverses LPS/D-Gal-induced elevation of plasma transaminase. Glutamine was administered 30 min before the LPS/D-Gal challenge. Plasma was collected 6 h after LPS/D-Gal injection to determine alanine aminotransferase (ALT, A) and aspartate aminotransferase (AST, B) activities. Values are means ± SD, n = 8 per group.
Figure 2.
Pretreatment with glutamine alleviates LPS/D-Gal-induced liver histological abnormalities. Liver samples were collected 6 h after LPS/D-Gal exposure and stained with hematoxylin and eosin. White arrows indicate hepatocyte necrosis; black arrows indicate inflammatory cell infiltration. Scale bar = 50 μm. (A color version of this figure is available in the online journal.)
Pretreatment with glutamine decreases the level of pro-inflammatory cytokines
TNF-α and IL-6 are the major pro-inflammatory cytokines involved in LPS/D-Gal-induced liver injury.19,20 LPS/D-Gal injection sharply increased plasma TNF-α and IL-6 concentrations at 1.5 and 6 h post-injection, respectively, which was suppressed by glutamine pretreatment (Figure 3).
Figure 3.
Pretreatment with glutamine decreases plasma concentrations of pro-inflammatory cytokines in LPS/D-Gal-induced liver injury. Plasma samples were collected 1.5 h after LPS/D-Gal exposure to measure TNF-α (A) and 6 h after LPS/D-Gal exposure to measure IL-6 levels (B). Values are means ± SD, n = 8 per group.
Pretreatment with glutamine suppresses hepatocytes apoptosis
LPS/D-Gal exposure induces massive apoptosis of hepatocytes, 21 which is mediated by the activation of caspase cascades and cleavage of a large number of functional proteins. 22 To confirm whether glutamine modulates LPS/D-Gal-induced hepatocyte apoptosis, the activities of caspases 3, 8 and 9 were measured. Pretreatment with glutamine suppressed activation of all three caspases in the liver of LPS/D-Gal-exposed mice (Figure 4). In addition, western blot analysis showed that pretreatment with glutamine attenuated procaspase-3 cleavage (Figure 5) and reduced the number of TUNEL-positive cells (Figure 6) in the LPS/D-Gal-challenged mice.
Figure 4.
Pretreatment with glutamine suppresses LPS/D-Gal-induced activation of caspases. Liver samples were collected 6 h after LPS/D-Gal exposure. The activity of (A) caspase-3, (B) caspase-8, and (C) caspase-9 in liver tissue were determined. Values are means ± SD, n = 8.
Figure 5.
Pretreatment with glutamine suppresses LPS/D-Gal-induced upregulation of cleaved caspase-3. Liver samples were collected 6 h after LPS/D-Gal exposure. (A) The level of cleaved caspase-3 in liver tissue were detected by immunoblot analysis. (B) The blots were semi-quantified. Values are means ± SD, n = 4.
Figure 6.
Pretreatment with glutamine decreases hepatocyte apoptosis in LPS/D-Gal-exposed mice. Liver samples were collected 6 h after LPS/D-Gal exposure. Apoptotic cells were detected using the TUNEL method, with the dark-brown nuclei representing the TUNEL-positive cells. (A) The representative liver sections of each group were shown. Scale bar = 100 μm. (B) 10 high power fields were randomly selected to count the number of TUNEL-positive cells. Values are means ± SD, n = 4. (A color version of this figure is available in the online journal.)
Post-treatment with glutamine attenuates LPS/D-Gal-induced liver damage
In order to investigate whether glutamine has therapeutic potential in liver injury induced by LPS/D-Gal, we administered glutamine 1.5 h after LPS/D-Gal injection. Glutamine post-treatment attenuated plasma ALT and AST activities and improved the ultrastructural abnormalities inflicted by LPS/D-Gal (Figure 7). In addition, caspase-3 activity and the TUNEL-positivity of cells in LPS/D-Gal-induced mice were also inhibited by glutamine post-treatment (Figure 8).
