Skip to main content

The relationship between the systemic immune inflammation index and the nonalcoholic fatty liver disease in American adolescents

Abstract

Background

Non-alcoholic fatty liver disease (NAFLD) is a growing health crisis in the general population of the United States (U.S.), but the relationship between systemic immune-inflammation (SII) index and NAFLD is not known.

Methods

We collected data from the National Health and Nutrition Examination Survey 2017–2018. Next, propensity score matching (PSM), collinearity analysis, restricted cubic spline (RCS) plot, logistic regression, quantile regression analysis, subgroup analysis, mediation analysis, and population attributable fraction were used to explore the association of the SII with risk of NAFLD.

Results

A total of 665 participants including the 532 Non-NAFLD and 133 NAFLD were enrolled for further analysis after PSM analysis. The RCS results indicated that there was a linear relationship between the SII and controlled attenuation parameter (p for nonlinear = 0.468), the relationship also existed after adjustment for covariates (p for nonlinear = 0.769). The logistic regression results indicated that a high SII level was an independent risk factor for NAFLD (OR = 3.505, 95% CI: 1.092–11.249, P < 0.05). The quantile regression indicated that at higher quantiles (0.90, and 0.95) the SII was significantly associated with NAFLD (p < 0.05). Mediation analysis indicated that alanine aminotransferase (ALT), triglycerides, and blood urea nitrogen (BUN) were partially contribute to the relationship between SII and NAFLD. The population attributable fractions indicated that 23.19% (95% CI: 8.22%, 38.17%) of NAFLD cases could be attributed to SII corresponding to 133 NAFLD cases.

Conclusion

There was a positive linear relationship between the SII and the risk of NAFLD. The ALT, triglycerides, and BUN had a partial mediating effect on the relationship between the SII and NAFLD.

Peer Review reports

Introduction

Nonalcoholic fatty liver disease (NAFLD) has become a significant public health concern due to its increasing prevalence and associated health risks. NAFLD is a spectrum of liver disorders characterized by excessive fat accumulation in the liver, which can progress to nonalcoholic steatohepatitis (NASH), fibrosis, cirrhosis, and ultimately hepatocellular carcinoma. The global prevalence of NAFLD is estimated to be around 25% [1] and among individuals with obesity, diabetes, and metabolic syndrome [2]. NAFLD also poses a serious threat to the health of adolescents. According to relevant literature, the prevalence of NAFLD among adolescents is continuously increasing. The global prevalence of NAFLD in adolescents rose from 3.73 to 4.71% in the last 20 years [3, 4]. Compared to healthy adolescents, children with this disease are more prone to cardiovascular diseases and metabolic syndrome, significantly impacting their quality of life [5, 6]. Therefore, the prevention and treatment of NAFLD in adolescents should receive adequate attention.

NAFLD is closely associated with inflammation. levels of inflammatory factors such as IL-1β, IL-6, and IL-17 are significantly increased in obese children with NAFLD [7]. The severity and composition of inflammatory infiltration are related to the steatosis and severity of NAFLD in children [8]. The Systemic Immune Inflammation (SII) index is a novel biomarker that reflects the systemic immune response and inflammation in various diseases. It is calculated based on the peripheral blood cell counts, including platelets, neutrophils, and lymphocytes, and is associated with the prognosis and outcomes of several diseases such as cancer, infectious diseases, and cardiovascular diseases [9,10,11,12]. The association of the SII with NAFLD has gained increasing attention due to the potential role of inflammation in NAFLD pathogenesis and progression [13]. Previous studies have suggested that elevated SII levels are associated with the severity of liver fibrosis in NAFLD patients [12]. Furthermore, SII has been proposed as a potential prognostic marker for NAFLD-related complications, such as hepatocellular carcinoma and cardiovascular events [13]. However, most of these studies have been conducted on adult patients with NAFLD, and the situation among adolescents is not well understood. Therefore, investigating the relationship between SII and NAFLD in adolescents may provide valuable insights into the underlying mechanisms of NAFLD progression and identify potential therapeutic targets.

In this cross-sectional study, we aim to investigate the correlation between SII and NAFLD in American adolescents, as well as its potential value in predicting NAFLD using the participant’s related information from the National Health and Nutrition Examination Survey (NHANES).

