Anti‑inflammatory effect of metformin against an experimental model of LPS‑induced cytokine storm

  • Authors:
    • Ibrahim Taher
    • Eman El‑Masry
    • Mohamed Abouelkheir
    • Ahmed E. Taha
  • View Affiliations

  • Published online on: July 13, 2023     https://doi.org/10.3892/etm.2023.12114
  • Article Number: 415
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Cytokine storm is one of the leading causes of death in patients with COVID‑19. Metformin has been shown to inhibit the action of a wide range of proinflammatory cytokines such as IL‑6, and TNF‑α which may ultimately affect cytokine storm due to Covid‑19. The present study analyzed the anti‑inflammatory effect of oral and intraperitoneal (IP) metformin administration routes in a mouse model of lipopolysaccharide (LPS)‑induced cytokine storm. A total of 60 female BALB/c mice were randomly assigned to one of six groups: i) Control; ii) LPS model; iii) oral saline + LPS; iv) oral metformin + LPS; v) IP saline + LPS; and vi) IP metformin + LPS. Metformin or saline were administered to the mice for 30 days, after which an IP injection of 0.5 mg/kg LPS induced a cytokine storm in the five treatment groups. Mice were sacrificed and serum cytokine levels were measured. Pretreatment of mice with either oral or IP metformin significantly reduced the increase in IL‑1, IL‑6 and TNF‑α following LPS injection. Both metformin administration routes significantly reduced IL‑1 and TNF‑α levels, although IP metformin appeared to be significantly more effective at reducing IL‑6 levels compared with oral metformin. Neither the oral or IP route of administration of metformin demonstrated a significant effect on IL‑17 levels. Based on its ability to suppress the proinflammatory LPS‑induced cytokine storm, metformin may have future potential benefits in ameliorating human diseases caused by elevated cytokine levels.

Introduction

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was the causative agent of the Coronavirus disease 2019 (COVID-19) pandemic. A number of drug treatments such as antiviral drugs; monoclonal antibodies; convalescent plasma and cytokine therapy targeting the SARS-CoV-2 immunopathological process have recently been either approved or tested to treat COVID-19 (1,2). While a majority of the cases of SARS-CoV-2 infection are asymptomatic or associated with mild symptoms, 10-20% of patients with COVID-19 may encounter acute respiratory distress syndrome (ARDS), particularly patients who are elderly or have co-morbidities (3). Recent studies have reported a fatal immunopathological process defined as a ‘cytokine storm’, which is a component of the macrophage activation syndrome, also known as secondary hemophagocytic lymphohistiocytosis that can lead to ARDS (4,5). Several cytokines, such as: IL-1; IL-5; IL-7; IL-9; IL-10 and TNF-α, were detected at higher concentrations in the serum of patients with a severe case of COVID-19, compared with those who had milder infections (6-10). Further research assessing the cytokine profile changes and their mechanisms are necessary to comprehend how COVID-19 infections can become severe, in addition to providing information that could be used to develop treatment options to control disease pathogenesis (11).

In order for the immune system to target an invading virus, antigen-presenting cells (APCs) process and present viral antigens to other cells of the immune system. Viral antigens are recognized by CD8+ cytotoxic T lymphocytes and natural killer (NK) cells, activating both the innate and adaptive branches of the immune system. Apoptosis is induced by NK cells and CD8+ cytotoxic T cells to kill virus-infected cells. To avoid the unnecessary initiation of cell death, APCs and CD8+ cytotoxic T cells are being eliminated through apoptosis after antigenic reactivity has ceased. However, defects in the cytotoxic activities of lymphocytes, whether acquired or genetic, can result in a failure of CD8+ cytotoxic T cells and NK cells to lyse infected cells and activate APCs, which leads to exaggerated and prolonged interactions between adaptive and innate immune cells. High levels of serum proinflammatory cytokines, such as IL-1, IL-6, IL-17 and TNF-α, are then released uncontrollably, resulting in a cytokine storm. Cytokine storm, ARDS, thrombotic tendency, disseminated intravascular coagulation, hepatic dysfunction and multi-organ failure can result from this immunopathologic process (5,11,12). This life-threatening phenomenon is regarded as the leading cause of death in patients with COVID-19 (13,14).

