Skip to main content
Erschienen in: BMC Infectious Diseases 1/2023

Open Access 01.12.2023 | Research

Fatty liver is associated with significant liver inflammation and increases the burden of advanced fibrosis in chronic HBV infection

verfasst von: Yi-Ning Dai, Cheng-Fu Xu, Hong-Ying Pan, Mei-Juan Chen, Chao-Hui Yu

Erschienen in: BMC Infectious Diseases | Ausgabe 1/2023

Abstract

Background

Concurrent non-alcoholic fatty liver disease (NAFLD) is common in patients with chronic HBV infection. But the impact of fatty liver on the histologic progression of HBV infection remains controversial.

Methods

Consecutive HBV-infected patients who underwent liver biopsy between 2016 and 2021 were included. Alcohol consumption and other types of viral hepatitis were excluded. All biopsies were scored for grading and staging by Scheuer’s score, and the steatosis was scored as an estimate of the percentage of liver parenchyma replaced by fat. Logistic regression analyses were applied to assess the associated factors for significant liver inflammation (G ≥ 2), significant fibrosis (S ≥ 2) and advanced fibrosis (S ≥ 3).

Results

Among the 871 HBV-infected patients, hepatic steatosis was prevalent in 255 patients (29.28%). Significant liver inflammation was present in 461 patients (52.93%). Significant fibrosis was observed in 527 patients (60.51%), while advanced liver fibrosis was observed in 171 patients (19.63%). Patients with concomitant NAFLD were more likely to have significant liver inflammation and advanced fibrosis. Fatty liver was an independent risk factor for significant liver inflammation (OR: 2.117, 95% CI: 1.500-2.988), but it could not predict the development of fibrosis. Especially, in HBV-infected patients with persistent normal ALT (immune tolerant and inactive carrier phase), the presence of significant liver inflammation was higher in NAFLD than those without NAFLD. The prevalence of advanced liver fibrosis was higher in NAFLD than non-NAFLD only in the immune tolerant phase, while NAFLD did not increase fibrosis burden in other stages of HBV infection. We developed a predictive model for significant liver inflammation with the area under receiver operating characteristic curve (AUROC) of 0.825, and a model for significant fibrosis with the AUROC of 0.760.

Conclusions

NAFLD is independently associated with significant liver inflammation, and increases the burden of advanced liver fibrosis in HBV-infected patients. The influence of NAFLD on the degree of liver inflammation and fibrosis is different in distinct clinical phases of chronic HBV infection.
Hinweise

Publisher’s Note

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

Background

With the dramatic rise in the prevalence of non-alcoholic fatty liver disease (NAFLD), it frequently coexists with other conditions such as alcohol consumption and viral hepatitis. In the meantime, chronic hepatitis B (CHB) caused by hepatitis B virus (HBV) infection is still one of the most common causes of chronic liver diseases in China [1]. Consequently, CHB and NAFLD are frequently observed together with an estimated 30% having hepatic steatosis among those with CHB [2].
Until now, the interplay between the two diseases has not been thoroughly evaluated. Several studies revealed that hepatic steatosis in chronic HBV infection did not appear to affect the severity of liver histology [3, 4]. Conversely, some recent researches concluded steatosis was associated with advanced fibrosis in CHB [58]. Therefore, the effect of fatty liver on the natural history of chronic HBV infection still remains controversial [9].
With this background, we aim to compare the histologic differences between simple chronic HBV infection and HBV infection with concomitant NAFLD, and to study whether fatty liver predict severe liver histology like significant liver inflammation or fibrosis in chronic HBV infection. Furthermore, noninvasive models were developed to accurately identify significant liver inflammation and significant fibrosis in this study.

Methods

Study design and patients

A retrospective cross-sectional study was conducted at Zhejiang Provincial People’s Hospital. We included patients with chronic HBV infection over 18 years-old who underwent a liver biopsy between 2016 and 2021. The exclusion criteria were as follows: (1) excessive alcohol consumption (ethanol consumption more than 140 g in men and 70 g in women per week); (2) other types of viral hepatitis (e.g., chronic hepatitis C virus infection); (3) other causes of liver injury (e.g., drug-induced liver disease, autoimmune liver disease, or hereditary disorders); (4) pregnancy, malignancy, severe cardiopulmonary disorders, or renal dysfunction.
The natural history of chronic HBV infection has been divided into four clinical phases as follows, taking into account the clinical data of patients including presence of hepatitis B e antigen (HBeAg), Hepatitis B virus DNA (HBV DNA) levels and alanine aminotransferase (ALT) values [10]. (1) HBeAg-positive chronic HBV infection, i.e. “immune tolerant” phase: positive serum HBeAg, very high levels of HBV DNA and ALT persistently within the normal range (upper limit of normal (ULN) of 40 IU/ml). (2) HBeAg-positive CHB: positive serum HBeAg, high levels of HBV DNA and abnormal ALT values. (3) HBeAg-negative chronic HBV infection, i.e. “inactive carrier” phase: negative serum HBeAg, undetectable or low (< 2000 IU/ml) HBV DNA levels and normal ALT; Some patients in this phase may have serum HBV DNA levels > 2000 IU/ml (usually < 20,000 IU/ml) accompanied by persistently normal ALT. (4) HBeAg-negative CHB: negative serum HBeAg, moderate to high levels of HBV DNA, and elevated or fluctuating ALT levels.
This study was approved by the Ethics Committee of People’s Hospital of Hangzhou Medical College and followed the guidelines for studies in humans. Informed consents were obtained from all subjects.

Clinical data and liver biopsy

Clinical, demographic and laboratory data were collected from the medical records of patients. Complete blood counts, biochemical and virological (HBV DNA, the positivity of HBeAg) data were recorded as the closest results to the date on which liver biopsy was performed.
Skilled doctors performed percutaneous liver biopsy using the MAX-CORE Disposable Core Biopsy Instrument (Bard Peripheral Vascular, Inc., Mexico). The specimens were fixed, paraffin-embedded, and stained by haematoxylin and eosin (H&E) and Masson’s trichrome for further pathological evaluation by an experienced liver pathologist. All liver biopsy slides should be qualified for scoring of histologic features.
A threshold of 5% macrovesicular steatosis made a diagnosis of NAFLD [11]. Steatosis was graded as the percentage of liver parenchyma replaced by fat: (1) 5 − 33%, (2) 34 − 66%, or (3) more than 66% [12]. Lobular inflammation was scored on a scale of 0–3: (0) none, (1) mild, (2) moderate, and (3) many. The degree of portal inflammation and hepatocellular ballooning were divided as: (0) none, (1) mild inflammation or few balloon cells, and (2) prominent inflammation or ballooning. Liver inflammation (G0 to G4) and fibrosis (S0 to S4) were assessed according to the Scheuer scoring system [13]. The grades of liver inflammation were classified into the following 5 stages: G0, no inflammation; G1, inflammatory but no necrosis; G2, focal necrosis or acidophil bodies; G3, severe focal cell damage; and G4, widely bridging necrosis and piecemeal necrosis. Significant liver inflammation was defined as inflammation grade of G2 to G4. Liver fibrosis was scored as follows: S0, no fibrosis; S1, portal fibrosis without septa; S2, portal fibrosis with rare septa; S3, numerous septa without cirrhosis; and S4, cirrhosis. Fibrotic stage of S2 to S4 was considered as significant fibrosis, while fibrotic stage of S3 to S4 was defined as advanced fibrosis [14].