Methods

Data source and study participants

Data were collected from the 2017–2018 survey cycles of NHANES. It is a program of studies designed to assess the health and nutritional status of individuals in the United States and is conducted by the National Center for Health Statistics which is a part of the Centers for Disease Control and Prevention (CDC). The survey collects data through interviews, physical examinations, and laboratory tests, and is a valuable resource for studying various health and nutrition-related outcomes. 9254 participants were collected. Of these participants, we excluded 3306 without the Vibration Controlled Transient Elastography (VCTE) data. Notable, the participants aged > 12 years old were allowed to undergo VCTE. Next, the 456 participants’ elastography examination status was ineligible, not performed, or partial, which were also excluded. We further excluded 43 with serologic positivity for viral hepatitis, 592 with 4 or 5 drinks a day, 205 without SII data, and 3691 with age > 20. 961 participants including 828 Non-NAFLD and 133 NAFLD remained and were used for performed PSM analysis. Finally, 665 participants (12 years old < age < 20 years old) including 532 Non-NAFLD and 133 NAFLD were enrolled in the study.

Diagnosis of nonalcoholic fatty liver disease

Controlled attenuation parameter (CAP) is a non-invasive technique used to assess hepatic steatosis by measuring the ultrasound attenuation in the liver. CAP has been shown to have good diagnostic accuracy for detecting hepatic steatosis in NAFLD patients [14]. The median CAP was categorized using 285 dB/m as a threshold for diagnosing liver steatosis, with optimal diagnostic performance [15].

Calculation of systemic immune-inflammation index

The systemic immune-inflammation index was calculated based on the Lymphocyte, neutrophil, and platelet counts by the following formula: SII= (platelet count × neutrophils count)/lymphocytes count. The three kinds of cells were measured via automated hematology analyzing devices and their count was expressed as x103cells/µL [16].

Covariates

This study enrolled some potential covariates that affect the relationship between the SII and NAFLD. These covariates included age, gender, asthma, smoke, physical activity, poverty income ratio, body mass index (BMI, kg/m2), alanine aminotransferase (ALT, U/L), alkaline phosphatase, aspartate aminotransferase (AST, U/L), blood urea nitrogen (BUN, mg/dL), creatinine, glucose, triglycerides, uric acid, and Non-high density lipoprotein (Non-HDL) cholesterol.

Statistical analyses

Propensity score matching (PSM) was used to eliminate bias and control for potential confounding variables. The SII was normalized by log due to the relatively large data range. The Mann-Whitney was used to compare differences in continuous characteristics between the Non-NAFLD and NAFLD groups, and the chi-square test was used to compare differences in categoric characteristics between two groups. The collinearity analysis was performed to evaluate and exclude variables with collinearity and variance inflation factor (VIF) > 5 considered to be collinear [17]. The Restricted cubic spline (RCS) curve was plotted to explore the relationship between the SII index with the NAFLD. Logistic regression analysis was used to explore the relationship between the SII with NAFLD. Quantile regression analysis was conducted to fit the correlation between SII and NAFLD better. We conducted subgroups to explore the relationship between SII and NAFLD in different subgroups via logistic regression. Stratification factors contained gender (male/female), race (Hispanic/Non-Hispanic White/Non-Hispanic Black/other races) asthma (yes/no), and physical activity (yes/no). Moreover, the interaction analysis was conducted to explore the differences between the subgroups. The mediation analysis explored the underlay mediate variable effect on the relationship between the SII and NAFLD. Population attributable fraction was conducted to quantify the NAFLD burden attributable to SII. All statistical analyses were performed using R 4.1.0, with two-tailed P < 0.05 indicating statistical significance.

Results

Participant characteristics

A total of 961 participants were included, of which 828 were Non-NAFLD, and 133 were NAFLD. There were significant differences in age, gender, physical activity, poverty income ratio, BMI, ALT, AST, triglycerides, uric acid, Non-HDL cholesterol, and Log SII between the Non-NAFLD group and NAFLD group (all p < 0.05). Then the PSM was performed based on 4:1, and finally, the 532 Non-NAFLD and 133 NAFLD were selected for further analysis. The results showed that there were significant differences in ALT, AST, triglycerides, Non-HDL cholesterol, body mass index, uric acid, Log SII, BUN, and poverty income ratio between the Non-NAFLD group and NAFLD group (Table 1, all p < 0.05). Triglycerides, uric acid, Non-HDL cholesterol, Log SII, and AST in Non-NAFLD group were significantly lower than those in the NAFLD group, while blood urea nitrogen in Non-NAFLD group was significantly higher than in the NAFLD group.