Metformin is an anti-diabetic medication with a well-characterized safety profile. Previous studies have reported that metformin may have other physiological effects in addition to lowering blood glucose levels (15). It may influence the AMP kinase (AMPK)/mTOR signaling pathway, which in turn regulates a variety of proinflammatory cytokines, such as IL-1; IL-2: IL-6; IL-12 and TNF-α (15). Metformin, an immunomodulatory drug with high tolerability, is an add-on drug in the treatment of certain malignant, autoimmune and aging-related conditions, such as antiproliferative and antioxidant effects, T2D, obesity associated inflammation, autoimmune diseases and cardio and nephro-protection (16-23). In patients with chronic inflammation, metformin can be used as a protective or therapeutic option (24,25), including conditions such as colitis-associated colon cancer (26), otitis media (27) and airway inflammation (28). Metformin has previously been reported to aid in the treatment of sepsis-related brain injury by inhibiting neuroinflammation, oxidative stress and apoptosis (29).

The aim of the present study was to evaluate the anti-inflammatory effect of oral and intraperitoneal (IP) metformin in a mouse model of lipopolysaccharide (LPS)-induced cytokine storm.

Materials and methods

Animal model

The present study included 60 female wild-type BALB/c mice aged 5-6 weeks and weighing ~25 g. The animal cages were well ventilated, and subjected to a 12-h light/dark cycle. The relative humidity was kept at around 45-55%. The rooms temperature was adjusted to a range of 22-24˚C. Food and water were freely available to the animals. After 2 weeks of acclimation, mice were randomly assigned to one of six groups (n=10/group): i) Control; ii) LPS model; iii) oral saline + LPS; iv) oral metformin + LPS; v) IP saline + LPS; and vi) IP metformin + LPS. The control group received no intervention other than the collection of blood samples at the end of the experiment to estimate the basal levels of serum cytokines. For 30 days, metformin or saline were administered to the mice via either the oral or IP route. Mice were administered metformin at a concentration of 250 mg/kg body weight diluted in 100 µl normal saline once daily via oral gavage or the IP route (28). Saline-treated groups of mice were administered an equivalent volume of saline (100 µl) daily, either orally or via the IP route. The Jouf University Local Committee of Bioethics approved all experimental procedures (approval no. 07-08-42; Sakaka, Saudi Arabia).

Murine model of LPS-induced cytokine storm

After 30 days of metformin or saline administration, LPS was administered to the five intervention groups of mice through IP injection at a concentration of 0.5 mg/kg body weight (from O111:B4 Escherichia coli; MilliporeSigma) to induce a cytokine storm. LPS was prepared in sterile PBS before use. At one hour following LPS was administration, mice were sacrificed by halothane at a lethal dose (5%) for 2-3 min after which animals were confirmed to be dead based on their physical status (unable to walk and lacking response to manipulations). Then blood was collected from the heart (30). Serum separator tubes were used to collect blood samples. Blood samples were allowed to clot for 2 h at room temperature, then centrifuged at 3.74 x g for 15 min at 4˚C, before being stored at -80˚C.

Measurement of cytokines levels

Serum levels of IL-1β, IL-6, IL-17 and TNF-α were determined using ELISA kits according to the manufacturer's instructions (cat. nos. CSB-E08054m, CSB-E04639m, CSB-E04608m and CSB-E04741m, respectively; Cusabio Technology LLC). All experimental measurements were repeated in triplicates.

Statistical analysis

Data are presented as the mean ± standard error of the mean and were analyzed using SPSS (v22; IBM Corp.). One-way ANOVA followed by Tukey's post hoc test was used to determine statistical significance between treatment groups. The correlation among the cytokine levels within each treatment group of mice was analyzed using Spearman's rank correlation coefficient analysis, and the results were presented as P- and rho values. P<0.05 was considered to indicate a statistically significant difference.

Results

Serum IL-1β levels

There were no significant differences in the body weight of the treatment groups of mice compared with the control group. LPS injection significantly increased the serum levels of IL-1β in all treatment groups compared with the control (P<0.05; Fig. 1). Pretreatment with metformin, either orally or IP, significantly reduced this rise in IL-1β levels (P<0.05), with no significant difference demonstrated between the efficacy of the two methods of metformin administration.

Serum IL-6 levels

Compared with the control group, injection of LPS caused a significant elevation of IL-6 levels in all the treatment groups (P<0.05; Fig. 2). Pretreatment with either oral or IP metformin significantly attenuated this increase in IL-6 levels (P<0.05). IP metformin significantly decreased serum IL-6 levels compared with the oral route of metformin administration (P<0.05).

Serum IL-17 levels

Injection of LPS caused a significant increase in serum IL-17 levels in all treatment groups compared with the control group (Fig. 3). However, serum levels of IL-17 were not significantly impacted by the route of metformin administration.