Figure 7.
Post-treatment with glutamine protects against LPS/D-Gal-induced liver injury. Glutamine was injected intraperitoneally 1.5 h after LPS/D-Gal exposure. The mice were sacrificed 6 h after LPS/D-Gal injection, and plasma was collected to determine (A) ALT and (B) AST levels. Values are means ± SD, n = 8 per group. Representative hematoxylin and eosin-stained liver sections are shown (C). White arrows indicate hepatocyte necrosis; black arrows indicate inflammatory cell infiltration. Scale bar = 50 μm. (A color version of this figure is available in the online journal.)
Figure 8.
Post-treatment with glutamine decreases hepatocyte apoptosis in LPS/D-Gal exposed mice. Liver was sampled 6 h after LPS/D-Gal exposure to determine caspase-3 activity (A). Apoptotic cells were detected using the TUNEL method, and those with dark-brown nuclei indicated TUNEL-positive cells (B) which were counted in 10 high-power fields. C: Representative sections showing TUNEL-positive cells. Values are means ± SD, n = 4. Scale bar = 100 μm. (A color version of this figure is available in the online journal.)
Discussion
A compelling body of evidence indicates that glutamine, the most abundant free amino acid in the mammalian body, has appreciable therapeutic potential in several pathological conditions.12,16,23 The present study demonstrated that treatment with glutamine effectively alleviated LPS/D-Gal-induced fulminant hepatitis, as evidenced by the suppressed plasma ALT and AST activities, and attenuated hepatic histological abnormalities. These findings corroborate and extend previous studies, where glutamine supplementation alleviated hepatic disorders induced by acetaminophen overdose, carbon tetrachloride (CCl4) intoxication, thioacetamide (TAA) exposure, and ischemia-reperfusion.24–27 Therefore, glutamine supplementation might provide hepatoprotective benefits in inflammatory-, oxidative stress- and metabolic disturbance-based liver injury.
In LPS/D-Gal-induced liver injury, inflammatory cells are activated and many pro-inflammatory cytokines are secreted. 28 TNF-α and IL-6 are the main pro-inflammatory cytokines that play crucial roles in the development of liver injury induced by LPS/D-Gal.19,20 In LPS/D-Gal-induced acute liver injury, TNF-α levels rise rapidly, usually peaking 1–2 h after LPS/D-Gal exposure.29,30 Subsequently, TNF-α, an early-phase pro-inflammatory factor, can stimulate neutrophils, monocytes, and vascular endothelial cells to release inflammatory factors, such as IL-6, resulting in cascade amplification and hyperactivation of the inflammatory response over the next several hours. 31 Therefore, in this study serum TNF-α and IL-6 concentrations were measured at 1.5 and 6 h after LPS/D-Gal exposure, respectively.32,33 In this study, glutamine downregulated the levels of TNF-α and IL-6 in LPS/D-Gal-challenged mice. Consistent with the present findings, a previous study showed that glutamine effectively reduced the release of TNF-α and IL-6 in LPS-injected rats. 34 Glutamine also attenuated plasma TNF-α and IL-6 in rats subjected to ischemia- and reperfusion-induced gut injury.4,35 In addition, glutamine decreased TNF-α production and protected against DSS-induced colitis in mice. 14 These results indicate that the protective effect of glutamine in LPS/D-Gal-induced liver injury may be related to its anti-inflammatory properties.