Statistical analysis

Statistical analyses were performed by SPSS (version 23) and Python 3.7. Continuous variables are described as the mean ± standard deviation (SD), and categorical variables are presented as numbers (percentages). Propensity score-matching (PSM) was used to adjust the potential confounding factors including age, gender, HBV DNA and the positivity of HBeAg. When evaluating differences between groups, the t-test or the chi-squared test was applied. A two-sided P value < 0.05 was considered statistically significant.
Logistic analysis was applied to assess the risk factors for significant liver inflammation, significant fibrosis and advanced fibrosis. The associated factors observed in the univariate analysis were utilized for model training. The establishments of models were based on the Scikit-Learn package of Scientific Python 3.7 libraries. A binary logistic regression was performed to predict the probability of significant liver inflammation and significant fibrosis. We used the LIBLINEAR library to carry out the computation. Area under receiver operating characteristic curve (AUROC) was used to evaluate the predictive accuracy of the model.

Results

Study Population

A total of 871 patients with chronic HBV infection were included in this study. The mean age of the study population was 39.14 (± 9.93) years, and 586 (67.28%) were male patients. Among them, 352 patients (40.41%) were HBeAg-positive. Majority of the patients (93.92%) did not receive any antiviral therapy. Hepatic steatosis was present in 255 patients (29.28%): 219 grade 1 steatosis, 33 grade 2 steatosis, and 3 grade 3 steatosis. Significant hepatic inflammation was present in 461 patients (52.62%). Significant fibrosis was observed in 527 patients (60.51%), while advanced liver fibrosis was observed in 171 patients (19.63%).

Comparisons between HBV-infected patients with and without hepatic steatosis

The characteristics of HBV-infected patients with and without hepatic steatosis are exhibited in Table 1. Compared with patients without fatty liver, chronic HBV infection with concurrent NAFLD were more likely to be male, HBeAg-negative, and to have hypertension and type 2 diabetes mellitus (T2DM). Furthermore, they had significantly higher height, weight, body mass index (BMI) and higher levels of white blood cells (WBC), albumin, total bilirubin, ALT, GGT and ALP.
Table 1
Clinical characteristics of HBV-infected patients with and without hepatic steatosis
 
HBV infection (N = 616)
HBV infection + NAFLD (N = 255)
P value
Age
38.87 ± 10.19
39.78 ± 9.25
0.222
Gender (M/F)
366/250
220/35
< 0.001
Height
164.63 ± 8.23
167.63 ± 7.97
< 0.001
Weight
60.69 ± 10.14
73.00 ± 10.71
< 0.001
BMI
22.33 ± 2.98
25.98 ± 3.55
< 0.001
WBC
5.61 ± 1.51
6.00 ± 1.37
< 0.001
PLT
184.83 ± 52.47
191.60 ± 52.89
0.084
ALB
43.77 ± 3.57
44.54 ± 3.40
0.003
GLB
29.22 ± 3.93
29.02 ± 4.00
0.482
Tbil
15.23 ± 6.22
16.51 ± 6.29
0.006
ALT
38.62 ± 32.75
52.02 ± 37.48
< 0.001
AST
33.30 ± 19.40
35.81 ± 17.58
0.075
GGT
27.24 ± 26.64
42.89 ± 36.26
< 0.001
ALP
81.10 ± 24.27
89.68 ± 25.51
< 0.001
HBV DNA (Log10 IU/mL)
4.55 ± 2.66
4.23 ± 2.52
0.093
HbeAg (+)
264 (42.86%)
88 (34.51%)
0.022
Anti-viral therapy
36 (5.84%)
17 (6.67%)
0.644
Hypertension
29 (4.71%)
32 (12.55%)
< 0.001
T2DM
7 (1.14%)
13 (5.10%)
< 0.001
NAFLD: Non-alcoholic fatty liver disease; M: Male; F: Female; BMI: Body mass index; WBC: White blood cell count; PLT: Platelet; ALB: Albumin; GLB: Globulin; Tbil: Total bilirubin; ALT: Alanine aminotransferase; AST: Aspartate transferase; GGT: Gamma-glutamyltransferase; ALP: Alkaline phosphatase; HBV: Hepatitis B virus; HbeAg: hepatitis B e antigen; T2DM: Type 2 diabetes mellitus
Compared to HBV-infected patients without fatty liver, chronic HBV infection with concurrent NAFLD had a higher severity of hepatic steatosis, hepatic inflammation especially portal inflammation, and hepatic ballooning (Table 2). There were no significant differences based on the grades of lobular inflammation or fibrosis. The prevalence of significant fibrosis was similar between the two groups. However, there was borderline difference in the presence of advanced liver fibrosis (P = 0.049).
Table 2
Histologic characteristics of HBV-infected patients with and without hepatic steatosis
 
HBV infection (N = 616)
HBV infection + NAFLD (N = 255)
P value
Steatosis
  
< 0.001
0
616 (100%)
0
1
0
219 (85.88%)
2
0
33 (12.94%)
3
0
3 (1.18%)
Lobular inflammation
  
0.323
0
1 (0.16%)
0
1
389 (63.15%)
150 (58.82%)
2
210 (34.09%)
93 (36.47%)
3
15 (2.44%)
12 (4.71%)
4
1 (0.16%)
0
Portal inflammation
  
< 0.001
1
360 (58.44%)
101 (39.61%)
2
227 (36.85%)
127 (49.80%)
3
26 (4.22%)
26 (10.20%)
4
3 (0.49%)
1 (0.39%)
Hepatic ballooning
   
0
608 (98.70%)
239 (93.73%)
 
1
8 (1.30%)
9 (3.53%)
< 0.001
2
0
7 (2.75%)
 
3
0
0
 
G
  
< 0.001
1
318 (51.62%)
92 (36.08%)
2
268 (43.51%)
135 (52.94%)
3
28 (4.55%)
27 (10.59%)
4
2 (0.32%)
1 (0.39%)
S
  
0.071
0
11 (1.79%)
0
1
236 (38.31%)
97 (38.04%)
2
259 (42.05%)
97 (38.04%)
3
54 (8.77%)
30 (11.76%)
4
56 (9.09%)
31 (12.16%)
Significant liver fibrosis
369 (59.90%)
158 (61.96%)
0.572
Advanced liver fibrosis
110 (17.857%)
61 (23.922%)
0.049
Significant liver inflammation
298 (48.38%)
163 (63.92%)
< 0.001
NAFLD: Non-alcoholic fatty liver disease; G: inflammation grade; S: fibrosis stage
To alleviate the effects of confounders, PSM was performed at a 1:1 ratio, yielding 254 matched pairs. Compared with HBV-infected patients without fatty liver, chronic HBV infection with concurrent NAFLD had higher possibility of having hypertension, T2DM, and had greater body weight, BMI, and greater concentrations of ALT, GGT and ALP (Table 3).
Table 3
Clinical characteristics of HBV-infected patients with and without hepatic steatosis after PSM
 
