HISTOLOGICAL AND IMMUNOHISTOCHEMICAL STUDY OF HEPATIC STELLATE CELLS PRE AND POST-TRANSPLANTED HUMAN LIVER

Document Type : Original Article

Authors

1 Department of Medical Histology and Cell Biology, Al-Azhar Faculty of Medicine in Cairo

2 Department of Medical Histology and Cell Biology, Al-Azhar Faculty of Medicine in Assiut

3 Department of Pathology, Military Medical Academy

Abstract

Background: Hepatic stellate cells (HSCs) regulate vitamin A metabolism and play a vital role during the activation of the immune response. During liver damage, HSCs transform to myofibroblast-like cells, leading to loss of their lipid content and synthesis of extracellular matrix (ECM) inducing liver fibrosis.
Objective: The present study aimed to establish the correlation between HSCs activity within different areas of hepatic tissue and the degree of liver fibrosis in individuals with normal liver, hepatitis C virus infected patients and post-transplanted liver.
Patients and methods: The study involved thirty four cases from the international medical center classified into three groups: group 1 included ten healthy individuals as control, group 2 included twelve patients with chronic hepatitis C virus and liver cirrhosis and group 3 included twelve patients with post-transplanted liver due to liver previous hepatitis C virus infection and hepatic cirrhosis subdivided into group 3a five patients who received antihepatitis C treatment, and group 3b seven patients who didn’t receive antihepatitis C treatment. We used H&E and Masson trichrome stains, immunohistochemical detection of á-smooth muscle actin (á-SMA), Glial Fibrillary acidic protein (GFAP) and transmission electron microscopy (TEM).
Results: H&E stain revealed hepatic tissue with preserved architecture in group 1, disrupted architecture and areas of feathery degeneration in group 2, normal tissue histology in group 3a and moderate cellular infiltration in group 3b. Masson trichrome stain revealed normal collagen fibers distribution in group1, advanced fibrosis in group 2, no fibrotic changes in group 3a and few foci of bridging fibrosis in group 3b.  Immunohistochemical analysis revealed increased expression of α-SMA in group 2 and group 3b in comparison to group 1 and group 3a. Immunohistochemical analysis revealed increased expression of  GFAP in group 3b in comparison to group 3a, group 2 and group 1. TEM showed increase in the fibrous tissue and degeneration in the ultrastructure of the hepatocyte with few lipid droplets in HSCs of group 2 compared to group 1.
Conclusion: The hepatic stellate cells play an important role in the fibrosis of the liver damaged by hepatitis C virus and in the post-transplanted liver which not treated from HCV.

Keywords


HISTOLOGICAL AND IMMUNOHISTOCHEMICAL STUDY OF HEPATIC STELLATE CELLS PRE AND POST-TRANSPLANTED HUMAN LIVER

By

 

Mahmoud A.A. Masoud1, Mohammad A. Kasem2, Ayman F. El-Sharwy1, Amr F. El-Sebaey3 and Tamer I. Abd El-Rahman2

Department of Medical Histology and Cell Biology, Al-Azhar Faculty of Medicine in Cairo1 and in Assiut2 and Department of Pathology, Military Medical Academy3

Correspondence: Tamer Ibrahim Abd El-Rahman Ahmed

E-mail: Tamer23101982@gmail.com, Tel.: 01222310146

                                                ABSTRACT                    

Background: Hepatic stellate cells (HSCs) regulate vitamin A metabolism and play a vital role during the activation of the immune response. During liver damage, HSCs transform to myofibroblast-like cells, leading to loss of their lipid content and synthesis of extracellular matrix (ECM) inducing liver fibrosis.

Objective: The present study aimed to establish the correlation between HSCs activity within different areas of hepatic tissue and the degree of liver fibrosis in individuals with normal liver, hepatitis C virus infected patients and post-transplanted liver.

Patients and methods: The study involved thirty four cases from the international medical center classified into three groups: group 1 included ten healthy individuals as control, group 2 included twelve patients with chronic hepatitis C virus and liver cirrhosis and group 3 included twelve patients with post-transplanted liver due to liver previous hepatitis C virus infection and hepatic cirrhosis subdivided into group 3a five patients who received antihepatitis C treatment, and group 3b seven patients who didn’t receive antihepatitis C treatment. We used H&E and Masson trichrome stains, immunohistochemical detection of á-smooth muscle actin (á-SMA), Glial Fibrillary acidic protein (GFAP) and transmission electron microscopy (TEM).

Results: H&E stain revealed hepatic tissue with preserved architecture in group 1, disrupted architecture and areas of feathery degeneration in group 2, normal tissue histology in group 3a and moderate cellular infiltration in group 3b. Masson trichrome stain revealed normal collagen fibers distribution in group1, advanced fibrosis in group 2, no fibrotic changes in group 3a and few foci of bridging fibrosis in group 3b.  Immunohistochemical analysis revealed increased expression of α-SMA in group 2 and group 3b in comparison to group 1 and group 3a. Immunohistochemical analysis revealed increased expression of  GFAP in group 3b in comparison to group 3a, group 2 and group 1. TEM showed increase in the fibrous tissue and degeneration in the ultrastructure of the hepatocyte with few lipid droplets in HSCs of group 2 compared to group 1.

Conclusion: The hepatic stellate cells play an important role in the fibrosis of the liver damaged by hepatitis C virus and in the post-transplanted liver which not treated from HCV.

Keywords: HSCs – HCV – fibrosis – post-transplanted liver.