Serum TNF-α levels

Compared with the control, LPS injection caused a significant increase in TNF-α levels across all treatment groups (P<0.05; Fig. 4). Metformin pretreatment significantly decreased the TNF-α levels in serum compared with the LPS + saline groups. There was no significant difference demonstrated by the route of metformin administration on the reduction of serum TNF-α levels.

Correlation analysis

The results of the Spearman's correlation analysis among measured proinflammatory cytokines within each of the different subgroups demonstrated that certain correlations were statistically significant (Table I). The magnitude of the correlation coefficient indicated the strength of the relationship between the variables, with larger absolute values indicating stronger relationships.

Table I

Spearman's correlation coefficient analysis between production of serum cytokines in a mouse model of LPS-induced cytokine storm pretreated with oral or IP metformin.

Table I

Spearman's correlation coefficient analysis between production of serum cytokines in a mouse model of LPS-induced cytokine storm pretreated with oral or IP metformin.

A, Control
CytokineIL-6IL-17TNF-α
IL-10.027 (0.633)a0.103 (0.438)0.028 (0.632)a
IL-6 0.006 (0.774)a0.210 (0.287)
IL-17  0.067 (0.515)
B, LPS
CytokineIL-6IL-17TNF-α
IL-10.037 (0.596)a0.049 (0.559)a0.156 (0.358)
IL-6 0.125 (0.399)0.021 (0.663)a
IL-17  0.171 (0.334)
C, LPS + oral saline
CytokineIL-6IL-17TNF-α
IL-10.027 (0.636)a0.116 (0.418)0.077 (0.491)
IL-6 0.033 (0.612)a0.214 (0.285)
IL-17  0.089 (0.467)
D, LPS + oral metformin
CytokineIL-6IL-17TNF-α
IL-10.041 (0.584)a0.033 (0.612)a0.006 (0.770)a
IL-6 0.221 (0.274)0.035 (0.602)a
IL-17  0.002 (0.830)a
E, LPS + IP saline
CytokineIL-6IL-17TNF-α
IL-10.005 (0.782)a0.005 (0.782)a0.000 (0.915)a
IL-6 0.008 (0.758)a0.015 (0.697)a
IL-17  0.002 (0.830)a
F, LPS + IP metformin
CytokineIL-6IL-17TNF-α
IL-10.0014 (0.851)a0.0008 (0.879)a0.005 (0.782)a
IL-6 0.003 (0.815)a0.003 (0.815)a
IL-17  0.003 (0.818)a

[i] Data are presented as P- and (rho) values.

[ii] aP<0.05 (statistically significant). Spearman's rho values range from -1 to +1, where a value of -1 indicates a perfect negative correlation, 0 indicates no correlation, and +1 indicates a perfect positive correlation. LPS, lipopolysaccharide; IP, intraperitoneal.

Discussion

Effective drug treatments targeting the SARS-CoV-2 immunopathological process are of particular interest to reduce the disease burden caused by these infections (1). Metformin, a widely available drug treatment for diabetes, was previously reported to have a cytokine-lowering effect in both diabetic and non-diabetic patients (24). In patients with COVID-19, this effect could be critical as the development of cytokine storm can lead to very lethal outcomes such as ARDS (31). The effect of metformin on the production of cytokines is reported to be caused by blocking the AMPK/mTOR cytokine receptor pathway, which results in a decrease in the expression of certain proinflammatory genes, such as IL-1α, IL-1β, IL-2, IL-6, IL-12 and TNF-α (15). Additional studies suggest that AMPK-independent mechanisms, such as altering the gut microbiota, may also be involved in this process (32-34).

Cytokine storm causes severe illness in patients with COVID-19, thereby significantly increasing the morbidity and mortality rates by ~5% (31). The present study sought to assess the anti-inflammatory effect of metformin in a mouse model of LPS-induced cytokine storm. The present results demonstrated that LPS injection caused a significant increase in serum IL-1β levels when compared with the control group. Oral and IP metformin significantly reduced the elevated IL-1β levels in BALB/c mice 1 h after IP LPS injection, with no significant difference demonstrated in efficacy between the two routes of metformin administration. These findings support previous reports that metformin is a potent inhibitor of the chronic inflammatory response. For example, metformin therapy has been reported to reduce reactive oxygen species and hypoxia-inducible factor-1 (HIF-1) levels, which in turn reduce the IL-1β expression levels after prolonged exposure to proinflammatory LPS stimuli (29,35). Additionally, it has been previously established that the IL-1β mediated inflammatory response is involved in COVID-19 pathogenesis (36). Therefore, lowering IL-1β levels could reduce inflammation and mortality in patients with COVID-19(37).