HBV infection (N = 254)
HBV infection + NAFLD (N = 254)
P value
Age
39.71 ± 9.33
39.79 ± 9.27
0.332
Gender (M/F)
219/35
219/35
/
Height
167.76 ± 6.61
167.62 ± 7.98
0.796
Weight
64.19 ± 9.27
73.01 ± 10.73
< 0.001
BMI
22.79 ± 2.94
25.98 ± 3.56
< 0.001
WBC
5.77 ± 1.60
6.01 ± 1.37
0.066
PLT
183.13 ± 53.90
191.55 ± 52.99
0.068
ALB
44.09 ± 3.45
44.53 ± 3.40
0.093
GLB
28.61 ± 3.89
29.03 ± 4.01
0.212
Tbil
16.10 ± 6.50
16.51 ± 6.31
0.456
ALT
41.76 ± 35.98
52.08 ± 37.54
0.001
AST
33.46 ± 19.05
35.84 ± 17.61
0.132
GGT
31.25 ± 30.39
42.90 ± 36.33
< 0.001
ALP
84.67 ± 21.86
89.56 ± 25.49
0.015
HBV DNA (Log10 IU/mL)
4.22 ± 2.51
4.25 ± 2.51
0.436
HbeAg (+)
88 (34.65%)
88 (34.65%)
/
Anti-viral therapy
16 (6.30%)
16 (6.30%)
/
Hypertension
12 (4.72%)
32 (12.60%)
0.002
T2DM
3 (1.18%)
13 (5.12%)
0.011
PSM: Propensity score-matching; NAFLD: Non-alcoholic fatty liver disease; M: Male; F: Female; BMI: Body mass index; WBC: White blood cell count; PLT: Platelet; ALB: Albumin; GLB: Globulin; Tbil: Total bilirubin; ALT: Alanine aminotransferase; AST: Aspartate transferase; GGT: Gamma-glutamyltransferase; ALP: Alkaline phosphatase; HBV: Hepatitis B virus; HbeAg: hepatitis B e antigen; T2DM: Type 2 diabetes mellitus
The histologic characteristics of the patients after PSM were exhibited in Table 4. It revealed that chronic HBV infection with concurrent NAFLD had a greater severity of hepatic steatosis, lobular and portal inflammation, hepatic ballooning and fibrosis. Chronic HBV infection with NAFLD is more prone to have significant inflammation and advanced liver fibrosis, while but the probability of having significant fibrosis between the two groups was similar.
Table 4
Histologic characteristics of HBV-infected patients with and without hepatic steatosis after PSM
 
HBV infection (N = 254)
HBV infection + NAFLD (N = 254)
P value
Steatosis
  
< 0.001
0
254 (100%)
0
1
0
218 (85.83%)
2
0
33 (12.99%)
3
0
3 (1.18%)
Lobular inflammation
  
< 0.001
0
1 (0.39%)
0
1
193 (75.98%)
150 (59.06%)
2
59 (23.23%)
92 (36.22%)
3
1 (0.39%)
12 (4.72%)
4
0
0
Portal inflammation
  
< 0.001
1
165 (64.96%)
100 (39.37%)
2
81 (31.89%)
127 (50.00%)
3
8 (3.15%)
26 (10.24%)
4
0
1 (0.39%)
Hepatic ballooning
  
0.001
0
253 (99.61%)
238 (93.70%)
1
1 (0.39%)
9 (3.54%)
2
0
7 (2.76%)
3
0
0
G
  
< 0.001
1
148 (58.27%)
92 (36.22%)
2
98 (38.58%)
134 (52.76%)
3
8 (3.15%)
27 (10.63%)
4
0
1 (0.39%)
S
  
0.010
0
9 (3.54%)
0
1
105 (41.34%)
97 (38.19%)
2
100 (39.37%)
97 (38.19%)
3
20 (7.87%)
29 (11.42%)
4
20 (7.87%)
31 (12.20%)
Significant liver fibrosis
140 (55.12%)
157 (61.81%)
0.126
Advanced liver fibrosis
40 (15.75%)
60 (23.62%)
0.034
Significant liver inflammation
106 (41.73%)
162 (63.78%)
< 0.001
PSM: Propensity score-matching; NAFLD: Non-alcoholic fatty liver disease; G: inflammation grade; S: fibrosis stage

Factors associated with significant liver inflammation, significant fibrosis and advanced liver fibrosis

Univariate analysis showed hepatic steatosis, ALT, AST, GGT, ALB, GLB, PLT and the positivity of HBeAg were predictors of significant liver inflammation. The above characteristics entered into the subsequent multivariate analysis, revealing that the following seven characteristics were associated with higher odds of having significant inflammation in chronic HBV infection: steatosis, decreased ALT and ALB, elevated AST, GGT and GLB, and the positivity of HBeAg (Table 5).
Table 5
Factors associated with significant liver inflammation
Variables
Univariate
Multivariate
P value
β ± SE
Wald
OR (95% CI)
P value
Age
0.544
    
Gender
0.690
    
HBV DNA (Log10 IU/mL)
0.863
    
HbeAg (+)
0.031
0.436 ± 0.157
7.760
1.547 (1.138–2.103)
0.005
BMI
0.242
    
Hypertension
0.377
    
T2DM
0.756
    
WBC
0.105
    
PLT
0.053
-0.003 ± 0.001
3.644
0.997 (0.994-1.000)
0.056
ALB
0.003
-0.080 ± 0.023
11.827
0.923 (0.881–0.966)
0.001
GLB
0.013
0.057 ± 0.021
7.602
1.059 (1.017–1.103)
0.006
Tbil
0.082
    
ALT
0.038
-0.013 ± 0.005
5.500
0.987 (0.977–0.998)
0.019
AST
< 0.001
0.054 ± 0.012
21.319
1.055 (1.031–1.079)
< 0.001
GGT
0.041
0.008 ± 0.004
4.606
1.008 (1.001–1.015)
0.032
ALP
0.178
    
Steatosis
0.001
0.750 ± 0.176
18.210
2.117 (1.500-2.988)
< 0.001
HBV: Hepatitis B virus; HbeAg: hepatitis B e antigen; BMI: Body mass index; T2DM: Type 2 diabetes mellitus; WBC: White blood cell count; PLT: Platelet; ALB: Albumin; GLB: Globulin; Tbil: Total bilirubin; ALT: Alanine aminotransferase; AST: Aspartate transferase; GGT: Gamma-glutamyltransferase; ALP: Alkaline phosphatase
Both univariate and multivariate analyses demonstrated associated factors for higher probability of significant fibrosis in chronic HBV infection were as follows: decreased levels of HBV DNA, hypertension, decreased ALT, elevated AST, GGT and GLB (Table 6).
Table 6
Factors associated with significant liver fibrosis
Variables
Univariate
Multivariate
P value
β ± SE
Wald
OR (95% CI)
P value
Age
0.196
    
Gender
0.211
    
HBV DNA (Log10 IU/mL)
0.004
-0.127 ± 0.029
19.317
0.881 (0.832–0.932)
< 0.001
HbeAg (+)
0.706
    
BMI
0.769
    
Hypertension
0.028
0.890 ± 0.341
6.802
2.434 (1.247–4.751)
0.009
T2DM
0.727
    
WBC
0.194
    
PLT
0.086
    
ALB
0.712
    
GLB
0.037
0.040 ± 0.020
3.977
1.041 (1.001–1.083)
0.046
Tbil
0.952
    
ALT
0.002
-0.017 ± 0.005
12.163
0.983 (0.974–0.993)
< 0.001
AST
< 0.001
0.045 ± 0.010
19.461
1.046 (1.025–1.067)
< 0.001
GGT
0.059
0.007 ± 0.003
4.711
1.007 (1.001–1.083)
0.030
ALP
0.952
    
Steatosis
0.784
    
HBV: Hepatitis B virus; HbeAg: hepatitis B e antigen; BMI: Body mass index; T2DM: Type 2 diabetes mellitus; WBC: White blood cell count; PLT: Platelet; ALB: Albumin; GLB: Globulin; Tbil: Total bilirubin; ALT: Alanine aminotransferase; AST: Aspartate transferase; GGT: Gamma-glutamyltransferase; ALP: Alkaline phosphatase
Table 7 showed factors related to higher risk of advanced liver fibrosis in chronic HBV infection included male gender, decreased levels of HBV DNA, decreased levels of PLT and ALT, and elevated levels of AST, GGT and GLB.
Table 7
Factors associated with advanced liver fibrosis
Variables
Univariate
Multivariate
P value
β ± SE
Wald
OR (95% CI)
P value
Age
0.067
    
Gender (if male)
0.036
0.644 ± 0.232
7.720
1.904 (1.209–2.998)
0.005
HBV DNA (Log10 IU/mL)
0.001
-0.139 ± 0.038
13.350
0.871 (0.808–0.938)
< 0.001
HbeAg (+)
0.099
    