Table 1 Patient characteristics

SII is an independent risk factor for NAFLD

To acquire the precise relationship between the SII and NAFLD, the collinearity analysis was performed before the RCS analysis. The collinearity analysis results showed no collinear relationships among the variables which had differences with statistical significance between the Non-NAFLD and NAFLD and obtained from Table 2(Table 2, all VIF < 5. Further, the RCS results indicated that there was a linear relationship between the SII and CAP (Fig. 1A, P for nonlinear = 0.468), the relationship also existed after adjustment for ALT, AST, triglycerides, Non-HDL cholesterol, uric acid, BUN, and poverty income ratio (Fig. 1B, P for nonlinear = 0.769). Therefore, the logistic regression was performed. The results of the univariate logistic regression indicated that high levels of SII were a risk factor for NAFLD (OR = 6.415, 95% CI: 2.655–15.504, P < 0.001, Table 3). After adjustment for significant factors identified in the baseline characteristics including ALT, AST, triglycerides, Non-HDL cholesterol, uric acid, BUN, and poverty income ratio, high SII level was an independent risk factor for NAFLD (OR = 3.505, 95% CI: 1.092–11.249, P < 0.05, Table 3).

Table 2 Collinearity analysis
Fig. 1
figure 1

Restricted cubic spline (RCS) curve for the relationship between the SII index with the controlled attenuation parameter (CAP) in patients with NAFLD. (A) RCS model (B) RCS model after adjusting for ALT, triglycerides, Non-HDL cholesterol, body mass index, uric acid, BUN, and poverty income ratio

Table 3 Logistic regression analysis of the association of SII with nonalcoholic fatty liver disease

For further exploring the relationship between the SII and NAFLD, the quantile regression was fitted on the 0.05,0.10,0.25,0.75,0.9,0.95 of SII distribution to investigate the relationship between the CAP and SII. The results indicated that at higher quantiles (0.90, and 0.95) the SII was significantly associated with NAFLD (Table 4p < 0.05).

Table 4 Quantile regression analysis of the association of SII with nonalcoholic fatty liver disease

Subgroup analysis

To further the relation between the SII and NAFLD, a subgroup analysis was performed, which was stratified by gender, race, asthma, and physical activity. We selected those four factors for further subgroup analyses because they were less studied, but important for NAFLD based on reported literatures [18,19,20]. The results indicated that SII was a risk factor for NAFLD events in male/female NAFLD patients, Non-Hispanic Black patients, patients with/without asthma, and patients without physical activity (p < 0.05, Table 5). Moreover, the interaction analysis showed that there was no statistical significance for gender, race, asthma, and physical activity (all P for interactions > 0.05, Table 5).

Table 5 Association of SII with nonalcoholic fatty liver disease in different subgroups

Mediation analyses and population attributable fraction

The multivariable logistic regression results showed that ALT, triglycerides, and BUN were independent risk factors for NAFLD. Mediation analysis was performed to further explore their function in the NAFLD. The results indicated that three variables partially contributed to the relationship between SII and NAFLD. ALT, triglycerides, and BUN mediated 19.71%,18.22%, and 9.84% of the effect of SII on NAFLD (Fig. 2A-C). Attributable cases (AT) and population attributable fractions (PAF) showed NAFLD burden attributable to SII (Table 6). 23.19% (95% CI: 8.22%, 38.17%) of NAFLD cases can be attribute to SII corresponding to 133 NAFLD cases.

Fig. 2
figure 2

The association of the SII with mediation variables and NAFLD. (A) ALT was the mediation variable. (B) The Triglycerides was the mediation variable. (C) The BUN was the mediation variable

Table 6 Estimated burden of NAFLD attributable to SII among adolescents

Discussion

In this study, we found that there was a positive linear relationship between the SII and NAFLD in American adolescents. In addition, ALT, triglycerides, and BUN had a partial mediating effect on the relationship between the SII and NAFLD.

The pathogenesis of NAFLD is a complex process involving the interaction of multiple factors such as genetics, environment, and metabolic abnormalities. Most related studies have shown that the mechanisms are different and include [1] Disorders of fat metabolism and Inflammatory response: The main feature of NAFLD is the accumulation of lipids in the liver, mainly triacylglycerol. This may be due to metabolic abnormalities in the synthesis and transport of fats in the liver. Fat accumulation triggers an inflammatory response in the liver. Activation of inflammatory cells and cytokines leads to inflammatory responses in liver tissue, including hepatocyte injury and apoptosis [21, 22]. [2] Oxidative stress and mitochondrial dysfunction: Oxidative stress and mitochondrial dysfunction also play an important role in the development of NAFLD. Cellular damage caused by lipid peroxidation and oxidative stress may increase the risk of NAFLD progression [23, 24]. [3] Liver fibrosis and scarring: Fibrosis is the excessive accumulation of collagen in liver tissue, and scarring is when fibrous tissue replaces normal liver tissue. [4] Interaction of genes with environmental factors: Genetic and environmental factors are also thought to have an impact on the development and progression of NAFLD. Some genetic mutations may increase the risk of developing NAFLD, while environmental factors such as diet, lifestyle, obesity, and metabolic syndrome increase the risk of developing NAFLD [25].