The present study demonstrated that LPS injection resulted in a significant increase in serum IL-6 levels in all treatment groups of animals tested. Pretreatment with either oral or IP metformin significantly reduced this increase in IL-6 levels. IP metformin was significantly more effective at reducing IL-6 levels compared with oral metformin. Hyun et al (38) reported that metformin reduces IL-1, IL-6 and TNF-α production at both the protein and mRNA level in a dose-dependent manner. Similarly, Chao et al (36) reported that metformin reduces the LPS-induced release of IL-6 in mouse livers. Additionally, metformin inhibits the acute inflammatory response in two macrophage-like cell lines by activating AMPK, but not HIF-1 or IL-10(39).

TNF-α is an inflammatory cytokine produced in response to bacterial and viral infections and can elicit tissue damage and fibrosis (27). The present study demonstrated that serum TNF-α levels were significantly increased after mice were injected with LPS compared with the controls. Oral or IP metformin were equally effective in attenuating the significantly elevated TNF-α levels, with no significant differences demonstrated between either route of administration. Kim et al (30) previously reported that oral administration of metformin to mice treated with LPS reduced the plasma, spleen and lungs tissue levels of both TNF-α and IL-6 leading to a reduction in the effect of LPS-induced inflammation.

Cho et al (27) investigated the anti-inflammatory effects of metformin on LPS-induced inflammation in human middle ear epithelial cell lines. LPS was found to elevate TNF-α and cyclooxygenase-2 levels. However, pretreatment with metformin reduced the production of these inflammatory factors. Metformin also decreased the production of sepsis-induced brain injury in mice inflammatory cytokines, such as IL-6, IL-1 β and TNF-α (29). These findings are consistent with the findings of the present study.

The present study demonstrated a non-significant increase in serum IL-17 levels in mice pretreated with LPS. Neither IP nor oral metformin had a significant effect on serum IL-17 levels. One possible explanation for these findings is that the effect of LPS is likely to be dose- or time-dependent. Sun et al (40) reported that IL-17A is upregulated in LPS-induced neuroinflammation and cognitive impairment in aged rats. Differences in the findings of the present study regarding IL-17 levels could be due to the time frame of sample collection following LPS injection. It could be possible that 1 h may not be sufficient time to demonstrate a significant increase in serum IL-17 levels. A previous study reported that whilst IL-6 and TNF-α levels peak within 1 h of LPS injection in mice, changes in IL-17 levels are slower and peak over 8 h (41).

Age-related increases in serum levels of IL-17A, IL-17F, IL-21 and IL-6 can be prevented by metformin treatment (42). In a mouse model of scleroderma treated with metformin, Moon et al (42) reported a decrease in the production of the proinflammatory cytokine IL-17 in dermal tissues and lymphocytes in a mice model of bleomycin-induced scleroderma. The pathophysiology of IL-17 has been studied in mice challenged with LPS, and it was reported that increased amounts of IL-17 are associated with excessive lung injury and inflammation (43). Neutralizing increased IL-17 production using anti-IL-17 antibodies improves survival and reduces lung injury (43).

Low-grade inflammation caused by microbiota dysbiosis is thought to promote the occurrence of metabolic syndrome (44,45). The gut microbiota may be involved in the regulation of immunity, inflammation and autoinflammatory diseases such as multiple sclerosis and osteomyelitis (46,47). Several studies have reported that metformin can influence the composition of the intestinal microbiota in certain clinical situations such as dysbiosis, intestinal inflammations disorders, and T2D (48-50). It has been reported that the ability of metformin to alleviate certain inflammatory diseases, such as inflammatory bowel disease is linked to its ability to modify the diversity of gut microbiota (51). By contrast, it has also been proposed that the ability of metformin to reduce indices of low-grade inflammation in metabolic syndrome is independent of its effect on the gut microbiota (52,53).

A limitation of the present study is that further studies are required to determine whether metformin has a direct or indirect effect on cultured immune system cells, such as macrophages and T cells. Investigating a potential dose-dependent response of both LPS and metformin and longer time scales between LPS injection and sample collection could also further elucidate the impact of metformin in the mouse model used in the present study.

In the present study, a mouse model of LPS-induced cytokine storm was pretreated with metformin, which was demonstrated to suppress the release of the pro-inflammatory cytokines IL-1β, IL-6, IL-17 and TNF-α. Certain serum cytokine levels demonstrated a positive correlation with other cytokines in the mouse model. These findings may potentially suggest a future role for metformin in the treatment of human diseases associated with the cytokine storm.

Acknowledgements

Not applicable.