BMI
0.465
    
Hypertension
0.560
    
T2DM
0.576
    
WBC
0.554
    
PLT
< 0.001
-0.010 ± 0.002
24.919
0.990 (0.987–0.994)
< 0.001
ALB
0.903
    
GLB
< 0.001
0.105 ± 0.024
19.018
1.111 (1.059–1.164)
< 0.001
Tbil
0.338
    
ALT
0.031
-0.011 ± 0.005
5.384
0.989 (0.980–0.998)
0.020
AST
0.007
0.023 ± 0.008
7.211
1.023 (1.006–1.040)
0.007
GGT
0.005
0.010 ± 0.003
9.924
1.010 (1.004–1.016)
0.002
ALP
0.533
    
Steatosis
0.229
    
HBV: Hepatitis B virus; HbeAg: hepatitis B e antigen; BMI: Body mass index; T2DM: Type 2 diabetes mellitus; WBC: White blood cell count; PLT: Platelet; ALB: Albumin; GLB: Globulin; Tbil: Total bilirubin; ALT: Alanine aminotransferase; AST: Aspartate transferase; GGT: Gamma-glutamyltransferase; ALP: Alkaline phosphatase

Liver histology based on the natural history of chronic HBV infection

The 818 patients who haven’t receive any antiviral therapy were divided into four disease phases of chronic HBV infection according to their clinical data. Among them, 238 patients had concurrent NAFLD, while the other 580 did not. We compared the presence of significant liver inflammation, significant fibrosis and advanced liver fibrosis between the two groups with or without NAFLD according to the natural history of chronic HBV infection. Patients with normal ALT (chronic HBV infection with no evidence of CHB) and elevated ALT values (CHB) were also analyzed separately.
As shown in Table 8, in patients with HBeAg-positive and HBeAg-negative chronic HBV infection (immune tolerant and inactive carrier phase), that is, in those HBV-infected patients with persistent normal ALT value, the presence of significant liver inflammation was higher in NAFLD group than that in the non-NAFLD group (68.750% vs. 43.571%, P = 0.011; 59.302% vs. 35.685%, P < 0.001; 61.864% vs. 38.583, P < 0.001 in chronic HBV infection with no evidence of CHB). Furthermore, in patients with HBeAg-positive chronic HBV infection (immune tolerant phase), the presence of advanced liver fibrosis was higher in NAFLD group than that in the non-NAFLD group (25% vs. 10.714%, P = 0.044).
Table 8
Liver histology based on the natural history of chronic HBV infection
Phase of chronic HBV infection
NAFLD or not
Total
Significant liver inflammation (%)
P value
Significant liver fibrosis (%)
P value
Advanced liver fibrosis (%)
P value
HBeAg-positive chronic HBV infection
Non-NAFLD
140
61 (43.571)
0.011
66 (47.143)
0.434
15 (10.714)
0.044
NAFLD
32
22 (68.750)
18 (56.25)
8 (25)
HBeAg-positive CHB
Non-NAFLD
111
78 (70.270)
0.455
64 (57.658)
1.000
18 (16.216)
0.508
NAFLD
51
39 (76.471)
29 (56.863)
11 (21.569)
HBeAg-negative chronic HBV infection
Non-NAFLD
241
86 (35.685)
< 0.001
150 (62.241)
1.000
44 (18.257)
0.749
NAFLD
86
51 (59.302)
53 (61.628)
17 (19.767)
HBeAg-negative CHB
Non-NAFLD
88
57 (64.773)
0.325
61 (69.318)
0.397
19 (21.591)
0.848
NAFLD
69
39 (56.522)
43 (62.319)
16 (23.188)
Chronic HBV infection with no evidence of CHB
Non-NAFLD
381
147 (38.583)
< 0.001
216 (56.693)
0.524
59 (15.486)
0.160
NAFLD
118
73 (61.864)
71 (60.169)
25 (21.186)
CHB
Non-NAFLD
199
135 (67.839)
0.625
125 (62.814)
0.636
37 (18.593)
0.471
NAFLD
120
78 (65)
72 (60)
27 (22.5)
NAFLD: Non-alcoholic fatty liver disease; HbeAg: hepatitis B e antigen; CHB: chronic hepatitis B

Subgroup analysis based on metabolic factors

To increase the strength of the correlation between the virus and NAFLD, we make a list of subgroup analyses (Table 9). Firstly, patients with T2DM, hypertension and overweight (defined as BMI ≥ 23 kg/m2) were excluded (Subgroup 1). In this subgroup of a total of 409 patients, 41 had concurrent NAFLD, while the other 368 did not. The presence of significant liver inflammation in HBV-infected patients with NAFLD but no metabolic factors is borderline higher than those without NAFLD (60.976% vs. 45.109%, P = 0.069), but the prevalence of significant or advanced liver fibrosis between the two groups was comparable.
Table 9
Subgroup analysis based on metabolic factors
Subgroups
NAFLD or not
Total
Significant liver inflammation (%)
P value
Significant liver fibrosis (%)
P value
Advanced liver fibrosis (%)
P value
Subgroup 1
Non-NAFLD
368
166 (45.109)
0.069
212 (57.609)
0.623
59 (16.033)
1.000
NAFLD
41
25 (60.976)
22 (53.659)
6 (14.634)
Subgroup 2
Non-NAFLD
377
170 (45.093)
0.035
218 (57.825)
0.871
62 (16.446)
0.670
NAFLD
43
27 (62.791)
24 (55.814)
8 (18.605)
NAFLD: Non-alcoholic fatty liver disease; Subgroup 1: individuals with T2DM, hypertension and overweight were excluded; Subgroup 2: only lean HBV-infected subjects were included
We observed the presence of steatosis in lean individuals (BMI < 23 kg/m2) with HBV infection in this study was approximately 10%, similar with the overall prevalence of steatosis in lean individuals according to literature [15]. There were 420 lean HBV-infected subjects totally, and liver histology was compared in the NAFLD and non-NAFLD group (Subgroup 2). NAFLD group had higher prevalence of significant liver inflammation than the non-NAFLD group (62.791% vs. 45.093%, P = 0.035), but the presence of significant or advanced liver fibrosis between the two groups was similar.

Noninvasive models for significant liver inflammation and significant fibrosis

All included patients with and without significant liver inflammation or significant fibrosis were randomly selected with a ratio of 1:1. 80% of the patients were included in the derivation cohort, while the other 20% were included in the validation cohort.
The derivation cohort for significant inflammation was composed of 656 patients. Model score = 0.00592 + 0.86997 * AST − 0.40814 * ALT + 0.19083 * GGT − 0.32590 * ALB + 0.29954 * GLB + 0.31760 * (if steatosis) + 0.17445 * (if HbeAg positive) − 0.16393 * PLT. The AUROC of the model was 0.735. In the validation cohort of 164 patients, the AUROC was 0.825 (Fig. 1). At a cut-off value of 0.478, the model could determine significant hepatic inflammation with a sensitivity of 0.705, a specificity of 0.810, and a diagnostic accuracy of 0.758.
The derivation cohort for significant fibrosis was consisted of 555 patients. A model was developed with an AUROC of 0.702. Model score = 0.08888 + 0.20617 * (if hypertension) − 0.16412 * ALT + 0.32559 * AST + 0.45615 * GGT + 0.24697 * GLB − 0.41932 * Log10 HBV DNA. In the validation cohort of 110 patients, the AUROC was 0.760 (Fig. 2). At a cut-off value of 0.500, the model could diagnose significant fibrosis in CHB with a sensitivity of 0.725, a specificity of 0.714, and a diagnostic accuracy of 0.719.