 

 

 

 

 

INTRODUCTION

     HSCs are cells characterized by the presence of cytoplasmic droplets of fat which contain vitamin A (Juan et al., 2019). Acute and chronic liver cell damage leads to activation and transformation of HSCs into myofibroblast-like cells which are responsible for synthesis of ECM and their loss of intracellular lipid droplets (Kasztelan-Szczerbińska et al., 2010 and Ionescu et al., 2013).

     Activation of HSCs consists of two major phases, i.e. initiation and perpetuation, followed by a resolution phase if the liver injury subsides (Friedman and Lee, 2011). During resolution, loss of activated stellate cells occurs by apoptosis or it becomes senescent or reverts to a quiescent phenotype (Kisseleva et al., 2012 and Troeger et al., 2012). The apoptosis and necrosis of the parenchymal cells may contribute to the activation of HSCs as well as liver macrophages. Activated HSCs and liver resident macrophages engage other immune cells such as circulating monocytes, T cells and neutrophils by secretion of pro-inflammatory mediators (Pellicoro et al., 2014).

     The majority of patients with chronic HCV infection develop hepatic cirrhosis, failure or hepatocellular carcinoma (Bhatia et al., 2014). The history of infection and the disease progression is influenced by various factors such as level of HCV infection, age of onset, sex, co-infection with hepatitis B virus and duration of infection (Barakat et al., 2013). Once the causative agent of damage is removed, the progression of liver disease is attenuated and recovery is a possible scenario even if the patient has state of fibrosis (Salas Villalobos et al., 2017).

     End stage liver disease due to hepatitis C virus (HCV) infection remains a leading indication for liver transplantation (LT) worldwide. Recurrent HCV infection is associated with more rapid fibrosis progression leading to higher rates of graft loss and patient mortality compared to patients transplanted for non-HCV etiologies. Re-transplantation for allograft cirrhosis due to recurrent HCV remains controversial (deLemos et al., 2014).

     The aim of this study was to establish a correlation between hepatic stellate cells (HSCs) activity and the degree of liver fibrosis in individuals with healthy liver, hepatitis C infected patients, and post-transplanted liver.

PATIENTS AND METHODS

     Patients were selected from the international medical center with ages ranged from 20-65 years in the period of March 2017 to April 2019. Liver biopsy specimens were obtained percutaneously, using an automatic Autovac gun with Tru-Cut® needle with a diameter of 18-G. Only samples longer than 20 mm with more than eight portal tracts were included in this study.  Informed consent from all patients and controls was obtained in accordance with the local ethical committee.

     Thirty four cases were included in this study and classified into 3 groups: group 1 included ten healthy individuals (donor for liver transplantation) serve as a control, group 2 included twelve patients with chronic hepatitis C and liver fibrosis and group 3 included twelve patients with post-transplanted liver divided into group 3a five patients who received anti-hepatitis C treatment and group 3b seven patients who did not receive anti-hepatitis C treatment.

     We used H&E stain for morphological examination, Masson trichrome stain to detect fibrosis, immunohistochemistry of α-SMA, GFAP to detect HSCs activation. We also used a transmission electron microscope to detect the ultrastructure of the hepatocytes and HSCs content. The quantification of hepatic stellate cells was performed in liver biopsies of all groups using Optimas image analysis software. SPSS program (version 16) was used to evaluate the results and assessment of immunoreactivity using the semi quantitative method by determining the percentage of positive cells by Olympus BX50F4 microscope.

Statistical analysis:

     Data were collected, revised coded and entered to the statistical package for social sciences (SPSS), version 16. Qualitative data were presented as number and percentages, while quantitative data were presented as mean, standard deviations, median and interquartile range. The comparison between four groups with qualitative data were done using Fisher exact test. The comparison between four groups regarding quantitative data with parametric distribution was done using ANOVA test followed by post hoc analysis for multiple comparison and as regarding quantitative data with non-parametric distribution was done using Kruskal-Wallis test. P-value was considered significant when P was < 0.05.


 

RESULTS

 

 

Hematoxylin and eosin sections:

Group 1: Showed normal liver architecture (figure 1a, b).

Group2: Liver tissues showed cirrhotic nodules and disrupted architecture and areas of feathery degeneration. The portal tracts revealed biliary epithelial dysplastic changes and moderate cellular infiltration (figure 1c). Other tissue samples showed steatosis infiltrated by malignant epithelial cells (figure 1d).

Group 3a: Showed preserved architecture with mild cellular infiltration (figure 1e).

Group 3b: Revealed areas of centrilobular degeneration in the form of marked diffuse macrovesicular and microvesicular steatosis and portal cellular infiltration (figure 1f).

Masson’s trichrome sections:

Group 1: Reveled normal collagen fibers distribution (figure 2a).

Group 2:  Reveled advanced fibrosis around the central vein and periportal fibrosis (figure 2b).

 Group 3a: Reveled no significant fibrosis (figure 2c).

 Group 3b: Reveled few foci of fibrosis (figure 2d).


 

 

 

 

 

 
   

Figure (1): Section in the liver showed a, b) Group1showed normal architechire of liver tissue (H and E X 200). c, d) Group2 showed disrupted architecture of liver tissue. The portal tracts reveal biliary epithelial dysplastic changes and mild cellular infiltration (H and E X 200). e) Group3a showed hepatocytes with preserved architecture with microvesicular Steatosis (H and E X 20). f) Group3b showed diffuse macrovesicular and microvesicular steatosis, portal inflammation with cellular infiltration and widening of portal tracts (H and E X 200).