In addition to the uncontrolled inflammatory response, accumulating evidence indicates that excessive hepatocyte apoptosis is a hallmark of LPS/D-Gal-induced hepatic damage.4,21,33,36 In the current study, glutamine treatment prevented LPS/D-Gal-induced activation of the caspase cascade, which initiated hepatocyte apoptosis. In addition, glutamine reduced the number of TUNEL-positive cells in the LPS/D-Gal-challenged liver. Collectively, these results demonstrate an anti-apoptotic effect of glutamine in LPS/D-Gal-induced liver injury. Concordant with our results, previous studies have shown that glutamine significantly decreases the number of apoptotic cells induced by ischemia/reperfusion in the liver. 37 Glutamine treatment also attenuated tubular cell apoptosis in kidney tissues of rats with myoglobinuric acute kidney injury. 38 In ischemia/reperfusion injury of the intestine, glutamine reduced the content of pro-apoptotic Bax, increased anti-apoptotic Bcl-2 content, and decreased the number of apoptotic cells in the ileum tissues of rats. 4 In addition, glutamine reduced apoptosis in rats with TNBS-induced colitis. 39 Thus, the protective effect of glutamine in LPS/D-Gal-induced liver injury may also be attributed to its antiapoptotic properties.
Apoptosis is triggered by the activation of death receptors via the extrinsic pathway. 40 Interaction of TNF-α with its death receptor leads to activation of the caspase cascade, culminating in apoptosis.19,40 In LPS/D-Gal-induced liver injury, the increased expression of TNF-α was suppressed by glutamine, suggesting TNF-α suppression may be a molecular mechanism underlying the anti-apoptotic actions of glutamine. These anti-apoptotic actions were confirmed by in vitro experiments. 41 For example, glutamine increased cell survival and decreased apoptosis in TNF-α/IFN-γ-induced young adult mouse colonic epithelial cells. 42 Glutamine also suppressed apoptosis in camptothecin-induced IEC-18 cells following heat shock. 43 In addition, glutamine decreased apoptosis induced by the endoplasmic reticular stressors brefeldin A and tunicamycin in Caco-2 cells. 39 Therefore, glutamine might modulate apoptotic pathways and provide anti-apoptotic benefits in LPS/D-Gal-induced liver injury.
Since glutamine pretreatment would be impractical in most clinical situations, we tested whether glutamine is hepatoprotective when administered after the insult. Interestingly, glutamine post-treatment decreased plasma ALT and AST activities and alleviated the liver histological abnormalities induced by LPS/D-Gal. The findings suggest that glutamine post-treatment may have therapeutic potential for treatment of acute liver injury.
Collectively, the results of this study demonstrated that treatment with glutamine alleviated LPS/D-Gal-induced liver injury, which may be attributed to its anti-inflammatory and antiapoptotic properties. Although further investigation is required to decipher the molecular mechanisms mediating glutamine hepatoprotection, this study indicates that glutamine might have potential value as a pharmacological intervention in acute liver injury.
Footnotes
Authors’ Contributions: Y-QY and LZ designed the experiments. M-XY and X-YZ conducted the experiments. Data analysis was performed by SZ, R-YS, K-RF, and KH. M-XY, LZ, and Y-QY wrote the manuscript. All authors reviewed the final manuscript.
The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.
Funding: The author(s) disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: This research was supported by a grant from the Science and Technology Research Program of the Chongqing Municipal Education Commission (No. KJQN201900435) and a grant from the National Natural Science Foundation of China (No. 81871606).