Discussion

The current study highlighted the pathological findings in HBV-infected patients with and without concurrent NAFLD. HBV-infected patients with fatty liver had a higher severity of hepatic steatosis, hepatic inflammation, hepatic ballooning and a higher probability of advanced liver fibrosis than patients with simple chronic HBV infection. Fatty liver was not a risk factor for significant or advanced fibrosis, but it could independently predict significant liver inflammation in chronic HBV infection. Especially, in patients with either HBeAg-positive or HBeAg-negative chronic HBV infection, that is, in HBV-infected patients with persistent normal ALT, the presence of significant liver inflammation was higher in NAFLD than those without NAFLD. The prevalence of advanced liver fibrosis was higher in NAFLD than non-NAFLD group only in HBeAg-positive chronic HBV infection, while NAFLD did not increase fibrosis burden in other stages of HBV infection. Furthermore, we developed noninvasive models for significant liver inflammation and significant fibrosis with good diagnostic performance.
In 2020, a group of experts suggested the nomenclature of NAFLD should be updated to metabolic associated fatty liver disease (MAFLD) [16, 17], because metabolic liver disease usually coexists with other conditions such as viral hepatitis, and should not be described as a condition of “exclusion” [18]. We need to pay attention to patients with dual aetiology fatty liver disease who meet the criteria of NAFLD and who also have other concomitant condition. It is generally recognized that both NAFLD and HBV infection are common types of chronic hepatitis, and both can cause cirrhosis, hepatic failure and hepatocellular carcinoma [19, 20]. The coexistence of NAFLD and chronic HBV infection happens frequently. Therefore, it’s of great importance to explore the relationship between hepatic steatosis and HBV infection.
The interaction between NAFLD and HBV infection is complex and unclear. HBV infection might be related to decreased risk of NAFLD [2, 21], but the mechanisms whereby HBV influences steatosis has not been well understood. On the other hand, how hepatic steatosis influences the clinical outcomes of chronic HBV infection is not entirely clear. Wong et al. has found fatty liver, measured by controlled attenuation parameter (CAP), is associated with advanced fibrosis [6]. Mak’s study demonstrated hepatic steatosis promoted fibrosis progression in virologically quiescent CHB [22]. Another study by Seto et al. revealed that severe steatosis was related to severe fibrosis in both treatment-naive and on-treatment patients with HBV infection [5]. However, the definition of liver fibrosis in the above studies was determined by liver stiffness measurement (LSM) under transient elastography. As we know, moderate to severe hepatic steatosis might result in overestimation of LSM in HBV-infected patients, which should be considered seriously to avoid misdiagnosing fibrosis [23]. Therefore, histology by liver biopsy is always the golden standard. Even though in studies reporting the relationship between concomitant NAFLD and the severity of liver histology in patients based on liver biopsy, the results were not consistent [3, 4, 7].
In this study, we found that HBV-infected patients with concomitant NAFLD had a higher severity of portal inflammation and hepatic ballooning, and were more likely to develop significant liver inflammation and advanced fibrosis (borderline difference). To lighten the effects of confounding factors including age, gender and virological profiles, we performed further evaluation in 254 matched pairs. The results showed that chronic HBV infection with concurrent NAFLD had a greater severity of lobular and portal inflammation, hepatic ballooning and fibrosis, and were more likely to have significant inflammation and advanced fibrosis than simple HBV infection. But the probability of having significant fibrosis between the two groups was similar. Multivariate logistic analyses identified that steatosis was an independent predictor for only significant liver inflammation but not significant or advanced fibrosis in chronic HBV infection. While histologic alterations represent a continuous process, and persistent liver inflammation will definitely lead to the progression of fibrosis [24], it’s better to carry out a longitudinal cohort study to explore whether concomitant NAFLD in HBV-infection could promote liver fibrosis.
As we know, the natural history of HBV infection follows four disease phases with different levels of viral replication and dynamics in liver disease progression. As reported in this study, the influence of concurrent NAFLD on liver histology is different in distinct clinical phases in chronic HBV infection. Briefly, NAFLD could aggravate liver inflammation in HBV-infected patients with persistent normal ALT (immune tolerant and inactive carrier phase), and NAFLD increased the burden of advanced fibrosis only in the immune tolerant phase. This is a pretty novel and interesting finding. We should further explore the inside mechanism relating the interaction between NAFLD and distinct clinical phases in chronic HBV infection.
HBV replication and subsequent liver inflammation and fibrosis account for disease progression in CHB. Significant liver necroinflammation (grade G ≥ 2) and significant fibrosis (stage S ≥ 2) by liver histology greatly increase the risk of cirrhosis, hepatocellular carcinoma and end-stage liver diseases. As a consequence, early and timely antiviral therapy is recommended in these situations. Nevertheless, as the only golden standard for evaluating significant liver inflammation and fibrosis, liver biopsy is limited in clinical practice due to its invasive nature. Current noninvasive predictions of liver fibrosis mainly include LSM and biomarkers like Chitinase 3-like 1 (CHI3L1) [25, 26], Golgi protein 73 [27], et al. The detection of the newly-developed noninvasive markers has not yet been applied widely and their diagnostic accuracies are still limited. LSM could be impacted by a variety of factors like BMI, waist circumference, fatty liver, skin capsular distance and so on [28, 29]. Central obesity and fatty liver will lead to an overestimation of LSM in viral hepatitis including CHB [23] and chronic hepatitis C [30]. Given the gradually increasing prevalence and severity of NAFLD in HBV-infected patients, it’s necessary and has always been a research hotspot to develop noninvasive diagnostic models for significant liver inflammation and fibrosis. Our models were based on routine widely available clinical and laboratory parameters, and have presented with good diagnostic performance.
Despite our best effort, there were still some limitations in this study. Firstly, it was limited to a cross-sectional study, which was unable to evaluate the clinical outcomes of patients with concurrent NAFLD with HBV infection. Long-period follow-up and repeated liver biopsy are of great necessity to understand the influence of progression or regression of fatty liver on the histologic characteristics and clinical outcomes of the disease. Secondly, lack of data on more metabolic factors such as waist circumference and insulin resistance could also affect the interpretation of results. Last but not least, the mechanism behind interactions between NAFLD and HBV infection have not been explored in this study. As literatures report that specific diet and drugs can prevent the progression of chronic HBV infection and NAFLD [31, 32], further mechanism research is warranted in the purpose of reducing the synergistic negative effects of HBV and NAFLD, alleviating liver injury, and preventing decompensation events.

Conclusions

In conclusion, HBV-infected patients with concomitant NAFLD had a higher severity of liver inflammation, and a higher probability of significant liver inflammation and advanced fibrosis than simple chronic HBV infection. Hepatic steatosis could independently predict significant liver inflammation in nondrinker patients with chronic HBV infection, but it was not a risk factor for significant or advanced fibrosis. NAFLD could aggravate liver inflammation in the immune tolerant and inactive carrier phase of HBV infection, and increase the burden of advanced fibrosis only in the immune tolerant phase. In addition, noninvasive models established in this study could efficiently predict significant liver inflammation and significant fibrosis in patients with chronic HBV infection.

Acknowledgements

Not applicable.