Figure (2): Section in the liver showed a) Normal collagen fibers distrubution in group1 (Masson trichrome stain X 200). b) Massive periportal fibrosis and surrounding central vein in group2 (Masson trichrome stain X 200). c) No fibrotic changes in group3a (Masson trichrome stain (X 200). d) Portal tract with foci of bridging fibrosis in group3b (Masson trichrome stain X 200).

 

Immunohistochemical results:

Control positive tissue: Tissue samples from surgically excised uterus after hysterectomy were fixed, processed and embedded in the same manner as the liver biopsies and used as positive control for immunohistochemical detection of monoclonal α-SMA (figure 3a). Tissue samples from surgically excised human astrocytoma were fixed, processed and embedded in the same manner as the liver biopsies and used as positive control for the GFAP (figure 3b).

Control negative tissue: Tissue samples from surgically excised lipoma were fixed, processed and embedded in the same manner as the liver biopsies and used as negative control for the monoclonal α-SMA (figure 3c). Tissue sections from liver biopsy were cut and processed by immunohistochemistry without adding GFAP antibodies were used as a negative control (figure 3d).

Alpha Smooth muscle actin:

Group1: Showed negative expression to α-SMA antibody (figure 4a).

Group 2: Considerable expression of α-SMA antibody reflecting fibrosis in tissue samples (figure 4b).

 

 
   

Group3a: Negative expression to α-SMA (figure 4c).

Group3b: Positive expression to α-SMA antibody (figure 4d).

Glial Fibrillary Acidic Protein:

Group1: Showed mild expression to GFAP (figure 5a).

Group2: There was no significant expression (figure 5b).

Group3a: Mild expression of GFAP in (figure 5c).

Group3b: Revealed the highest percentage of GFAP-positive hepatic stellate cells indicating activation of HSCs (figure 5d).

Transmission electron microscopy:

Group1: Ultrathin sections of liver cells revealed normal hepatic architecture (figures 6a and 6b).

Group 2: Showed marked alteration in the normal structure of the hepatocyte. The nuclei of liver cells were irregular in shape with heterochromatin. The cytoplasm was highly vacuolated with a large lipid droplet, the rough endoplasmic reticulum appeared swollen and destructed, the mitochondria were swollen with destructed cristae (figure 7 a). HSCs surrounded by fibrous tissue with a few fat droplets in its cytoplasm.  The space of Disse contained collagen bundles and the blood sinusoids (SD) contain Kupffer cell (K) with intended nucleus (figure 7 b).


Figure (3): (a) Control positive uterus for α-SMA (X 200). (b) Control positive human astrocytoma tissue to GFAP (x200). (c) Control negative lipoma to α-SMA (x400). (d) Control negative liver tissue to GFAP without adding the GFAP primary monoclonal antibody (x400).

 

 
   

Figure (4): Sections stained by α-SMA antibody showed a) Negative immunoreactivity to α-SMA antibody in group1 (X200). b) Positive staining was distributed widely within the lobule in group 2 (X 200). c) Negative expression to α-SMA in group 3a d) Positive staining distributed in the periphery and within portal tract in group 3b.

Figure (5): Section stained by GFAP antibody showed a) Mild positive staining (blue arrow) in group1 (X 200). b) Mild positive staining (blue arrow) in group2 (X200). C) Mild positive staining in group3a (X 200). d) Positive staining near to portal tract and along hepatic lobule in group3b (blue arrow) (X200).

 

 

Figure (6): TEM of liver section in group 1 showed a) Normal hepatocyte (H) with the rounded euchromatic nucleus and crowded cytoplasm with cell organelles, mitochondria (M), (RER) and glycogen granules (X10000). b) Stellate cells adherent to the outer surface of the endothelium. The cytoplasm filed with lipid droplets and the nucleus is  indented (X10000).

Figure (7): TEM of group 2 showing a) Cytoplasm with a large fat droplet (F). rER appeared swollen destructed. The mitochondria were swollen with destructed cristae. (X10000). b) The cytoplasm appeared pale and highly vacuolated. HSC surrounded by fibrous tissue showing few fat droplets in the cytoplasm and the space of Disse contained many collagen bundles. (K) Kupffer cell with the intended nucleus. (X2500).

 
   

 

 

Characteristics and age of the studied groups:

     The obtained results  showed  that The mean age of the studied groups  among the control was  30.7 years versus (59.25, 55.14& 55.6) years among hepatitis C virus patients, post-transplant without HCV treatment patients and post-transplant with HCV treatment patients  respectively. There was a statistical significant difference between the studied groups regard age (table 1).


 

Table (1): Characteristics and age of the studied groups

P

Post-transplant with HCV treatment

Post-transplant without HCV treatment

HCV

Control

Groups

Items

<0.01*

n = 5

n=7

n=12

n=10

Number of patients / group

55.6 ± 4.9

55.14 ± 8.7

59.25 ± 3.8

30.7 ± 7.5

Age (years)

(Mean + SD)

 

P1value < 0.01*                  P2value < 0.01*            P 3value < 0.01*

P 4value= 0.2                     P 5value=0.9                P 6value=0.1

P1 == represent the statistical difference between control and HCV cases

P2 = represent the statistical difference between control and post-transplant without HCV treatment cases.

P3= represent the statistical difference between control and post-transplant with HCV treatment cases.

P4= represent the statistical difference between post-transplant with HCV treatment cases and chronic HCV cases.