Funding

Funding: The present study was funded by the Deanship of Scientific Research at Jouf University (grant no. DSR 2020-04-2543).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

All of the authors have made substantial contributions towards the completion of the present study. MA and AET conceived the present study, performed the experiments, were project administrators and prepared the draft manuscript. IAT, EAEM, MA and AET collected the data, obtained resources, performed data analysis and critically reviewed and edited the manuscript. IAT and EAEM acquired funding. IAT, MA and AET supervised the project. IAT and AET confirm the authenticity of the raw data. All authors read and approved the final version of the manuscript.

Ethics approval and consent to participate

Ethical approval was obtained from the Local Committee of Bioethics of Jouf University (approval no. 07-08-42; Sakaka, Saudi Arabia).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Catanzaro M, Fagiani F, Racchi M, Corsini E, Govoni S and Lanni C: Immune response in COVID-19: Addressing a pharmacological challenge by targeting pathways triggered by SARS-CoV-2. Signal Transduct Target Ther. 5(84)2020.PubMed/NCBI View Article : Google Scholar

2 

Yuan Y, Jiao B, Qu L, Yang D and Liu R: The development of COVID-19 treatment. Front Immunol. 14(1125246)2023.PubMed/NCBI View Article : Google Scholar

3 

Pfortmueller CA, Spinetti T, Urman RD, Luedi MM and Schefold JC: COVID-19-associated acute respiratory distress syndrome (CARDS): Current knowledge on pathophysiology and ICU treatment-A narrative review. Best Pract Res Clin Anaesthesiol. 35:351–368. 2021.PubMed/NCBI View Article : Google Scholar

4 

Huang C, Wang Y, Li X, Ren L, Zhao J, Hu Y, Zhang L, Fan G, Xu J, Gu X, et al: Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet. 395:497–506. 2020.PubMed/NCBI View Article : Google Scholar

5 

Crayne CB, Albeituni S, Nichols KE and Cron RQ: The immunology of macrophage activation syndrome. Front Immunol. 10(119)2019.PubMed/NCBI View Article : Google Scholar

6 

Kindler E, Thiel V and Weber F: Interaction of SARS and MERS coronaviruses with the antiviral interferon response. Adv Virus Res. 96:219–243. 2016.PubMed/NCBI View Article : Google Scholar

7 

Min CK, Cheon S, Ha NY, Sohn KM, Kim Y, Aigerim A, Shin HM, Choi JY, Inn KS, Kim JH, et al: Comparative and kinetic analysis of viral shedding and immunological responses in MERS patients representing a broad spectrum of disease severity. Sci Rep. 6(25359)2016.PubMed/NCBI View Article : Google Scholar

8 

Channappanavar R and Perlman S: Pathogenic human coronavirus infections: Causes and consequences of cytokine storm and immunopathology. Semin Immunopathol. 39:529–539. 2017.PubMed/NCBI View Article : Google Scholar

9 

Liang Y, Wang ML, Chien CS, Yarmishyn AA, Yang YP, Lai WY, Luo YH, Lin YT, Chen YJ, Chang PC and Chiou SH: Highlight of immune pathogenic response and hematopathologic effect in SARS-CoV, MERS-CoV, and SARS-Cov-2 Infection. Front Immunol. 11(1022)2020.PubMed/NCBI View Article : Google Scholar

10 

Yao Z, Zheng Z, Wu K and Junhua Z: Immune environment modulation in pneumonia patients caused by coronavirus: SARS-CoV, MERS-CoV and SARS-CoV-2. Aging (Albany NY). 12:7639–7651. 2020.PubMed/NCBI View Article : Google Scholar

11 

Mehta P, McAuley DF, Brown M, Sanchez E, Tattersall RS and Manson JJ: HLH Across Speciality Collaboration, UK. COVID-19: Consider cytokine storm syndromes and immunosuppression. Lancet. 395:1033–1034. 2020.PubMed/NCBI View Article : Google Scholar

12 

Al-Samkari H and Berliner N: Hemophagocytic lymphohistiocytosis. Annu Rev Pathol. 13:27–49. 2018.PubMed/NCBI View Article : Google Scholar

13 

Li X, Geng M, Peng Y, Meng L and Lu S: Molecular immune pathogenesis and diagnosis of COVID-19. J Pharm Anal. 10:102–108. 2020.PubMed/NCBI View Article : Google Scholar

14 

Sarzi-Puttini P, Giorgi V, Sirotti S, Marotto D, Ardizzone S, Rizzardini G, Antinori S and Galli M: COVID-19, cytokines and immunosuppression: What can we learn from severe acute respiratory syndrome? Clin Exp Rheumatol. 38:337–342. 2020.PubMed/NCBI View Article : Google Scholar