Declarations

Competing interests

The authors declare no competing interests.
This study was approved by the Ethics Committee of People’s Hospital of Hangzhou Medical College (2021QT257) and followed the guidelines for studies in humans. Informed consents were obtained from all subjects.
Not applicable.
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.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Global prevalence. Treatment, and prevention of hepatitis B virus infection in 2016: a modelling study. Lancet Gastroenterol Hepatol. 2018;3(6):383–403.CrossRef Global prevalence. Treatment, and prevention of hepatitis B virus infection in 2016: a modelling study. Lancet Gastroenterol Hepatol. 2018;3(6):383–403.CrossRef
2.
Zurück zum Zitat Joo EJ, Chang Y, Yeom JS, Ryu S. Hepatitis B virus infection and decreased risk of nonalcoholic fatty liver disease: a cohort study. Hepatology. 2017;65(3):828–35.CrossRefPubMed Joo EJ, Chang Y, Yeom JS, Ryu S. Hepatitis B virus infection and decreased risk of nonalcoholic fatty liver disease: a cohort study. Hepatology. 2017;65(3):828–35.CrossRefPubMed
3.
Zurück zum Zitat Shi JP, Fan JG, Wu R, Gao XQ, Zhang L, Wang H, Farrell GC. Prevalence and risk factors of hepatic steatosis and its impact on liver injury in chinese patients with chronic hepatitis B infection. J Gastroenterol Hepatol. 2008;23(9):1419–25.CrossRefPubMed Shi JP, Fan JG, Wu R, Gao XQ, Zhang L, Wang H, Farrell GC. Prevalence and risk factors of hepatic steatosis and its impact on liver injury in chinese patients with chronic hepatitis B infection. J Gastroenterol Hepatol. 2008;23(9):1419–25.CrossRefPubMed
4.
Zurück zum Zitat Machado MV, Oliveira AG, Cortez-Pinto H. Hepatic steatosis in hepatitis B virus infected patients: meta-analysis of risk factors and comparison with hepatitis C infected patients. J Gastroenterol Hepatol. 2011;26(9):1361–7.CrossRefPubMed Machado MV, Oliveira AG, Cortez-Pinto H. Hepatic steatosis in hepatitis B virus infected patients: meta-analysis of risk factors and comparison with hepatitis C infected patients. J Gastroenterol Hepatol. 2011;26(9):1361–7.CrossRefPubMed
5.
Zurück zum Zitat Seto WK, Hui R, Mak LY, Fung J, Cheung KS, Liu K, Wong DK, Lai CL, Yuen MF. Association between hepatic steatosis, measured by controlled attenuation parameter, and Fibrosis Burden in Chronic Hepatitis B. Clin Gastroenterol Hepatol. 2018;16(4):575–83.CrossRefPubMed Seto WK, Hui R, Mak LY, Fung J, Cheung KS, Liu K, Wong DK, Lai CL, Yuen MF. Association between hepatic steatosis, measured by controlled attenuation parameter, and Fibrosis Burden in Chronic Hepatitis B. Clin Gastroenterol Hepatol. 2018;16(4):575–83.CrossRefPubMed
6.
Zurück zum Zitat Wong SW, Chan WK, Mohamed R. Fatty liver is associated with advanced fibrosis but does not predict adverse outcomes in patients with chronic hepatitis B. J Viral Hepat. 2020;27(12):1297–305.CrossRefPubMed Wong SW, Chan WK, Mohamed R. Fatty liver is associated with advanced fibrosis but does not predict adverse outcomes in patients with chronic hepatitis B. J Viral Hepat. 2020;27(12):1297–305.CrossRefPubMed
7.
Zurück zum Zitat Petta S, Camma C, Di Marco V, Macaluso FS, Maida M, Pizzolanti G, Belmonte B, Cabibi D, Di Stefano R, Ferraro D, Guarnotta C, Venezia G, Craxi A. Hepatic steatosis and insulin resistance are associated with severe fibrosis in patients with chronic hepatitis caused by HBV or HCV infection. LIVER INT. 2011;31(4):507–15.CrossRefPubMed Petta S, Camma C, Di Marco V, Macaluso FS, Maida M, Pizzolanti G, Belmonte B, Cabibi D, Di Stefano R, Ferraro D, Guarnotta C, Venezia G, Craxi A. Hepatic steatosis and insulin resistance are associated with severe fibrosis in patients with chronic hepatitis caused by HBV or HCV infection. LIVER INT. 2011;31(4):507–15.CrossRefPubMed
8.
Zurück zum Zitat Mak LY, Seto WK, Hui RW, Fung J, Wong DK, Lai CL, Yuen MF. Fibrosis evolution in chronic hepatitis B e antigen-negative patients across a 10-year interval. J Viral Hepat. 2019;26(7):818–27.CrossRefPubMed Mak LY, Seto WK, Hui RW, Fung J, Wong DK, Lai CL, Yuen MF. Fibrosis evolution in chronic hepatitis B e antigen-negative patients across a 10-year interval. J Viral Hepat. 2019;26(7):818–27.CrossRefPubMed
9.
Zurück zum Zitat Brunt EM, Ramrakhiani S, Cordes BG, Neuschwander-Tetri BA, Janney CG, Bacon BR, Di Bisceglie AM. Concurrence of histologic features of steatohepatitis with other forms of chronic liver disease. Mod Pathol. 2003;16(1):49–56.CrossRefPubMed Brunt EM, Ramrakhiani S, Cordes BG, Neuschwander-Tetri BA, Janney CG, Bacon BR, Di Bisceglie AM. Concurrence of histologic features of steatohepatitis with other forms of chronic liver disease. Mod Pathol. 2003;16(1):49–56.CrossRefPubMed
10.
Zurück zum Zitat EASL. 2017 Clinical Practice Guidelines on the management of hepatitis B virus infection. J HEPATOL 2017; 67(2): 370–398. EASL. 2017 Clinical Practice Guidelines on the management of hepatitis B virus infection. J HEPATOL 2017; 67(2): 370–398.
11.
Zurück zum Zitat Angulo P, Kleiner DE, Dam-Larsen S, Adams LA, Bjornsson ES, Charatcharoenwitthaya P, Mills PR, Keach JC, Lafferty HD, Stahler A, Haflidadottir S, Bendtsen F. Liver fibrosis, but No other histologic features, is Associated with Long-term outcomes of patients with nonalcoholic fatty liver disease. Gastroenterology. 2015;149(2):389–97.CrossRefPubMed Angulo P, Kleiner DE, Dam-Larsen S, Adams LA, Bjornsson ES, Charatcharoenwitthaya P, Mills PR, Keach JC, Lafferty HD, Stahler A, Haflidadottir S, Bendtsen F. Liver fibrosis, but No other histologic features, is Associated with Long-term outcomes of patients with nonalcoholic fatty liver disease. Gastroenterology. 2015;149(2):389–97.CrossRefPubMed
12.
Zurück zum Zitat Younossi ZM, Stepanova M, Rafiq N, Makhlouf H, Younoszai Z, Agrawal R, Goodman Z. Pathologic criteria for nonalcoholic steatohepatitis: interprotocol agreement and ability to predict liver-related mortality. Hepatology. 2011;53(6):1874–82.CrossRefPubMed Younossi ZM, Stepanova M, Rafiq N, Makhlouf H, Younoszai Z, Agrawal R, Goodman Z. Pathologic criteria for nonalcoholic steatohepatitis: interprotocol agreement and ability to predict liver-related mortality. Hepatology. 2011;53(6):1874–82.CrossRefPubMed
13.
Zurück zum Zitat Scheuer PJ. Classification of chronic viral hepatitis: a need for reassessment. J HEPATOL. 1991;13(3):372–4.CrossRefPubMed Scheuer PJ. Classification of chronic viral hepatitis: a need for reassessment. J HEPATOL. 1991;13(3):372–4.CrossRefPubMed
14.
Zurück zum Zitat Consensus on the diagnosis and treatment of hepatic fibrosis. (2019). J Dig Dis 2020; 21(3): 127–138. Consensus on the diagnosis and treatment of hepatic fibrosis. (2019). J Dig Dis 2020; 21(3): 127–138.
15.
Zurück zum Zitat Gao Y, Zhao T, Song S, Duo Y, Gao J, Yuan T, Zhao W. Lean nonalcoholic fatty liver disease and risk of incident type 2 diabetes mellitus: a literature review and meta-analysis. Diabetes Res Clin Pract. 2023;200:110699.CrossRefPubMed Gao Y, Zhao T, Song S, Duo Y, Gao J, Yuan T, Zhao W. Lean nonalcoholic fatty liver disease and risk of incident type 2 diabetes mellitus: a literature review and meta-analysis. Diabetes Res Clin Pract. 2023;200:110699.CrossRefPubMed
16.
Zurück zum Zitat Eslam M, Sanyal AJ, George J. MAFLD: A Consensus-Driven proposed nomenclature for metabolic Associated fatty liver disease. Gastroenterology. 2020;158(7):1999–2014.CrossRefPubMed Eslam M, Sanyal AJ, George J. MAFLD: A Consensus-Driven proposed nomenclature for metabolic Associated fatty liver disease. Gastroenterology. 2020;158(7):1999–2014.CrossRefPubMed
17.
Zurück zum Zitat Eslam M, Newsome PN, Sarin SK, Anstee QM, Targher G, Romero-Gomez M, Zelber-Sagi S, Wai-Sun WV, Dufour JF, Schattenberg JM, Kawaguchi T, Arrese M, Valenti L, Shiha G, Tiribelli C, Yki-Jarvinen H, Fan JG, Gronbaek H, Yilmaz Y, Cortez-Pinto H, Oliveira CP, Bedossa P, Adams LA, Zheng MH, Fouad Y, Chan WK, Mendez-Sanchez N, Ahn SH, Castera L, Bugianesi E, Ratziu V, George J. A new definition for metabolic dysfunction-associated fatty liver disease: an international expert consensus statement. J HEPATOL. 2020;73(1):202–9.CrossRefPubMed Eslam M, Newsome PN, Sarin SK, Anstee QM, Targher G, Romero-Gomez M, Zelber-Sagi S, Wai-Sun WV, Dufour JF, Schattenberg JM, Kawaguchi T, Arrese M, Valenti L, Shiha G, Tiribelli C, Yki-Jarvinen H, Fan JG, Gronbaek H, Yilmaz Y, Cortez-Pinto H, Oliveira CP, Bedossa P, Adams LA, Zheng MH, Fouad Y, Chan WK, Mendez-Sanchez N, Ahn SH, Castera L, Bugianesi E, Ratziu V, George J. A new definition for metabolic dysfunction-associated fatty liver disease: an international expert consensus statement. J HEPATOL. 2020;73(1):202–9.CrossRefPubMed
18.