P5= represent the statistical difference between post-transplant with HCV treatment cases and post-transplant without HCV treatment cases.

P6= represent the statistical difference between post-transplant without HCV treatment cases and chronic HCV cases.

 

 

     The obtained results showed that there was a statistical significant difference between means and frequencies of the studied groups regarding fibrosis scale and fibrosis stage respectively (table 2).

 

 

Table (2): Difference between studied groups as regard fibrosis

P

Post-transplant with HCV treatment (N=5)

Post-transplant without HCV treatment (N=7)

HCV (N=12)

 

Control (N=10)

Groups

 

Parameters

< 0.01*

 

 

 

1.2 ± 0.6

1

(0.7 – 1.7)

 

 

 

0.9 ± 0.8

1

(0 – 1.5)

 

 

2.4 ± 1.01

2

(1.6 – 3.3)

 

 

0.04 ± 0.05

0.00

(0 - 0.1)

Fibrosis scale

 

-Mean± S.D.

-Median

-Interquartile range

< 0.01*

 

%

No

%

No

%

No

%

No

 

 

 

 

Fibrosis stage:

No

Mild to moderate

Sever

 

 

20

80

0

 

 

1

4

0

 

 

42.9

57.1

0

 

 

3

4

0

 

 

0

58.3

41.7

 

 

0

7

5

 

 

100

0

0

 

 

10

0

0

 

 

     The obtained results showed statistically significant increase in area stained by α smooth muscle actin (α SMA) by (40% & 51.6%) in HCV group and  post-transplant without HCV treatment  group respectively in comparison to the control group. However, there was a statistically significant decrease in area stained by α smooth muscle actin (α SMA) by 9.8% in post-transplant with HCV treatment  group in comparison to the control group (table 3).

 

 

Table (3): Immunohistochemical staining of α smooth muscle actin

Groups

 

 

 

Parameter

Control (n=10)

HCV

(n=12)

Post-transplant without HCV treatment (n=7)

Post-transplant with HCV treatment (n=5)

P

Area stained by

α smooth muscle actin (α SMA)

 

-Mean± S.D.

-Median

-Interquartile range

 

 

 

 

10.02 ± 5.11

11.05

(5.08 – 13.8)

 

 

 

 

14.1 ± 4.2

9.7

(10.2 – 18.4)

 

 

 

 

9.1 ± 3.1

8.2

(7.9 – 12.9)

 

 

 

 

15.2 ± 3.8

14

( 12.1 – 18.8)

 

< 0.03*

 

 

     The obtained results showed  a statistically significant increase in area stained by Glial Fibrillary acidic protein (GFAP) by (12.5% , 87.5% &12.5%) in HCV group,  post-transplant without HCV treatment  group and post-transplant with HCV treatment  group respectively in comparison to the control group (table 4).


 

Table (4): Immunohistochemical staining of Glial Fibrillary Acidic Protein

Groups

 

 

 

Parameter

Control (n=10)

HCV (n=12)

Post-transplant without HCV treatment (n=7)

Post-transplant with HCV treatment (n=5)

P

Area stained by Glial Fibrillary Acidic Protein

(GFAP)

 

-Mean± S.D.

-Median

-Interquartile range

 

 

 

 

 

0.8 ± 0.3

0.81

(0.4  - 1)

 

 

 

 

 

0.9 ± 0.2

0.9

(0.7 – 1)

 

 

 

 

 

1.5 ± 0.8

1.1

(1.05 – 1.6)

 

 

 

 

 

0.9 ± 0.05

0.85

(0.7 – 0.9)

 

< 0.01*

 

 

DISCUSSION

     Chronic hepatitis C leading to end-stage liver disease and one of the most important indications for liver transplantation (Lin et al., 2014). Liver lesions in the hepatitis C virus is attributed to the interaction of several pathways in the host immune response, disruption of pathogen associated pattern and interference with cellular immune-regulation and subversion of natural killer (NK) cell activity (Kaplan, 2015).

     In our study, we found a strong relationship between the number of activated HSCs and liver fibrosis in chronic HCV group which was in agreement with most studies that demonstrate the role of HSCs in the development of liver fibrosis (Săndulescu et al., 2011, Ionescu et al., 2013 and Bakiera et al., 2016).

     In the present study, significant numbers of α-SMA positive HSCs were detected in patients with chronic HCV infection group, but in normal donors group α-SMA positive HSCs were not detected, this finding was in agreement with Zakaria et al. (2010) and Hirabaru et al. (2014) whose postulated that the activity of HSCs stimulates the expression of tissue inhibitors of matrix metalloproteinase (TIMP-1 & TIMP-2). This effect leads to the alteration in the balance between the production and degradation of ECM components and its accumulation in the hepatic tissue (Ionescu A.G. et al., 2013). We found in our study that α-SMA showed a well-known and reliable mesenchymal marker of HSCs activation this was in correspondence to Olsen et al. (2011).

     In the present study, GFAP-positive HSCs in post-transplant HCV recurrence positively correlated with fibrosis progression. Moreover, GFAP immunostaining seemed to identify HSCs detectable in liver fibrosis mainly in the early stages compared to α-SMA in fact, the closer to transplantation recurrence was observed, the more diffusely GFAP expression was found this was in agreement with Busletta et al. (2011).