The “multiple-hit model” is currently the widely accepted theory for NAFLD in adolescents [26]. This model suggests that NAFLD results from the interplay of genetic and environmental dietary factors, along with crosstalk between various organs and tissues, leading to extensive metabolic dysfunction. The initial step in NAFLD for both adults and adolescents involves triglyceride accumulation and insulin resistance (IR) [27,28,29,30]. Our study indicates that triglyceride levels are significantly higher in NAFLD patients compared to those without the condition. Triglycerides are formed by the esterification of glycerol and free fatty acids (FFAs). The accumulation of triglycerides suggests an excess of FFAs in the liver, which, when activated by acetyl-CoA synthase, may trigger esterification or β-oxidation pathways. This can lead to oxidative stress, generating reactive oxygen species and resulting in mitochondrial dysfunction [31, 32]. This dysfunction activates inflammatory pathways such as JNK-AP-1 and IKK-NF-κBD [33]. The production of inflammatory cytokines can induce IR. Hepatic IR prevents insulin from inhibiting lipolysis, leading to reduced fatty acids [34], lower hepatic acetyl-CoA levels, and reduced pyruvate carboxylase activity, thereby inhibiting the conversion of pyruvate to glucose [35]. These processes activate the production of pro-inflammatory cytokines in the liver, exposing it to high levels of these cytokines and leading to histological changes similar to non-alcoholic steatohepatitis [36].

SII is reminiscent of the interaction of thrombocytosis, inflammation, and immunity. Song et al. indicated that SII could be used as a simple and affordable way to identify hepatic steatosis because there was a positive relationship between SII and hepatic steatosis [37]. The pathological progression of NAFLD follows three steps including steatosis, lipotoxicity, and inflammation [38]. In our study, we found that the patients with high SII had a high risk of NAFLD. SII, secreted by lymphocytes and central granulocytes, leads to NAFLD through the release of inflammatory factors such as Tumor Necrosis Factor-alpha (TNF-alpha), Interferon-gamma (IFN-gamma), Interleukin 2(IL-2), Interleukin 2 (IL-6), Interleukin 1 (IL-1), etc. [39,40,41]. These inflammatory factors can trigger the generation of oxidative stress, disrupting the cellular redox balance and producing excessive reactive oxygen species, such as free radicals. Excessive oxidative stress can damage cell membranes, proteins, and nucleic acids, leading to cell injury and death. This cell damage, in turn, can provoke more inflammatory reactions, forming a vicious cycle and causing changes in liver cell function [42,43,44]. In our study, there was a linear relationship between the SII and the NAFLD. The result was not consistent with the Zhao et al. Their research showed that the relationship between the SII and the risk of NAFLD was U-shaped [45]. There was a difference in the definition of NAFLD. NAFLD was defined using the US fatty liver index (FLI) in their study, while we used the CAP to diagnose the NAFLD. In addition, our study population was adolescents, while their study population was adults. In the study of Xie et al., the CAP was used to diagnose hepatic steatosis and they found that there was an inverted U-shaped relationship between SII and CAP [46]. This difference is likely due to dietary differences in adolescentss and adults, particularly sugar intake. Pediatric/adolescents consume more sugar compared to adults. Studies have shown that excessive sugar intake can lead to increased subclinical inflammation and is positively correlated with certain inflammatory markers [47, 48]. For example, fructose-induced liver fat accumulation involves stress pathways, leading to increased gluconeogenesis, fat synthesis, and reduced fat oxidation [49,50,51]. Additionally, long-term fructose exposure in the gut can trigger inflammation by increasing serum TNF-α levels [52], thereby affecting the gut-liver axis and influencing the development of NAFLD [26, 53].

We also found that ALT, triglycerides, and BUN had a partial mediating effect on the relationship between the SII and NAFLD. ALT plays a crucial role in amino acid metabolism, particularly in the reciprocal conversion between alanine and alpha-ketoglutarate. ALT is capable of transferring the amino group from alanine to alpha-ketoglutarate, generating glutamate and pyruvate. This reaction plays a key role in protein metabolism and energy metabolism. SII responds to an index of inflammation and immune balance in the body. When the SII is high, it triggers oxidative stress and produces excess ROS, which in turn damages liver cells, resulting in an increase in ALT, which means that the liver is inflamed. Inflammatory factors can lead to IR, indicating a reduced response of liver cells to insulin. Insulin typically inhibits the activity of ALT. However, in IR states, this inhibitory effect is diminished, leading to an elevation in ALT levels. Additionally, when liver cells are damaged or undergo cell death, ALT is released into the bloodstream [33, 54,55,56,57]. A higher SII means that the patient has a high level of inflammation in the body and more inflammatory factors. Some inflammatory factors, such as IL-6 and TNF-alpha, can promote an increase in lipid synthesis within liver cells, leading to the accumulation of triglycerides in liver cells. Additionally, inflammatory factors can interfere with the insulin signaling pathway, causing IR. Insulin resistance is an important hormone that inhibits lipid breakdown and clearance, resulting in its accumulation within liver cells [33, 54, 58]. These processes contribute to the occurrence and development of NAFLD. An elevation in triglyceride levels may impact the filtration function of the kidneys. In situations where triglyceride levels are elevated, the kidneys may need to process more metabolic products, including urea nitrogen. Therefore, elevated triglycerides may be associated with an increase in urea nitrogen levels. Insulin resistance and abnormalities in glucose metabolism contribute to these metabolic changes, potentially affecting the production and clearance of urea nitrogen [59, 60]. An increase in urea nitrogen levels may be a characteristic feature of NAFLD. In summary, SII may mediate inflammatory pathways and affect the levels of the three, thereby exacerbating inflammation or insulin resistance and promoting disease progression. However, the specific detailed mechanism needs to be further explored by experiments.