ORCID IDs: Mengxin Yang
https://orcid.org/0000-0003-0602-0582
Xinyue Zhang
https://orcid.org/0000-0003-1003-0214
Ruyue Shao
https://orcid.org/0000-0001-7714-2427
Yongqiang Yang
https://orcid.org/0000-0003-2048-8309
References
- 1. Bernal W, Auzinger G, Dhawan A, Wendon J. Acute liver failure. Lancet 2010;376:190–201 [DOI] [PubMed] [Google Scholar]
- 2. Puengel T, Tacke F. Repair macrophages in acute liver failure. Gut 2018;67:202–3 [DOI] [PubMed] [Google Scholar]
- 3. Fan XG, Pei SY, Zhou D, Zhou PC, Huang Y, Hu XW, Li T, Wang Y, Huang ZB, Li N. Melittin ameliorates inflammation in mouse acute liver failure via inhibition of PKM2-mediated Warburg effect. Acta Pharmacol Sin 2021;42:1256–66 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Mondello S, Galuppo M, Mazzon E, Domenico I, Mondello P, Carmela A, Cuzzocrea S. Glutamine treatment attenuates the development of ischaemia/reperfusion injury of the gut. Eur J Pharmacol 2010;643:304–15 [DOI] [PubMed] [Google Scholar]
- 5. Ambade A, Catalano D, Lim A, Mandrekar P. Inhibition of heat shock protein (molecular weight 90 kDa) attenuates proinflammatory cytokines and prevents lipopolysaccharide-induced liver injury in mice. Hepatology 2012;55:1585–95 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Fu YJ, Xu B, Huang SW, Luo X, Deng XL, Luo S, Liu C, Wang Q, Chen JY, Zhou L. Baicalin prevents LPS-induced activation of TLR4/NF-kappaB p65 pathway and inflammation in mice via inhibiting the expression of CD14. Acta Pharmacol Sin 2021;42:88–96 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Yang S, Kuang G, Zhang L, Wu S, Zhao Z, Wang B, Yin X, Gong X, Wan J. Mangiferin attenuates LPS/D-GalN-induced acute liver injury by promoting HO-1 in Kupffer cells. Front Immunol 2020;11:285. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Yue S, Wang T, Yang Y, Fan Y, Zhou L, Li M, Fu F. Lipopolysaccharide/D-galactosamine-induced acute liver injury could be attenuated by dopamine receptor agonist rotigotine via regulating NF-κB signaling pathway. Int Immunopharmacol 2021;96:107798. [DOI] [PubMed] [Google Scholar]
- 9. Yang C, He L, Wang C, Huang Y, Wang A, Li X, Ao J. Dexmedetomidine alleviated lipopolysaccharide/D-galactosamine-induced acute liver injury in mice. Int Immunopharmacol 2019;72:367–73 [DOI] [PubMed] [Google Scholar]
- 10. Bernfeld E, Foster DA. Glutamine as an essential amino acid for KRas-driven cancer cells. Trends Endocrinol Metab 2019;30:357–68 [DOI] [PubMed] [Google Scholar]
- 11. Durante W. The emerging role of l-glutamine in cardiovascular health and disease. Nutrients 2019;11:2092. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Oliveira GP, Oliveira MB, Santos RS, Lima LD, Dias CM, Ab’ Saber AM, Teodoro WR, Capelozzi VL, Gomes RN, Bozza PT, Pelosi P, Rocco PR. Intravenous glutamine decreases lung and distal organ injury in an experimental model of abdominal sepsis. Crit Care 2009;13:R74 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Singleton KD, Serkova N, Beckey VE, Wischmeyer PE. Glutamine attenuates lung injury and improves survival after sepsis: role of enhanced heat shock protein expression*. Crit Care Med 2005;33:1206–13 [DOI] [PubMed] [Google Scholar]
- 14. Yan S, Hui Y, Li J, Xu X, Li Q, Wei H. Glutamine relieves oxidative stress through PI3K/Akt signaling pathway in DSS-induced ulcerative colitis mice. Iran J Basic Med Sci 2020;23:1124–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Singleton KD, Serkova N, Beckey VE, Wischmeyer PE. Glutamine attenuates lung injury and improves survival after sepsis: role of enhanced heat shock protein expression. Crit Care Med 2005;33:1206–13 [DOI] [PubMed] [Google Scholar]