     Our results for GFAP expression identified resting hepatic stellate cells and there was a high expression in the post transplanted liver not received treatment indicating recurrence of hepatitis C, while in the chronic injury low expression was detected. This result was in agreement with Hassan et al. (2014). Our observation of strong association of GFAP with the gold standard immunohistochemical marker α-SMA suggested that GFAP could be a useful indicator of early HSCs activation in post transplanted liver patients.

     HSCs, portal fibroblast, and myofibroblasts have been reported to comprise > 90% of the collagen expressing cells suggesting that they were the major source of collagen expressing cells in the fibrotic liver. HSCs are generally accepted as major contributors to liver fibrosis that give rise to hepatic myofibroblasts in response to liver injury (Iwaisako et al., 2014).

     In our study, the preportal fibrosis associated with chronic hepatitis C increase number of HSCs as fibrous tissue increased compared with the typical HSCs observed in normal liver. The cells encountered in this pathological condition have fewer lipid droplets and an elongated indented nucleus. This was in agreement with Nafady et al. (2017) who have postulated that activation of fat storing cells evidenced by transforming to fibroblast-like cell producing extraordinary amounts of collagen fibers in chronic stages. These fibers were seen around them in space of Disse, and in between the hepatic cells.

     In our study, liver transplanted patients without hepatitis C treatment showed an increased amount of activated HSCs compared to patients with liver transplanted patients with hepatitis C treatment, and normal livers. These data strongly suggest a role for HSCs in the pathogenesis of graft fibrosis and graft loss in hepatitis C recurrence this observation was in agreement with Terrault (2012). The most effective means of preventing HCV recurrence is eradicating HCV prior to liver transplantion.

     The newer direct antiviral agents (DAAs) for patients with HCV infection are highly effective, resulting in sustained virological response (SVR) in greater than 90% of patients (Van der Meer et al., 2012). Our study demonstrated that treatment of post-transplanted patients with antihepatitis C treatment played an important role in the prevention of recurrent HCV infection this was in accordance with Curry et al. (2015) whose postulated that pre-transplantation treatment of HCV prevents HCV recurrence. Another study by Ferrarese et al. (2018) supported our results who have suggested that Hepatitis C virus-related cirrhosis decreased since the introduction of direct-acting antivirals agents.

LIMITATIONS

     Limited availability of post transplanted hepatic tissue for TEM because assessment of graft fibrosis in patients with recurrent hepatitis C after liver transplantation‏ was done now by transient elastography and it's considered now good alternative to liver biopsy in post-liver transplantation.

CONCLUSION

     The hepatic stellate cells play an important role in the fibrosis of liver damaged by the hepatitis C virus and in the post-transplanted liver which was not treated from HCV. So, antiviral treatment led to improvement cases of post-transplantion.