15 

Chung MM, Nicol CJ, Cheng YC, Lin KH, Chen YL, Pei D, Lin CH, Shih YN, Yen CH, Chen SJ, et al: Metformin activation of AMPK suppresses AGE-induced inflammatory response in hNSCs. Exp Cell Res. 352:75–83. 2017.PubMed/NCBI View Article : Google Scholar

16 

Yin Y, Choi SC, Xu Z, Perry DJ, Seay H, Croker BP, Sobel ES, Brusko TM and Morel L: Normalization of CD4+ T cell metabolism reverses lupus. Sci Transl Med. 7(274ra18)2015.PubMed/NCBI View Article : Google Scholar

17 

Lee SY, Moon SJ, Kim EK, Seo HB, Yang EJ, Son HJ, Kim JK, Min JK, Park SH and Cho ML: Metformin suppresses systemic autoimmunity in roquin san/san mice through inhibiting B cell differentiation into plasma cells via regulation of AMPK/mTOR/STAT3. J Immunol. 198:2661–2670. 2017.PubMed/NCBI View Article : Google Scholar

18 

Jing Y, Wu F, Li D, Yang L, Li Q and Li R: Metformin improves obesity-associated inflammation by altering macrophages polarization. Mol Cell Endocrinol. 461:256–264. 2018.PubMed/NCBI View Article : Google Scholar

19 

Kim EK, Lee SH, Lee SY, Kim JK, Jhun JY, Na HS, Kim SY, Choi JY, Yang CW, Park SH and Cho ML: Metformin ameliorates experimental-obesity-associated autoimmune arthritis by inducing FGF21 expression and brown adipocyte differentiation. Exp Mol Med. 50(e432)2018.PubMed/NCBI View Article : Google Scholar

20 

Schuiveling M, Vazirpanah N, Radstake TRDJ, Zimmermann M and Broen JCA: Metformin, a new era for an old drug in the treatment of immune mediated disease? Curr Drug Targets. 19:945–959. 2018.PubMed/NCBI View Article : Google Scholar

21 

Ba W, Xu Y, Yin G, Yang J, Wang R, Chi S, Wang Y and Li C: Metformin inhibits pro-inflammatory responses via targeting nuclear factor-κB in HaCaT cells. Cell Biochem Funct. 37:4–10. 2019.PubMed/NCBI View Article : Google Scholar

22 

Jang SG, Lee J, Hong SM, Kwok SK, Cho ML and Park SH: Metformin enhances the immunomodulatory potential of adipose-derived mesenchymal stem cells through STAT1 in an animal model of lupus. Rheumatology (Oxford). 59:1426–1438. 2020.PubMed/NCBI View Article : Google Scholar

23 

Sciannimanico S, Grimaldi F, Vescini F, De Pergola G, Iacoviello M, Licchelli B, Guastamacchia E, Giagulli VA and Triggiani V: Metformin: Up to date. Endocr Metab Immune Disord Drug Targets. 20:172–181. 2020.PubMed/NCBI View Article : Google Scholar

24 

Saisho Y: Metformin and inflammation: Its potential beyond glucose lowering effect. Endocr Metab Immune Disord Drug Targets. 15:196–205. 2015.PubMed/NCBI View Article : Google Scholar

25 

Ismaiel AA, Espinosa-Oliva AM, Santiago M, García-Quintanilla A, Oliva-Martín MJ, Herrera AJ, Venero JL and de Pablos RM: Metformin, besides exhibiting strong in vivo anti-inflammatory properties, increases mptp-induced damage to the nigrostriatal dopaminergic system. Toxicol Apple Pharmacol. 298:19–30. 2016.PubMed/NCBI View Article : Google Scholar

26 

Koh SJ, Kim JM, Kim IK, Ko SH and Kim JS: Anti-inflammatory mechanism of metformin and its effects in intestinal inflammation and colitis-associated colon cancer. J Gastroenterol Hepatol. 29:502–510. 2014.PubMed/NCBI View Article : Google Scholar

27 

Cho JG, Song JJ, Choi J, Im GJ, Jung HH and Chae SW: The suppressive effects of metformin on inflammatory response of otitis media model in human middle ear epithelial cells. Int J Pediatr Otorhinolaryngol. 89:28–32. 2016.PubMed/NCBI View Article : Google Scholar

28 

Park CS, Bang BR, Kwon HS, Moon KA, Kim TB, Lee KY, Moon HB and Cho YS: Metformin reduces airway inflammation and remodeling via activation of AMP-activated protein kinase. Biochem Pharmacol. 84:1660–1670. 2012.PubMed/NCBI View Article : Google Scholar