Although the study used a variety of data analysis methods to obtain relatively accurate conclusions in American adolescents, there were still some limitations. It should be noted that the NHANES dataset is based on cross-sectional data, which limits the ability to establish causality. Additionally, the data is self-reported and subject to recall and reporting biases, which may introduce measurement errors in the analysis. In addition, only one cycle of study data was enrolled because of year limitations, which may improve the bias of data. Lastly, the use of CAP instead of biopsy to diagnose NAFLD in adolescents is controversial and may affect the accuracy of the conclusions. These limitations should be considered when interpreting the results of this study.

In summary, there was a positive linear relationship between the SII and the risk of NAFLD. The ALT, triglycerides, and BUN had a partial mediating effect on the relationship between the SII and NAFLD. The potential mechanism of SII affecting NAFLD events needs further experimental exploration.

Data availability

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

References

  1. Younossi ZM, Koenig AB, Abdelatif D, Fazel Y, Henry L, Wymer M. Global epidemiology of nonalcoholic fatty liver disease-Meta-analytic assessment of prevalence, incidence, and outcomes. Hepatology. 2016;64(1):73–84.

    Article  PubMed  Google Scholar 

  2. Hardy T, Oakley F, Anstee QM, Day CP. Nonalcoholic fatty liver disease: Pathogenesis and Disease Spectrum. Annu Rev Pathol. 2016;11:451–96.

    Article  CAS  PubMed  Google Scholar 

  3. Zhang X, Wu M, Liu Z, Yuan H, Wu X, Shi T, et al. Increasing prevalence of NAFLD/NASH among children, adolescents and young adults from 1990 to 2017: a population-based observational study. BMJ Open. 2021;11(5):e042843.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Hartmann P, Zhang X, Loomba R, Schnabl B. Global and national prevalence of nonalcoholic fatty liver disease in adolescents: an analysis of the global burden of disease study 2019. Hepatology. 2023;78(4):1168–81.

    Article  PubMed  Google Scholar 

  5. Nobili V, Alisi A, Valenti L, Miele L, Feldstein AE, Alkhouri N. NAFLD in children: new genes, new diagnostic modalities and new drugs. Nat Rev Gastroenterol Hepatol. 2019;16(9):517–30.

    Article  PubMed  Google Scholar 

  6. Schwimmer JB, Pardee PE, Lavine JE, Blumkin AK, Cook S. Cardiovascular risk factors and the metabolic syndrome in pediatric nonalcoholic fatty liver disease. Circulation. 2008;118(3):277–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Duan Y, Luo J, Pan X, Wei J, Xiao X, Li J, et al. Association between inflammatory markers and non-alcoholic fatty liver disease in obese children. Front Public Health. 2022;10:991393.

    Article  PubMed  PubMed Central  Google Scholar 

  8. De Vito R, Alisi A, Masotti A, Ceccarelli S, Panera N, Citti A, et al. Markers of activated inflammatory cells correlate with severity of liver damage in children with nonalcoholic fatty liver disease. Int J Mol Med. 2012;30(1):49–56.

    PubMed  Google Scholar 

  9. Hu B, Yang XR, Xu Y, Sun YF, Sun C, Guo W, et al. Systemic immune-inflammation index predicts prognosis of patients after curative resection for hepatocellular carcinoma. Clin Cancer Res. 2014;20(23):6212–22.

    Article  CAS  PubMed  Google Scholar 

  10. Chen L, Yan Y, Zhu L, Cong X, Li S, Song S, et al. Systemic immune-inflammation index as a useful prognostic indicator predicts survival in patients with advanced gastric cancer treated with neoadjuvant chemotherapy. Cancer Manag Res. 2017;9:849–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Templeton AJ, McNamara MG, Seruga B, Vera-Badillo FE, Aneja P, Ocana A, et al. Prognostic role of neutrophil-to-lymphocyte ratio in solid tumors: a systematic review and meta-analysis. J Natl Cancer Inst. 2014;106(6):dju124.