REFERENCES

  1. Bakiera KK, Gawda EW, Kwiatkowska BC and Jedrych BJ. (2016): Hepatic stellate cells activation and liver fibrosis after chronic administration of ethanol. Curr. Issues Pharm. Med. Sci; 29(2): 66-70.
  2. Barakat EM, El Wakeel LM and  Hagag RS. (2013): Effects of Nigella sativa on outcome of hepatitis C in Egypt. World J Gastroenterol; 19(16): 2529–2536.
  3. Bhatia HK, Singh H, Grewal N and Natt NK. (2014): Sofosbuvir A novel treatment option for chronic hepatitis C infection. Molecules of the Millennium; 5(4): 278-284.
  4. Busletta C, Novo E, Cannito S, Paternostro C and Parola M. (2011): Hepatic myofibroblasts in liver fibrogenesis. Trends in Alcoholic Liver Disease Research - Clinical and Scientific Aspects.
  5. Curry MP, Forns X, Chung RT, Terrault NA, Brown R and Fenkel JM. (2015): Sofosbuvir and ribavirin prevent recurrence of HCV infection after liver transplantation: an open-label study. Gastroenterology; 148:100 –107.
  6. DeLemos AS, Schmeltzer PA and Russo MW. (2014): Recurrent hepatitis C after liver transplant. World J Gastroenterol; 20(31):10668–10681.
  7. Ferrarese A, Germani G, Gambato M, Russo FP, Senzolo M, Zanetto A, Shalaby S, Cillo U, Zanus G, Angeli P and Patrizia Burra P. (2018): Hepatitis C virus related cirrhosis decreased as indication to liver transplantation since the introduction of direct-acting antivirals: A single-center study. World J Gastroenterol24 (38): 4403–4411.
  8. Friedman SL and Lee UE. (2011): Mechanisms of Hepatic Fibrogenesis. Best Pract Res Clin Gastroenterol; 25(2): 195–206.
  9. Hassan S, Syed S, and Imdad Kehar S. (2014): Glial Fibrillary Acidic Protein (GFAP) as a Mesenchymal marker of Early Hepatic Stellate Cells Activation in Liver Fibrosis in Chronic Hepatitis C Infection. Pak J Med Sci; 30(5): 1027-1032.
  10. Hirabaru M, Mochizuki K, Takatsuki M, Soyama A, Kosaka T, Kuroki T, Shimokawa I and Eguchi S. (2014): Expression of alpha smooth muscle actin in living donor liver transplant recipients. World J Gastroenterol; 14: 20(22): 7067–7074.
  11. Ionescu AG, Letitia AMS, Vere CC, Ciurea ME, Streba CT, Ionescu M, Comanescu M , Irimia E and Rogoveanu O. (2013): Histopathological and immunohistochemical study of hepatic stellate cells in patients with viral C chronic liver disease. Rom J Morphol Embryol; 54(4):983–991.
  12. Iwaisako K, Jiang C, Zhang M, Cong M, Moore-Morris TJ, Park TJ, Liu X, Xu J, Wang P, Paik YH, Meng F, Asagiri M, Murray LA, Hofmann AF, Iida T, Glass CK, Brenner DA and Kisseleva T. (2014): Origin of myofibroblasts in the fibrotic liver in mice. Proc Natl Acad Sci U S A; 111:3297-3305
  13. Juan L , Ying-Ren Z and Zhen T. (2019): Roles of hepatic stellate cells in acute liver failure: From the perspective of inflammation and fibrosis. World J Hepatol; 11(5): 412–420.
  14. Kaplan DE. (2015): Immunopathogenesis of Hepatitis C Virus Infection. Gastroenterol Clin; 44: 735–760.
  15. Kasztelan-Szczerbińska B, Komórkigwia-ździstejakogłówny and sygnalizac-jimiędzykomórkowej. (2010): Stellate cells as a central regulator of intracellular signaling in the process of liver fibrosis. PostępyNaukMedycznych; 1: 69-74.
  16. Kisseleva T, Cong M, Paik Y, Scholten D, Jiang C and Benner C. (2012): Myofibroblasts revert to an inactive phenotype during regression of liver fibrosis. Proc Natl Acad Sci U S A; 109: 9448–9453.
  17. Lin Ji, Jian-Feng Wu, Zhang Qi, Hong-Wei Zhang, and Guang-Wen Cao. (2014): Virus-related liver cirrhosis: Molecular basis and therapeutic options. World J Gastroenterol; 20 (21): 6457–6469.
  18. Nafady AM, Ahmed OB, Ghafeer HH. (2017): Scanning and transmission electron microscopy of the cells forming the hepatic sinusoidal wall of rat in acetaminophen- and Escherichia coli endotoxin-induced hepatotoxicity. Journal of Microscopy and Ultrastructure; 5 (1): 21-27.
  19. Olsen AL, Bloomer SA, Chan EP, Gaça MD, Georges PC and Sackey B. (2011): Hepatic stellate cells require a stiff environment for myofibroblastic differentiation. Am J Physiol Gastrointest Liver Physiol; 301(1):110–118.
  20. Pellicoro A, Ramachandran P and Iredale JP. (2014): Liver fibrosis and repair: immune regulation of wound healing in a solid organ. Nat Rev Immunol; 14:181–194.
  21. Salas-Villalobos TB. Lozano-Sepúlveda SA. Rincón-Sánchez AR. Govea-Salas MG and Rivas-Estilla AM. (2017): Mechanisms involved in liver damage resolution after hepatitis C virus clearance. Medicina Universitaria; 19(75): 100-107.
  22. Săndulescu L, Rogoveanu I, Ciurea T, Comănescu MV, Streba CT, Ionescu AG, Oproaica A and Ene M. (2011): Immunohistochemical study of stellate cells in patients with chronic viral hepatitis C genotype 1. Rom J Morphol Embryol; 52(1):137-43.
  23. Terrault N. (2012): Liver transplantation in the setting of chronic HCV. Best Pract Res Clin Gastroenterol ., 26:531–548.
  24. Troeger JS., Mederacke I, Gwak GY, Dapito DH, Mu X and Hsu CC. (2012): Deactivation of hepatic stellate cells during liver fibrosis resolution in mice. Gastroenterology; 143: e1022.
  25. Zakaria S, Youssef M, Moussa M, Akl M, El-Ahwany E, El-Raziky M, Mostafa O, Helmy A.H, and El-Hindawi A. (2010): Value of α-smooth muscle actin and glial fibrillary acidic protein in predicting early hepatic fibrosis in chronic hepatitis C virus infection. Arch Med Sci; 30: 6(3): 356–365.

 

دراسة هستولوجیة وهستوکیمیائیة مناعیة للخلایا النجمیة الکبدیة قبل وبعد زراعة الکبد البشرى

محمود احمد عبد الحلیم مسعود، محمد أحمد  قاسم، أیمن فتحى الشعراوى، عمرو فرج السباعى*، تامر ابراهیم عبد الرحمن احمد

قسم الهستولوجیا وبیولوجیا الخلیة بطب الازهر، وقسم الباثولوجى بالاکادیمیة الطبیة العسکریة*

خلفیة البحث: الخلایا النجمیة الکبدیة هى التى تخزن الدهون وتنظم التمثیل الغذائى لفیتامین (أ) وتلعب دور هام اثناء النشاط المناعى عند حدوث اى ضررلخلایا للکبد, مما یؤدى الى زیادة فى نشاطها ونقص فى وظائفها الفسیولوجیة وتتحول الى خلایا تشبه الخلایا اللیفیة العضلیة التى تبدأ فى تخلیق الانسجة اللیفیة مما یؤدى الى تلیف الکبد.

الهدف من البحث: الکشف عن العلاقة بین نشاط الخلایا النجمیة الکبدیة ودرجة تلیف الکبد فى المرضى المصابین بفیروس الکبد الوبائى سی و بعد عملیة زراعة الکبد.

مواد وطرق البحث: اشتمل البحث على اربعة وثلاثین حالة تم اخذ عینات الکبد منهم من المرکز الطبى العالمى تم تقسیمهم الى ثلاثة مجموعات:

         المجموعة الاولى عشرة عینات کبدیة لأشخاص طبیعین من المتبرعین بالکبد ولا یعانون من أى امراض، والمجموعة الثانیة اثنتا عشرة عینة کبدیة لاشخاص یعانون من التهاب الکبدى الوبائى سی وتلیف فى الکبد والمجموعة الثالثة اثنتا عشرة عینة کبدیة ممن خضعوا لعملیة زراعة الکبد، وقد تم تقسیمهم الى مجموعتین فرعیتین مجموعة اخذت علاج الکبد الوبائى سی ومجموعة اخرى لم تاخذ العلاج.