- 16. Singleton KD, Beckey VE, Wischmeyer PE. Glutamine prevents activation of NF-kappaB and stress kinase pathways, attenuates inflammatory cytokine release, and prevents acute respiratory distress syndrome (ARDS) following sepsis. Shock 2005;24:583–9 [DOI] [PubMed] [Google Scholar]
- 17. Wischmeyer PE, Kahana M, Wolfson R, Ren H, Musch MM, Chang EB. Glutamine induces heat shock protein and protects against endotoxin shock in the rat. J Appl Physiol 2001;90:2403–10 [DOI] [PubMed] [Google Scholar]
- 18. Ozer J, Ratner M, Shaw M, Bailey W, Schomaker S. The current state of serum biomarkers of hepatotoxicity. Toxicology 2008;245:194–205 [DOI] [PubMed] [Google Scholar]
- 19. Yang F, Li X, Wang LK, Wang LW, Han XQ, Zhang H, Gong ZJ. Inhibitions of NF-kappaB and TNF-alpha result in differential effects in rats with acute on chronic liver failure induced by d-Gal and LPS. Inflammation 2014;37:848–57 [DOI] [PubMed] [Google Scholar]
- 20. Li L, Duan C, Zhao Y, Zhang X, Yin H, Wang T, Huang C, Liu S, Yang S, Li X. Preventive effects of interleukin-6 in lipopolysaccharide/d-galactosamine induced acute liver injury via regulating inflammatory response in hepatic macrophages. Int Immunopharmacol 2017;51:99–106 [DOI] [PubMed] [Google Scholar]
- 21. Hu K, Gong X, Ai Q, Lin L, Dai J, Cai L, Jiang R, Ge P, Zhang L. Endogenous AMPK acts as a detrimental factor in fulminant hepatitis via potentiating JNK-dependent hepatocyte apoptosis. Cell Death Dis 2017;8:e2637 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Julien O, Wells JA. Caspases and their substrates. Cell Death Differ 2017; 24:1380–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Jing L, Wu Q, Wang F. Glutamine induces heat-shock protein and protects against Escherichia coli lipopolysaccharide-induced vascular hyporeactivity in rats. Crit Care 2007;11:R34 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Sözen S, Kisakürek M, Yildiz F, Gönültaş M, Dinçel AS. The effects of glutamine on hepatic ischemia reperfusion injury in rats. Hippokratia 2011;15:161–6 [PMC free article] [PubMed] [Google Scholar]
- 25. Shrestha N, Chand L, Han MK, Lee SO, Kim CY, Jeong YJ. Glutamine inhibits CCl4 induced liver fibrosis in mice and TGF-β1 mediated epithelial-mesenchymal transition in mouse hepatocytes. Food Chem Toxicol 2016;93:129–37 [DOI] [PubMed] [Google Scholar]
- 26. Schemitt EG, Hartmann RM, Colares JR, Licks F, Salvi JO, Marroni CA, Marroni NP. Protective action of glutamine in rats with severe acute liver failure. World J Hepatol 2019;11:273–86 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Hong RW, Rounds JD, Helton WS, Robinson MK, Wilmore DW. Glutamine preserves liver glutathione after lethal hepatic injury. Ann Surg 1992;215:114–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Lv H, An B, Yu Q, Cao Y, Liu Y, Li S. The hepatoprotective effect of myricetin against lipopolysaccharide and D-galactosamine-induced fulminant hepatitis. Int J Biol Macromol 2020;155:1092–104 [DOI] [PubMed] [Google Scholar]