    Article  PubMed  Google Scholar 

  12. Xie QK, Chen P, Hu WM, Sun P, He WZ, Jiang C, et al. The systemic immune-inflammation index is an independent predictor of survival for metastatic colorectal cancer and its association with the lymphocytic response to the tumor. J Transl Med. 2018;16(1):273.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Estep M, Armistead D, Hossain N, Elarainy H, Goodman Z, Baranova A, et al. Differential expression of miRNAs in the visceral adipose tissue of patients with non-alcoholic fatty liver disease. Aliment Pharmacol Ther. 2010;32(3):487–97.

    Article  CAS  PubMed  Google Scholar 

  14. Karlas T, Petroff D, Sasso M, Fan JG, Mi YQ, de Ledinghen V, et al. Individual patient data meta-analysis of controlled attenuation parameter (CAP) technology for assessing steatosis. J Hepatol. 2017;66(5):1022–30.

    Article  PubMed  Google Scholar 

  15. Siddiqui MS, Vuppalanchi R, Van Natta ML, Hallinan E, Kowdley KV, Abdelmalek M, et al. Vibration-controlled transient elastography to assess fibrosis and steatosis in patients with nonalcoholic fatty liver disease. Clin Gastroenterol Hepatol. 2019;17(1):156–63. e2.

    Article  PubMed  Google Scholar 

  16. Qin Z, Li H, Wang L, Geng J, Yang Q, Su B, et al. Systemic Immune-inflammation index is Associated with increased urinary albumin excretion: a Population-based study. Front Immunol. 2022;13:863640.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Zhu JL, Xu XM, Yin HY, Wei JR, Lyu J. Development and validation of a nomogram for predicting hospitalization longer than 14 days in pediatric patients with ventricular septal defect-a study based on the PIC database. Front Physiol. 2023;14:1182719.

    Article  PubMed  PubMed Central  Google Scholar 

  18. Schwimmer JB, McGreal N, Deutsch R, Finegold MJ, Lavine JE. Influence of gender, race, and ethnicity on suspected fatty liver in obese adolescents. Pediatrics. 2005;115(5):e561–5.

    Article  PubMed  Google Scholar 

  19. Roh JH, Lee H, Yun-Jeong B, Park CS, Kim HJ, Yoon SY. A nationwide survey of the association between nonalcoholic fatty liver disease and the incidence of asthma in Korean adults. PLoS ONE. 2022;17(1):e0262715.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Mahady SE, George J. Exercise and diet in the management of nonalcoholic fatty liver disease. Metabolism. 2016;65(8):1172–82.

    Article  CAS  PubMed  Google Scholar 

  21. Chung GE, Yim JY, Kim D, Lim SH, Yang JI, Kim YS, et al. The influence of metabolic factors for nonalcoholic fatty liver disease in women. Biomed Res Int. 2015;2015:131528.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Romeo S, Kozlitina J, Xing C, Pertsemlidis A, Cox D, Pennacchio LA, et al. Genetic variation in PNPLA3 confers susceptibility to nonalcoholic fatty liver disease. Nat Genet. 2008;40(12):1461–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Alvarez CS, Graubard BI, Thistle JE, Petrick JL, McGlynn KA. Attributable fractions of nonalcoholic fatty liver disease for mortality in the United States: results from the Third National Health and Nutrition Examination Survey with 27 years of follow-up. Hepatology. 2020;72(2):430–40.

    Article  CAS  PubMed  Google Scholar 

  24. Cobbina E, Akhlaghi F. Non-alcoholic fatty liver disease (NAFLD) - pathogenesis, classification, and effect on drug metabolizing enzymes and transporters. Drug Metab Rev. 2017;49(2):197–211.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Patel SS, Siddiqui MS. Current and emerging therapies for non-alcoholic fatty liver disease. Drugs. 2019;79(1):75–84.

    Article  CAS  PubMed  Google Scholar 

  26. Fang YL, Chen H, Wang CL, Liang L. Pathogenesis of non-alcoholic fatty liver disease in children and adolescence: from two hit theory to multiple hit model. World J Gastroenterol. 2018;24(27):2974–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Ratziu V, Bellentani S, Cortez-Pinto H, Day C, Marchesini G. A position statement on NAFLD/NASH based on the EASL 2009 special conference. J Hepatol. 2010;53(2):372–84.