Zurück zum Zitat Fouad Y, Waked I, Bollipo S, Gomaa A, Ajlouni Y, Attia D. What’s in a name? Renaming ‘NAFLD’ to ‘MAFLD’. LIVER INT. 2020;40(6):1254–61.CrossRefPubMed Fouad Y, Waked I, Bollipo S, Gomaa A, Ajlouni Y, Attia D. What’s in a name? Renaming ‘NAFLD’ to ‘MAFLD’. LIVER INT. 2020;40(6):1254–61.CrossRefPubMed
19.
Zurück zum Zitat Chan TT, Chan WK, Wong GL, Chan AW, Nik MN, Chan SL, Chong CC, Mahadeva S, Shu SS, Lai PB, Chan HL, Wong VW. Positive Hepatitis B Core antibody is Associated with cirrhosis and Hepatocellular Carcinoma in nonalcoholic fatty liver disease. AM J GASTROENTEROL. 2020;115(6):867–75.CrossRefPubMed Chan TT, Chan WK, Wong GL, Chan AW, Nik MN, Chan SL, Chong CC, Mahadeva S, Shu SS, Lai PB, Chan HL, Wong VW. Positive Hepatitis B Core antibody is Associated with cirrhosis and Hepatocellular Carcinoma in nonalcoholic fatty liver disease. AM J GASTROENTEROL. 2020;115(6):867–75.CrossRefPubMed
20.
Zurück zum Zitat Chan AW, Wong GL, Chan HY, Tong JH, Yu YH, Choi PC, Chan HL, To KF, Wong VW. Concurrent fatty liver increases risk of hepatocellular carcinoma among patients with chronic hepatitis B. J Gastroenterol Hepatol. 2017;32(3):667–76.CrossRefPubMed Chan AW, Wong GL, Chan HY, Tong JH, Yu YH, Choi PC, Chan HL, To KF, Wong VW. Concurrent fatty liver increases risk of hepatocellular carcinoma among patients with chronic hepatitis B. J Gastroenterol Hepatol. 2017;32(3):667–76.CrossRefPubMed
21.
Zurück zum Zitat Huang J, Jing M, Wang C, Wang M, You S, Lin S, Zhu Y. The impact of hepatitis B virus infection status on the prevalence of nonalcoholic fatty liver disease: a population-based study. J MED VIROL. 2020;92(8):1191–7.CrossRefPubMed Huang J, Jing M, Wang C, Wang M, You S, Lin S, Zhu Y. The impact of hepatitis B virus infection status on the prevalence of nonalcoholic fatty liver disease: a population-based study. J MED VIROL. 2020;92(8):1191–7.CrossRefPubMed
22.
Zurück zum Zitat Mak LY, Hui RW, Fung J, Liu F, Wong DK, Cheung KS, Yuen MF, Seto WK. Diverse effects of hepatic steatosis on fibrosis progression and functional cure in virologically quiescent chronic hepatitis B. J HEPATOL. 2020;73(4):800–6.CrossRefPubMed Mak LY, Hui RW, Fung J, Liu F, Wong DK, Cheung KS, Yuen MF, Seto WK. Diverse effects of hepatic steatosis on fibrosis progression and functional cure in virologically quiescent chronic hepatitis B. J HEPATOL. 2020;73(4):800–6.CrossRefPubMed
23.
Zurück zum Zitat Shen F, Mi YQ, Xu L, Liu YG, Wang XY, Pan Q, Zhang RN, Hu XQ, Xu LM, Fan JG. Moderate to severe hepatic steatosis leads to overestimation of liver stiffness measurement in chronic hepatitis B patients without significant fibrosis. Aliment Pharmacol Ther. 2019;50(1):93–102.CrossRefPubMed Shen F, Mi YQ, Xu L, Liu YG, Wang XY, Pan Q, Zhang RN, Hu XQ, Xu LM, Fan JG. Moderate to severe hepatic steatosis leads to overestimation of liver stiffness measurement in chronic hepatitis B patients without significant fibrosis. Aliment Pharmacol Ther. 2019;50(1):93–102.CrossRefPubMed
24.
Zurück zum Zitat Sonneveld MJ, Brouwer WP, Hansen BE, Chan HL, Piratvisuth T, Jia JD, Zeuzem S, Chien RN, Choi H, de Knegt RJ, Wat C, Pavlovic V, Gaggar A, Xie Q, Buti M, de Man RA, Janssen H. Very low probability of significant liver inflammation in chronic hepatitis B patients with low ALT levels in the absence of liver fibrosis. Aliment Pharmacol Ther. 2020;52(8):1399–406.CrossRefPubMedPubMedCentral Sonneveld MJ, Brouwer WP, Hansen BE, Chan HL, Piratvisuth T, Jia JD, Zeuzem S, Chien RN, Choi H, de Knegt RJ, Wat C, Pavlovic V, Gaggar A, Xie Q, Buti M, de Man RA, Janssen H. Very low probability of significant liver inflammation in chronic hepatitis B patients with low ALT levels in the absence of liver fibrosis. Aliment Pharmacol Ther. 2020;52(8):1399–406.CrossRefPubMedPubMedCentral
25.
Zurück zum Zitat Huang H, Wu T, Mao J, Fang Y, Zhang J, Wu L, Zheng S, Lin B, Pan H. CHI3L1 is a Liver-Enriched, Noninvasive Biomarker that can be used to stage and diagnose substantial hepatic fibrosis. OMICS. 2015;19(6):339–45.CrossRefPubMedPubMedCentral Huang H, Wu T, Mao J, Fang Y, Zhang J, Wu L, Zheng S, Lin B, Pan H. CHI3L1 is a Liver-Enriched, Noninvasive Biomarker that can be used to stage and diagnose substantial hepatic fibrosis. OMICS. 2015;19(6):339–45.CrossRefPubMedPubMedCentral
26.
Zurück zum Zitat Wang L, Liu T, Zhou J, You H, Jia J. Changes in serum chitinase 3-like 1 levels correlate with changes in liver fibrosis measured by two established quantitative methods in chronic hepatitis B patients following antiviral therapy. HEPATOL RES. 2018;48(3):E283–90.CrossRefPubMed Wang L, Liu T, Zhou J, You H, Jia J. Changes in serum chitinase 3-like 1 levels correlate with changes in liver fibrosis measured by two established quantitative methods in chronic hepatitis B patients following antiviral therapy. HEPATOL RES. 2018;48(3):E283–90.CrossRefPubMed
27.
Zurück zum Zitat Cao Z, Li Z, Wang Y, Liu Y, Mo R, Ren P, Chen L, Lu J, Li H, Zhuang Y, Liu Y, Wang X, Zhao G, Tang W, Xiang X, Wang H, Cai W, Liu L, Zhu C, Bao S, Xie Q. Assessment of serum golgi protein 73 as a biomarker for the diagnosis of significant fibrosis in patients with chronic HBV infection. J Viral Hepat. 2017;24(Suppl 1):57–65.CrossRefPubMed Cao Z, Li Z, Wang Y, Liu Y, Mo R, Ren P, Chen L, Lu J, Li H, Zhuang Y, Liu Y, Wang X, Zhao G, Tang W, Xiang X, Wang H, Cai W, Liu L, Zhu C, Bao S, Xie Q. Assessment of serum golgi protein 73 as a biomarker for the diagnosis of significant fibrosis in patients with chronic HBV infection. J Viral Hepat. 2017;24(Suppl 1):57–65.CrossRefPubMed
28.
Zurück zum Zitat Wong GL, Wong VW, Chim AM, Yiu KK, Chu SH, Li MK, Chan HL. Factors associated with unreliable liver stiffness measurement and its failure with transient elastography in the chinese population. J Gastroenterol Hepatol. 2011;26(2):300–5.CrossRefPubMed Wong GL, Wong VW, Chim AM, Yiu KK, Chu SH, Li MK, Chan HL. Factors associated with unreliable liver stiffness measurement and its failure with transient elastography in the chinese population. J Gastroenterol Hepatol. 2011;26(2):300–5.CrossRefPubMed
29.
Zurück zum Zitat Karlas T, Petroff D, Sasso M, Fan JG, Mi YQ, de Ledinghen V, Kumar M, Lupsor-Platon M, Han KH, Cardoso AC, Ferraioli G, Chan WK, Wong VW, Myers RP, Chayama K, Friedrich-Rust M, Beaugrand M, Shen F, Hiriart JB, Sarin SK, Badea R, Lee HW, Marcellin P, Filice C, Mahadeva S, Wong GL, Crotty P, Masaki K, Bojunga J, Bedossa P, Keim V, Wiegand J. Impact of controlled attenuation parameter on detecting fibrosis using liver stiffness measurement. Aliment Pharmacol Ther. 2018;47(7):989–1000.CrossRefPubMed Karlas T, Petroff D, Sasso M, Fan JG, Mi YQ, de Ledinghen V, Kumar M, Lupsor-Platon M, Han KH, Cardoso AC, Ferraioli G, Chan WK, Wong VW, Myers RP, Chayama K, Friedrich-Rust M, Beaugrand M, Shen F, Hiriart JB, Sarin SK, Badea R, Lee HW, Marcellin P, Filice C, Mahadeva S, Wong GL, Crotty P, Masaki K, Bojunga J, Bedossa P, Keim V, Wiegand J. Impact of controlled attenuation parameter on detecting fibrosis using liver stiffness measurement. Aliment Pharmacol Ther. 2018;47(7):989–1000.CrossRefPubMed
30.
Zurück zum Zitat Dulay MT, Zaman N, Jaramillo D, Mody AC, Zare RN. Pathogen-Imprinted Organosiloxane Polymers as selective biosensors for the detection of targeted E. coli. C (Basel). 2018;4(2):29.PubMed Dulay MT, Zaman N, Jaramillo D, Mody AC, Zare RN. Pathogen-Imprinted Organosiloxane Polymers as selective biosensors for the detection of targeted E. coli. C (Basel). 2018;4(2):29.PubMed
31.
Zurück zum Zitat Di Francia R, Rinaldi L, Cillo M, Varriale E, Facchini G, D’Aniello C, Marotta G, Berretta M. Antioxidant diet and genotyping as tools for the prevention of liver disease. Eur Rev Med Pharmacol Sci. 2016;20(24):5155–63.PubMed Di Francia R, Rinaldi L, Cillo M, Varriale E, Facchini G, D’Aniello C, Marotta G, Berretta M. Antioxidant diet and genotyping as tools for the prevention of liver disease. Eur Rev Med Pharmacol Sci. 2016;20(24):5155–63.PubMed
32.
Zurück zum Zitat Palmiero G, Cesaro A, Vetrano E, Pafundi PC, Galiero R, Caturano A, Moscarella E, Gragnano F, Salvatore T, Rinaldi L, Calabro P, Sasso FC. Impact of SGLT2 inhibitors on Heart failure: from pathophysiology to Clinical Effects. INT J MOL SCI 2021; 22(11). Palmiero G, Cesaro A, Vetrano E, Pafundi PC, Galiero R, Caturano A, Moscarella E, Gragnano F, Salvatore T, Rinaldi L, Calabro P, Sasso FC. Impact of SGLT2 inhibitors on Heart failure: from pathophysiology to Clinical Effects. INT J MOL SCI 2021; 22(11).
Metadaten
Titel
Fatty liver is associated with significant liver inflammation and increases the burden of advanced fibrosis in chronic HBV infection
verfasst von
Yi-Ning Dai
Cheng-Fu Xu
Hong-Ying Pan
Mei-Juan Chen
Chao-Hui Yu
Publikationsdatum
01.12.2023
Verlag
BioMed Central
Erschienen in
BMC Infectious Diseases / Ausgabe 1/2023
Elektronische ISSN: 1471-2334
DOI
https://doi.org/10.1186/s12879-023-08632-y