         وقد تم أخذ العینات من اکباد الاشخاص الذین أجرى علیهم البحث لتحضیرها وصباغتها بواسطة الهیماتوکسیلین والأیوسین للکشف الظاهرى على انسجة الکبد وخلایاه وصبغة الماسون ترایکوروم للکشف على التلیف داخل انسجة الکبد. کما تم صبغ عینات أخرى بواسطة الصبغات المناعیة الخاصة بها وهى (الاکتین العضلى الاملس الفا والبروتین الحمضى اللیفى) للکشف على الخلایا النجمیة الکبدیة. کما تم تحضیر عینات اخرى للفحص بالمیکروسکوب الالیکترونى للکشف على البنیة الدقیقة للخلایا الکبدیة. تم فحص الشرائح وتصویرها وعمل الاحصائیات الحسابیة اللازمة لها.

نتائج البحث: أظهرت صبغة الهیماتوکسلین والأیوسین فى المجموعه الأولى بناء نسیجى طبیعى للکبد، أما فى المجموعه الثانیة فقد لوحظ تغیرأ وخللا فى شکل وترکیب خلایا الکبد، بینما فى المجموعة الثالثة لوحظ التهابات بسیطة إلى متوسطة بالقناة البابیة فى الاشخاص الذین لم یاخذوا علاج الکبد الوبائى سی بینما ألاشخاص الذین اخذوا العلاج کانت خلایا الکبد طبیعیة کما أظهرت صبغة الماسون ترایکوروم عن وجود اکبر نسبة تلیف فى المرضى المصابین بتلیف الکبدى الوبائى سی وذلک بالمقارنة بالاشخاص الذین قاموا بزراعة الکبد ولم یاخذوا علاج الکبدى الوبائى سی او الاشخاص الطبیعین.

         وقد أظهر التحلیل الاحصائى المناعى لعینات الکبد زیادة فى مستوى المناطق الموجبة للصبغة المناعیة (الاکتین العضلى الاملس الفا) للمجموعة المصابة بفیروس إلتهاب الکبدى الوبائى سی ویعانون من تلیف فى الکبد عن المجموعة التى قامت بزراعة الکبد ولم یاخذوا علاج الالتهاب الکبدى الوبائى سی بینما عینات الأشخاص الطبیعین لم یسجلوا اى مناطق موجبة للصبغة. وکذلک أظهر التحلیل المناعى لعینات الکبد (للبروتین الحمضى اللیفى) زیادة فى مستوى المناطق الموجبة للصبغة المناعیة للمجموعة الثانیة التى قامت بزراعة الکبد وکذلک لبعض الاشخاص الطبیعین من المجموعة الاولى بینما انخفضت تدریجیا فى المجموعة المصابة بالتهاب الکبدى الوبائى سی مع وجود تلیفات وتشمع بالکبد مما یؤکد ان هذه الصبغة المناعیة الخاصة تعتبر مؤشر جید للکشف الاولى عن نشاط الخلایا النجمیة الکبدیة.

         وقد لوحظ بالمیکروسکوب ألالیکترونى أن الخلایا النجمیة الکبدیة یحدث بها زیادة فى الأنسجة اللیفیة وانخفاض بنسبة الدهون الموجودة بها مع زیادة نشاطها فى الکبد المصاب بفیروس سی عن الکبد السلیم.

الاستنتاج: الخلایا النجمیة الکبدیة لها دور محورى واساسى فى عملیة تلیف الکبد وذلک بسبب التغیرات التى تحدث بها وزیادة نشاطها کما انها تلعب دور مهم فى التغیرات التى تحدث بعد عملیة زراعة الکبد وعلاج فیروس الکبدى الوبائى سی قبل عملیة زراعة الکبد یؤدى إلى قلة نشاط الخلایا النجمیة الکبدیة وتجنب تخلیق نسیج لیفى فى الکبد مما یؤدى الى زیادة عمر الکبد المزروع.