    Article  PubMed  Google Scholar 

  28. Berardis S, Sokal E. Pediatric non-alcoholic fatty liver disease: an increasing public health issue. Eur J Pediatr. 2014;173(2):131–9.

    Article  CAS  PubMed  Google Scholar 

  29. Alisi A, Cianfarani S, Manco M, Agostoni C, Nobili V. Non-alcoholic fatty liver disease and metabolic syndrome in adolescents: pathogenetic role of genetic background and intrauterine environment. Ann Med. 2012;44(1):29–40.

    Article  CAS  PubMed  Google Scholar 

  30. Ayonrinde OT, Olynyk JK, Marsh JA, Beilin LJ, Mori TA, Oddy WH, et al. Childhood adiposity trajectories and risk of nonalcoholic fatty liver disease in adolescents. J Gastroenterol Hepatol. 2015;30(1):163–71.

    Article  PubMed  Google Scholar 

  31. Ferramosca A, Zara V. Modulation of hepatic steatosis by dietary fatty acids. World J Gastroenterol. 2014;20(7):1746–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Buzzetti E, Pinzani M, Tsochatzis EA. The multiple-hit pathogenesis of non-alcoholic fatty liver disease (NAFLD). Metabolism. 2016;65(8):1038–48.

    Article  CAS  PubMed  Google Scholar 

  33. Hotamisligil GS. Inflammation and metabolic disorders. Nature. 2006;444(7121):860–7.

    Article  CAS  PubMed  Google Scholar 

  34. Rebrin K, Steil GM, Mittelman SD, Bergman RN. Causal linkage between insulin suppression of lipolysis and suppression of liver glucose output in dogs. J Clin Invest. 1996;98(3):741–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Petersen MC, Shulman GI. Roles of Diacylglycerols and Ceramides in hepatic insulin resistance. Trends Pharmacol Sci. 2017;38(7):649–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Tomita K, Tamiya G, Ando S, Ohsumi K, Chiyo T, Mizutani A, et al. Tumour necrosis factor alpha signalling through activation of Kupffer cells plays an essential role in liver fibrosis of non-alcoholic steatohepatitis in mice. Gut. 2006;55(3):415–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Song Y, Guo W, Li Z, Guo D, Li Z, Li Y. Systemic immune-inflammation index is associated with hepatic steatosis: evidence from NHANES 2015–2018. Front Immunol. 2022;13:1058779.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Jou J, Choi SS, Diehl AM. Mechanisms of disease progression in nonalcoholic fatty liver disease. Semin Liver Dis. 2008;28(4):370–9.

    Article  CAS  PubMed  Google Scholar 

  39. Van Laere K, Koole M, Kauppinen T, Monsieurs M, Bouwens L, Dierck R. Nonuniform transmission in brain SPECT using 201Tl, 153Gd, and 99mTc static line sources: anthropomorphic dosimetry studies and influence on brain quantification. J Nucl Med. 2000;41(12):2051–62.

    PubMed  Google Scholar 

  40. Vacchina P, Tripodi KE, Escalante AM, Uttaro AD. Characterization of bifunctional sphingolipid Delta4-desaturases/C4-hydroxylases of trypanosomatids by liquid chromatography-electrospray tandem mass spectrometry. Mol Biochem Parasitol. 2012;184(1):29–38.

    Article  CAS  PubMed  Google Scholar 

  41. Karin M, Cao Y, Greten FR, Li ZW. NF-kappaB in cancer: from innocent bystander to major culprit. Nat Rev Cancer. 2002;2(4):301–10.

    Article  CAS  PubMed  Google Scholar 

  42. Zhang X, Milton CC, Poon CL, Hong W, Harvey KF. Wbp2 cooperates with yorkie to drive tissue growth downstream of the Salvador-Warts-Hippo pathway. Cell Death Differ. 2011;18(8):1346–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Cereda E, Cilia R, Klersy C, Canesi M, Zecchinelli AL, Mariani CB, et al. Swallowing disturbances in Parkinson’s disease: a multivariate analysis of contributing factors. Parkinsonism Relat Disord. 2014;20(12):1382–7.

    Article  PubMed  Google Scholar 

  44. Leinonen R, Akhtar R, Birney E, Bonfield J, Bower L, Corbett M, et al. Improvements to services at the European Nucleotide Archive. Nucleic Acids Res. 2010;38(Database issue):D39–45.

    Article  CAS  PubMed  Google Scholar 

  45. Zhao B, Liu Y, Yang Y, He J. Association of systemic Immune-inflammation index with non-alcoholic fatty liver disease: a Population-based cross-sectional study. Risk Manag Healthc Policy. 2023;16:1581–92.