29 

Tang G, Yang H, Chen J, Shi M, Ge L, Ge X and Zhu G: Metformin ameliorates sepsis-induced brain injury by inhibiting apoptosis, oxidative stress and neuroinflammation via the PI3K/Akt signaling pathway. Oncotarget. 8:97977–97989. 2017.PubMed/NCBI View Article : Google Scholar

30 

Kim J, Kwak HJ, Cha JY, Jeong YS, Rhee SD, Kim KR and Cheon HG: Metformin suppresses lipopolysaccharide (LPS)-induced inflammatory response in murine macrophages via activating transcription factor-3 (ATF-3) induction. J Biol Chem. 289:23246–23255. 2014.PubMed/NCBI View Article : Google Scholar

31 

Montazersaheb S, Hosseiniyan Khatibi SM, Hejazi MS, Tarhriz V, Farjami A, Ghasemian Sorbeni F, Farahzadi R and Ghasemnejad T: COVID-19 infection: An overview on cytokine storm and related interventions. Virol J. 19(92)2022.PubMed/NCBI View Article : Google Scholar

32 

Zhao X, Cao F, Liu Q, Li X, Xu G, Liu G, Zhang Y, Yang X, Yi S, Xu F, et al: Behavioral, inflammatory and neurochemical disturbances in LPS and UCMS-induced mouse models of depression. Behav Brain Res. 364:494–502. 2019.PubMed/NCBI View Article : Google Scholar

33 

Kelly B, Tannahill GM, Murphy MP and O'Neill LA: Metformin inhibits the production of reactive oxygen species from NADH: Ubiquinone oxidoreductase to limit induction of interleukin-1β (IL-1β) and boosts interleukin-10 (IL-10) in lipopolysaccharide (LPS)-activated macrophages. J Biol Chem. 290:20348–20359. 2015.PubMed/NCBI View Article : Google Scholar

34 

Dehkordi EH, Sattari F, Khoshdel A and Kasiri K: Effect of folic acid and metformin on insulin resistance and inflammatory factors of obese children and adolescents. J Res Med Sci. 21(71)2016.PubMed/NCBI View Article : Google Scholar

35 

Dias SSG, Soares VC, Ferreira AC, Sacramento CQ, Fintelman-Rodrigues N, Temerozo JR, Teixeira L, Nunes da Silva MA, Barreto E, Mattos M, et al: Lipid droplets fuel SARS-CoV-2 replication and production of inflammatory mediators. PLoS Pathog. 16(e1009127)2020.PubMed/NCBI View Article : Google Scholar

36 

Chao L, Hui-Jie G, Fang Y, Ni K, Bao-Kub W and Teng-yuan Z: Anti-inflammatory effect of metformin LPS-induced inflammation in mice. Basic Clin Med. 39(1001-6325-1248-04)2019.

37 

Cavalli G, Larcher A, Tomelleri A, Campochiaro C, Della-Torre E, De Luca G, Farina N, Boffini N, Ruggeri A, Poli A, et al: Interleukin-1 and interleukin-6 inhibition compared with standard management in patients with COVID19 and hyperinfammation: A cohort study. Lancet Rheumatol. 3:E253–E261. 2021.PubMed/NCBI View Article : Google Scholar

38 

Hyun B, Shin S, Lee A, Lee S, Song Y, Ha NJ, Cho KH and Kim K: Metformin Down-regulates TNF-α secretion via suppression of scavenger receptors in macrophages. Immune Netw. 13:123–132. 2013.PubMed/NCBI View Article : Google Scholar

39 

Postler TS, Peng V, Bhatt DM and Ghosh S: Metformin selectively dampens the acute inflammatory response through an AMPK-dependent mechanism. Sci Rep. 11(18721)2021.PubMed/NCBI View Article : Google Scholar

40 

Sun J, Zhang S, Zhang X, Zhang X, Dong H and Qian Y: IL-17A is implicated in lipopolysaccharide-induced neuroinflammation and cognitive impairment in aged rats via microglial activation. J Neuroinflammation. 12(165)2015.PubMed/NCBI View Article : Google Scholar

41 

Furuya S, Kono H, Hara M, Hirayama K, Sun C and Fujii H: Interleukin 17A plays a role in lipopolysaccharide/D-galactosamine-induced fulminant hepatic injury in mice. J surg Res. 199:487–493. 2015.PubMed/NCBI View Article : Google Scholar

42 

Moon J, Lee SY, Choi JW, Lee AR, Yoo JH, Moon SJ, Park SH and Cho ML: Metformin ameliorates scleroderma via inhibiting Th17 cells and reducing mTOR-STAT3 signaling in skin fibroblasts. J Transl Med. 19(192)2021.PubMed/NCBI View Article : Google Scholar