  1. REFERENCES

    1. Bakiera KK, Gawda EW, Kwiatkowska BC and Jedrych BJ. (2016): Hepatic stellate cells activation and liver fibrosis after chronic administration of ethanol. Curr. Issues Pharm. Med. Sci; 29(2): 66-70.
    2. Barakat EM, El Wakeel LM and  Hagag RS. (2013): Effects of Nigella sativa on outcome of hepatitis C in Egypt. World J Gastroenterol; 19(16): 2529–2536.
    3. Bhatia HK, Singh H, Grewal N and Natt NK. (2014): Sofosbuvir A novel treatment option for chronic hepatitis C infection. Molecules of the Millennium; 5(4): 278-284.
    4. Busletta C, Novo E, Cannito S, Paternostro C and Parola M. (2011): Hepatic myofibroblasts in liver fibrogenesis. Trends in Alcoholic Liver Disease Research - Clinical and Scientific Aspects.
    5. Curry MP, Forns X, Chung RT, Terrault NA, Brown R and Fenkel JM. (2015): Sofosbuvir and ribavirin prevent recurrence of HCV infection after liver transplantation: an open-label study. Gastroenterology; 148:100 –107.
    6. DeLemos AS, Schmeltzer PA and Russo MW. (2014): Recurrent hepatitis C after liver transplant. World J Gastroenterol; 20(31):10668–10681.
    7. Ferrarese A, Germani G, Gambato M, Russo FP, Senzolo M, Zanetto A, Shalaby S, Cillo U, Zanus G, Angeli P and Patrizia Burra P. (2018): Hepatitis C virus related cirrhosis decreased as indication to liver transplantation since the introduction of direct-acting antivirals: A single-center study. World J Gastroenterol24 (38): 4403–4411.
    8. Friedman SL and Lee UE. (2011): Mechanisms of Hepatic Fibrogenesis. Best Pract Res Clin Gastroenterol; 25(2): 195–206.
    9. Hassan S, Syed S, and Imdad Kehar S. (2014): Glial Fibrillary Acidic Protein (GFAP) as a Mesenchymal marker of Early Hepatic Stellate Cells Activation in Liver Fibrosis in Chronic Hepatitis C Infection. Pak J Med Sci; 30(5): 1027-1032.
    10. Hirabaru M, Mochizuki K, Takatsuki M, Soyama A, Kosaka T, Kuroki T, Shimokawa I and Eguchi S. (2014): Expression of alpha smooth muscle actin in living donor liver transplant recipients. World J Gastroenterol; 14: 20(22): 7067–7074.
    11. Ionescu AG, Letitia AMS, Vere CC, Ciurea ME, Streba CT, Ionescu M, Comanescu M , Irimia E and Rogoveanu O. (2013): Histopathological and immunohistochemical study of hepatic stellate cells in patients with viral C chronic liver disease. Rom J Morphol Embryol; 54(4):983–991.
    12. Iwaisako K, Jiang C, Zhang M, Cong M, Moore-Morris TJ, Park TJ, Liu X, Xu J, Wang P, Paik YH, Meng F, Asagiri M, Murray LA, Hofmann AF, Iida T, Glass CK, Brenner DA and Kisseleva T. (2014): Origin of myofibroblasts in the fibrotic liver in mice. Proc Natl Acad Sci U S A; 111:3297-3305
    13. Juan L , Ying-Ren Z and Zhen T. (2019): Roles of hepatic stellate cells in acute liver failure: From the perspective of inflammation and fibrosis. World J Hepatol; 11(5): 412–420.
    14. Kaplan DE. (2015): Immunopathogenesis of Hepatitis C Virus Infection. Gastroenterol Clin; 44: 735–760.
    15. Kasztelan-Szczerbińska B, Komórkigwia-ździstejakogłówny and sygnalizac-jimiędzykomórkowej. (2010): Stellate cells as a central regulator of intracellular signaling in the process of liver fibrosis. PostępyNaukMedycznych; 1: 69-74.
    16. Kisseleva T, Cong M, Paik Y, Scholten D, Jiang C and Benner C. (2012): Myofibroblasts revert to an inactive phenotype during regression of liver fibrosis. Proc Natl Acad Sci U S A; 109: 9448–9453.
    17. Lin Ji, Jian-Feng Wu, Zhang Qi, Hong-Wei Zhang, and Guang-Wen Cao. (2014): Virus-related liver cirrhosis: Molecular basis and therapeutic options. World J Gastroenterol; 20 (21): 6457–6469.
    18. Nafady AM, Ahmed OB, Ghafeer HH. (2017): Scanning and transmission electron microscopy of the cells forming the hepatic sinusoidal wall of rat in acetaminophen- and Escherichia coli endotoxin-induced hepatotoxicity. Journal of Microscopy and Ultrastructure; 5 (1): 21-27.
    19. Olsen AL, Bloomer SA, Chan EP, Gaça MD, Georges PC and Sackey B. (2011): Hepatic stellate cells require a stiff environment for myofibroblastic differentiation. Am J Physiol Gastrointest Liver Physiol; 301(1):110–118.
    20. Pellicoro A, Ramachandran P and Iredale JP. (2014): Liver fibrosis and repair: immune regulation of wound healing in a solid organ. Nat Rev Immunol; 14:181–194.
    21. Salas-Villalobos TB. Lozano-Sepúlveda SA. Rincón-Sánchez AR. Govea-Salas MG and Rivas-Estilla AM. (2017): Mechanisms involved in liver damage resolution after hepatitis C virus clearance. Medicina Universitaria; 19(75): 100-107.
    22. Săndulescu L, Rogoveanu I, Ciurea T, Comănescu MV, Streba CT, Ionescu AG, Oproaica A and Ene M. (2011): Immunohistochemical study of stellate cells in patients with chronic viral hepatitis C genotype 1. Rom J Morphol Embryol; 52(1):137-43.
    23. Terrault N. (2012): Liver transplantation in the setting of chronic HCV. Best Pract Res Clin Gastroenterol ., 26:531–548.
    24. Troeger JS., Mederacke I, Gwak GY, Dapito DH, Mu X and Hsu CC. (2012): Deactivation of hepatic stellate cells during liver fibrosis resolution in mice. Gastroenterology; 143: e1022.
    25. Zakaria S, Youssef M, Moussa M, Akl M, El-Ahwany E, El-Raziky M, Mostafa O, Helmy A.H, and El-Hindawi A. (2010): Value of α-smooth muscle actin and glial fibrillary acidic protein in predicting early hepatic fibrosis in chronic hepatitis C virus infection. Arch Med Sci; 30: 6(3): 356–365.