    Article  PubMed  PubMed Central  Google Scholar 

  46. Xie R, Xiao M, Li L, Ma N, Liu M, Huang X, et al. Association between SII and hepatic steatosis and liver fibrosis: a population-based study. Front Immunol. 2022;13:925690.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Correa-Rodriguez M, Pocovi-Gerardino G, Callejas-Rubio JL, Rios Fernandez R, Martin-Amada M, Cruz-Caparros MG et al. Dietary intake of free sugars is Associated with Disease Activity and Dyslipidemia in systemic Lupus Erythematosus patients. Nutrients. 2020;12(4).

  48. O’Connor L, Imamura F, Brage S, Griffin SJ, Wareham NJ, Forouhi NG. Intakes and sources of dietary sugars and their association with metabolic and inflammatory markers. Clin Nutr. 2018;37(4):1313–22.

    Article  PubMed  PubMed Central  Google Scholar 

  49. Lanaspa MA, Sanchez-Lozada LG, Choi YJ, Cicerchi C, Kanbay M, Roncal-Jimenez CA, et al. Uric acid induces hepatic steatosis by generation of mitochondrial oxidative stress: potential role in fructose-dependent and -independent fatty liver. J Biol Chem. 2012;287(48):40732–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Lanaspa MA, Cicerchi C, Garcia G, Li N, Roncal-Jimenez CA, Rivard CJ, et al. Counteracting roles of AMP deaminase and AMP kinase in the development of fatty liver. PLoS ONE. 2012;7(11):e48801.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Cicerchi C, Li N, Kratzer J, Garcia G, Roncal-Jimenez CA, Tanabe K, et al. Uric acid-dependent inhibition of AMP kinase induces hepatic glucose production in diabetes and starvation: evolutionary implications of the uricase loss in hominids. FASEB J. 2014;28(8):3339–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Spruss A, Kanuri G, Wagnerberger S, Haub S, Bischoff SC, Bergheim I. Toll-like receptor 4 is involved in the development of fructose-induced hepatic steatosis in mice. Hepatology. 2009;50(4):1094–104.

    Article  CAS  PubMed  Google Scholar 

  53. Rotman Y, Sanyal AJ. Current and upcoming pharmacotherapy for non-alcoholic fatty liver disease. Gut. 2017;66(1):180–90.

    Article  CAS  PubMed  Google Scholar 

  54. Yu JH, Zhu BM, Wickre M, Riedlinger G, Chen W, Hosui A, et al. The transcription factors signal transducer and activator of transcription 5A (STAT5A) and STAT5B negatively regulate cell proliferation through the activation of cyclin-dependent kinase inhibitor 2b (Cdkn2b) and Cdkn1a expression. Hepatology. 2010;52(5):1808–18.

    Article  CAS  PubMed  Google Scholar 

  55. Roe DR, Cheatham TE 3. PTRAJ and CPPTRAJ: Software for Processing and Analysis of Molecular Dynamics Trajectory Data. J Chem Theory Comput. 2013;9(7):3084–95.

    Article  CAS  PubMed  Google Scholar 

  56. Adams LA, Lymp JF, St Sauver J, Sanderson SO, Lindor KD, Feldstein A, et al. The natural history of nonalcoholic fatty liver disease: a population-based cohort study. Gastroenterology. 2005;129(1):113–21.

    Article  PubMed  Google Scholar 

  57. Pitz S, Megonigal JP. Temperate forest methane sink diminished by tree emissions. New Phytol. 2017;214(4):1432–9.

    Article  CAS  PubMed  Google Scholar 

  58. DeBerardinis RJ, Thompson CB. Cellular metabolism and disease: what do metabolic outliers teach us? Cell. 2012;148(6):1132–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Haque R, Winfree CJ. Transforaminal nerve root stimulation: a technical report. Neuromodulation. 2009;12(3):254–7.

    Article  PubMed  Google Scholar 

  60. O’Brien E. Sleepers versus nonsleepers: another twist to the dipper/nondipper concept. Hypertension. 2007;49(4):769–70.

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

Not applicable.

Author information

Authors and Affiliations

Authors

Contributions

DFF contributed to the conception and design. DFF and BC contributed to the collection and assembly of data. DFF and Bin Chen analyzed and interpreted the data. All authors wrote and approved the final manuscript.

Corresponding author

Correspondence to Bin Chen.

Ethics declarations

Ethics approval and consent to participate

The Ethics Committee of Hangzhou Xiaoshan First People’s Hospital deemed that this research is based on open-source data, so the need for ethics approval was waived.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Fu, Df., Chen, B. The relationship between the systemic immune inflammation index and the nonalcoholic fatty liver disease in American adolescents. BMC Gastroenterol 24, 233 (2024). https://doi.org/10.1186/s12876-024-03324-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12876-024-03324-6

Keywords