43 

Li Q, Gu Y, Tu Q, Wang K, Gu X and Ren T: Blockade of Interleukin-17 restrains the development of acute lung injury. Scand J Immunol. 83:203–211. 2016.PubMed/NCBI View Article : Google Scholar

44 

Carvalho FA, Aitken JD, Vijay-Kumar M and Gewirtz AT: Toll-like receptor-gut microbiota interactions: Perturb at your own risk! Annu Rev. Physiol. 74:177–198. 2012.PubMed/NCBI View Article : Google Scholar

45 

Rodriguez J, Hiel S and Delzenne NM: Metformin: Old friend, new ways of action-implication of the gut microbiome? Curr Opin Clin Nutr Metab Care. 21:294–301. 2018.PubMed/NCBI View Article : Google Scholar

46 

Lee YK, Menezes JS, Umesaki Y and Mazmanian SK: Proinflammatory T-cell responses to gut microbiota promote experimental autoimmune encephalomyelitis. Proc Natl Acad Sci USA. 108 (Suppl 1):S4615–S4622. 2011.PubMed/NCBI View Article : Google Scholar

47 

Lukens JR, Gurung P, Vogel P, Johnson GR, Carter RA, McGoldrick DJ, Bandi SR, Calabrese CR, Vande Walle L, Lamkanfi M and Kanneganti TD: Dietary modulation of the microbiome affects autoinflammatory disease. Nature. 516:246–249. 2014.PubMed/NCBI View Article : Google Scholar

48 

Elbere I, Kalnina I, Silamikelis I, Konrade I, Zaharenko L, Sekace K, Radovica-Spalvina I, Fridmanis D, Gudra D, Pirags V and Klovins J: Association of metformin administration with gut microbiome dysbiosis in healthy volunteers. PLoS One. 13(e0204317)2018.PubMed/NCBI View Article : Google Scholar

49 

Zhang W, Xu JH, Yu T and Chen QK: Effects of berberine and metformin on intestinal inflammation and gut microbiome composition in db/db mice. Biomed Pharmacother. 118(109131)2019.PubMed/NCBI View Article : Google Scholar

50 

Zhang Q and Hu N: Effects of metformin on the gut microbiota in obesity and type 2 diabetes mellitus. Diabetes Metab Cinder Obes. 13:5003–5014. 2020.PubMed/NCBI View Article : Google Scholar

51 

Liu Z, Liao W, Zhang Z, Sun R, Luo Y, Chen Q, L X, Lu R and Ying Y: Metformin affects gut microbiota composition and diversity associated with amelioration of dextran sulfate sodium-induced colitis in mice. Front Pharmacol. 12(640347)2021.PubMed/NCBI View Article : Google Scholar

52 

Adeshirlarijaney A, Zou J, Tran HQ, Chassaing B and Gewirtz AT: Amelioration of metabolic syndrome by metformin associates with reduced indices of low-grade inflammation independently of the gut microbiota. Am J Physiol Endocrinol Metab. 317:E1121–E1130. 2019.PubMed/NCBI View Article : Google Scholar

53 

Petrakis D, Margină D, Tsarouhas K, Tekos F, Stan M, Nikitovic D, Kouretas D, Spandidos DA and Tsatsakis A: Obesity a risk factor for increased COVID19 prevalence, severity and lethality (Review). Mol Med Rep. 22:9–19. 2020.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

September-2023
Volume 26 Issue 3

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Taher I, El‑Masry E, Abouelkheir M and Taha AE: Anti‑inflammatory effect of metformin against an experimental model of LPS‑induced cytokine storm. Exp Ther Med 26: 415, 2023
APA
Taher, I., El‑Masry, E., Abouelkheir, M., & Taha, A.E. (2023). Anti‑inflammatory effect of metformin against an experimental model of LPS‑induced cytokine storm. Experimental and Therapeutic Medicine, 26, 415. https://doi.org/10.3892/etm.2023.12114
MLA
Taher, I., El‑Masry, E., Abouelkheir, M., Taha, A. E."Anti‑inflammatory effect of metformin against an experimental model of LPS‑induced cytokine storm". Experimental and Therapeutic Medicine 26.3 (2023): 415.
Chicago
Taher, I., El‑Masry, E., Abouelkheir, M., Taha, A. E."Anti‑inflammatory effect of metformin against an experimental model of LPS‑induced cytokine storm". Experimental and Therapeutic Medicine 26, no. 3 (2023): 415. https://doi.org/10.3892/etm.2023.12114