- 29. Bahrami S, Redl H, Leichtfried G, Yu Y, Schlag G. Similar cytokine but different coagulation responses to lipopolysaccharide injection in D-galactosamine-sensitized versus nonsensitized rats. Infect Immun 1994;62:99–105 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Yang H, Li TW, Zhou Y, Peng H, Liu T, Zandi E, Martínez-Chantar ML, Mato JM, Lu SC. Activation of a novel c-Myc-miR27-prohibitin 1 circuitry in cholestatic liver injury inhibits glutathione synthesis in mice. Antioxid Redox Signal 2015;22:259–74 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Zelová H, Hošek J. TNF-α signalling and inflammation: interactions between old acquaintances. Inflamm Res 2013;62:641–51 [DOI] [PubMed] [Google Scholar]
- 32. Peng X, Yang Y, Tang L, Wan J, Dai J, Li L, Huang J, Shen Y, Lin L, Gong X, Zhang L. Therapeutic benefits of apocynin in mice with lipopolysaccharide/D-galactosamine-induced acute liver injury via suppression of the late stage pro-apoptotic AMPK/JNK pathway. Biomed Pharmacother 2020;125:110020. [DOI] [PubMed] [Google Scholar]
- 33. Lv X, Zhou H, Hu K, Lin L, Yang Y, Li L, Tang L, Huang J, Shen Y, Jiang R, Wan J, Zhang L. Activation of PKM2 metabolically controls fulminant liver injury via restoration of pyruvate and reactivation of CDK1. Pharmacol Res 2021;172:105838. [DOI] [PubMed] [Google Scholar]
- 34. Huang J, Liu J, Chang G, Wang Y, Ma N, Roy AC, Shen X. Glutamine supplementation attenuates the inflammation caused by LPS-induced acute lung injury in mice by regulating the TLR4/MAPK signaling pathway. Inflammation 2021;44:2180–92 [DOI] [PubMed] [Google Scholar]
- 35. Zabot GP, Carvalhal GF, Marroni NP, Hartmann RM, da Silva VD, Fillmann HS. Glutamine prevents oxidative stress in a model of mesenteric ischemia and reperfusion. World J Gastroenterol 2014;20: 11406–14 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Luo M, Zhao A, Li J, Chen Y, Tian D, Wang C, Hu Z, Gao J. Acute liver injury attenuation of a novel recombinant sTNFR through blocking hepatic apoptosis. Immunopharmacol Immunotoxicol 2015;37:295–300 [DOI] [PubMed] [Google Scholar]
- 37. Xu F, Dai CL, Peng SL, Zhao Y, Jia CJ, Xu YQ. Preconditioning with glutamine protects against ischemia/reperfusion-induced hepatic injury in rats with obstructive jaundice. Pharmacology 2014;93:155–65 [DOI] [PubMed] [Google Scholar]
- 38. Kim YS, Jung MH, Choi MY, Kim YH, Sheverdin V, Kim JH, Ha HJ, Park DJ, Kang SS, Cho GJ, Choi WS, Chang SH. Glutamine attenuates tubular cell apoptosis in acute kidney injury via inhibition of the c-Jun N-terminal kinase phosphorylation of 14-3-3. Crit Care Med 2009;37:2033–44 [DOI] [PubMed] [Google Scholar]
- 39. Crespo I, San-Miguel B, Prause C, Marroni N, Cuevas MJ, Gonzalez-Gallego J, Tunon MJ. Glutamine treatment attenuates endoplasmic reticulum stress and apoptosis in TNBS-induced colitis. Plos One 2012; 7:e50407 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Brenner C, Galluzzi L, Kepp O, Kroemer G. Decoding cell death signals in liver inflammation. J Hepatol 2013;59:583–94 [DOI] [PubMed] [Google Scholar]
- 41. Jiao N, Xu D, Qiu K, Wang L, Wang L, Piao X, Yin J. Restoring mitochondrial function and normalizing ROS-JNK/MAPK pathway exert key roles in glutamine ameliorating bisphenol A-induced intestinal injury. FASEB J 2020;34:7442–61 [DOI] [PubMed] [Google Scholar]
- 42. Xue H, Sufit AJ, Wischmeyer PE. Glutamine therapy improves outcome of in vitro and in vivo experimental colitis models. JPEN J Parenter Enteral Nutr 2011;35:188–97 [DOI] [PubMed] [Google Scholar]
- 43. Ropeleski MJ, Riehm J, Baer KA, Musch MW, Chang EB. Anti-apoptotic effects of L-glutamine-mediated transcriptional modulation of the heat shock protein 72 during heat shock. Gastroenterology 2005;129:170–84 [DOI] [PubMed] [Google Scholar]