Weitere Artikel der Ausgabe 1/2023

BMC Infectious Diseases 1/2023 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Erhebliches Risiko für Kehlkopfkrebs bei mäßiger Dysplasie

29.05.2024 Larynxkarzinom Nachrichten

Fast ein Viertel der Personen mit mäßig dysplastischen Stimmlippenläsionen entwickelt einen Kehlkopftumor. Solche Personen benötigen daher eine besonders enge ärztliche Überwachung.

Nach Herzinfarkt mit Typ-1-Diabetes schlechtere Karten als mit Typ 2?

29.05.2024 Herzinfarkt Nachrichten

Bei Menschen mit Typ-2-Diabetes sind die Chancen, einen Myokardinfarkt zu überleben, in den letzten 15 Jahren deutlich gestiegen – nicht jedoch bei Betroffenen mit Typ 1.

15% bedauern gewählte Blasenkrebs-Therapie

29.05.2024 Urothelkarzinom Nachrichten

Ob Patienten und Patientinnen mit neu diagnostiziertem Blasenkrebs ein Jahr später Bedauern über die Therapieentscheidung empfinden, wird einer Studie aus England zufolge von der Radikalität und dem Erfolg des Eingriffs beeinflusst.

Costims – das nächste heiße Ding in der Krebstherapie?

28.05.2024 Onkologische Immuntherapie Nachrichten

„Kalte“ Tumoren werden heiß – CD28-kostimulatorische Antikörper sollen dies ermöglichen. Am besten könnten diese in Kombination mit BiTEs und Checkpointhemmern wirken. Erste klinische Studien laufen bereits.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.