• 검색 결과가 없습니다.

Gene expression associated with fibrosis

3. In clinical syudy

3.2. Secondary outcome

3.2.2. Gene expression associated with fibrosis

After stem cell administration, the relative expressions of transforming growth factor β1 (TGF-1), type 1 collagen (collagen-1), and α-smooth muscle actin (α-SMA) in real-time PCR significantly decreased from 1 ± 0.36 to 0.69 ± 0.32 (P<0.01), from 1 ± 0.58 to 0.70 ± 0.62 (P<0.01), and from 1 ± 0.44 to 0.55 ± 0.37 (P<0.01), respectively (Fig. 15).

69 A B

C

Figure 15. Gene expression analysis associated with fibrosis.

After administration of BM-MSCs therapy, the relative expressions of (A) TGF-1, (B) collagen-1, and (C) α-SMA by real-time PCR decreased significantly from 1 ± 0.36 to 0.69 ± 0.32 (P<0.01), from 1 ± 0.58 to 0.70 ± 0.62 (P<0.01), and from 1 ± 0.44 to 0.55 ± 0.37 (P<0.01), respectively. Values are expressed as means ± standard deviation. *P<0.01.

70

3.3. Safety

All 11 patients tolerated the therapy. They were monitored regularly for any sign of possible side effects of the stem cell therapy such as fever, hypersensitivity reaction, and acute rejection fever. There was no evidence of stem cell therapy related tumor developing during the follow-up period.

71

IV. Discussion

At the overt cirrhotic stage, end-stage liver disease often necessitates liver transplantation 35. However, it has several limitations, such as shortage of organ donors, high medical cost, a widening donor recipient gap, and life long dependence on immunological suppression. Hence, stem cell transplantation has been suggested as an effective alternate therapy for hepatic disease 10, 12, 13

.

Stem cell therapies have shown promising benefits for hepatic fibrosis in experimental and clinical studies 13, 14, 19, 22

. BM comprises two main populations of stem cells, hematopoietic stem cells and MSCs, of which the latter have been considered as alternative cell sources for liver or hepatocyte transplantation 24. In liver damage, MSCs are able to differentiate into hepatocytes, stimulate the regeneration of endogenous parenchymal cells, migrate to damaged sites, and enhance fibrous matrix degradation (antifibrotic effects). Furthermore, several clinical studies have demonstrated favorable effects of BM-MSCs treatment such as improving the liver function in patients with hepatitis B or C virus-related cirrhosis 22, 36. However, no previous study has examined the effect of autologous BM-MCs on hepatic fibrosis in patients with alcoholic cirrhosis. We aimed to determine the safety and antifibrosis effect of MSCs on alcohol-related hepatic fibrosis in pre-clinical and clinical study.

In pre-clinical studies, we investigated whether BM-MSCs can attenuate hepatic fibrosis in a thioacetamide induced cirrhotic rat model. Thioacetamide (TAA)-induced cirrhosis is well-known classical experimental cirrhosis model. In present study, TAA administration results in deficiencies in liver functions and a progress

72

increase in collagen accumulation in the liver with periportal cirrhotic characterized by portal-portal fibrous septa surrounding the hepatic lobules. These results were in agreement with 37 who indicated that TAA-induced cirrhosis characterized by cellular necrosis, fibrosis, micronodular reorganization of the parenchyma, duct proliferation, transaminase increase, peroxidation of lipid membranes, and macronodular cirrhosis

38, 39

.

Previous studies have suggested that MSC have antifibrotic effects on injured liver in animal models of liver fibrosis 40. Consistent with their result, we found that BM-MSCs treated group showed histologically significant improvement of hepatic fibrosis compared to untreated cirrhotic group. These results were further confirmed by immunohistochemical staining revealing α-SMA expression and Picrosirius Red staining. Additionally, the percentage of collagen proportionate area and content of hepatic hydroxyproline significantly decreased after BM-MSCs treatment. There results suggest that BM-MSCs improved liver structure in TAA-induced cirrhotic rats.

In present study, we investigated the effect of BM-MSCs on TGF-1, collagen-1,and α-SMA in TAA-induced cirrhotic rats. TGF-1 is a mediator particularly in liver fibrogenesis 41. TGF-1 promotes HSC to transit into MFB, stimulates the synthesis of ECM such as type I collagen and inhibits its degradation 42. And the expression of α-SMA in the liver is an indicator of HSC activation, which is recognized as the key player in hepatic fibrosis and cirrhosis 43. Likewise, this study showed that BM-MSCs recovered liver function, as indicated by decreasing gene expression of TGF-1, collagen-1, and α-SMA. Furthermore, the protein expression of α-SMA significantly decreased after BM-MSCs treatment. The results of our study are in agreement with

73

those of Campbell JS et al 44. The finding a reduction in HSC activation during regeneration of the fibrotic liver in the rat after BM-MSCs treatment.

The signaling pathway of TGF-β1 was activated and catalyse Smad2/Smad3 phosphorylation, entering into the nucleus, binding to transcriptional factors and regulating the expression of target genes (e.g.,collagen, tissue inhibitor of matrix metallproteinases). Indeed, activation of TGF-β/Smad signaling is a key mechanism of liver fibrosis in both experimental and human chronic liver diseases 45–47. Similarly, the protein level of TGF-β1, p-Smad2, and p-Smad3 in the untreated cirrhotic group were increased compared to the sham, indicating that the TGF-β1/Smad signaling pathway was activated during the hepatic fibrosis. BM-MSCs in treated group significantly reduced the protein level of TGF-β1, p-Smad2, and p-Smad3 consistent to its inhibitory effect on hepatic fibrosis. There results suggest that main antifibrotic process by BM-MSCs treatment could be related to the inhibition of TGF-β/Smad signaling pathway.

In vitro study, we investigated the effects of paracrine factors secreted by

BM-MSCs on activated HSCs, the primary extracellular matrix-producing cell type in the liver. Bone marrow derived MSCs have the ability to repair damaged liver. In vivo activation of HSCs is divided into a fibrogenic and hyperplastic response 48 that is mediated by many autocrine and paracrine signals. And strategies targeting removal of activated HSCs have shown to reduce fibrosis and augment liver function 49. In the liver, IL-10 is produced by several types of cells including hepatocytes, sinusoidal cells, kupffer cells, stellate cells and liver-associated lymphocytes 50. It was reported that endogenous IL-10 in the liver increased for downregulating inflammatory

74

response and improving liver fibrosis 51. IL-10 is a potent anti-inflammatory cytokine, which can protect hepatocytes, promote ECM degradation and modulate hepatic fibrogenesis by downregulating the inflammatory response, inhibiting the synthesis of pro-inflammatory cytokines and activating nuclear factor kB 52, 53.

HGF was identified as a mitogen for mature hepatocytes 54, and is considered to be important for the development and regeneration of liver 55. HGF showed the anti-apoptotic activity in hepatocytes and suppressed liver fibrosis 56, 57.

Previous studies have shown that the underlying mechanisms in the modulation of HSC activity by MSC were attributed to paracrine mediators, IL-10, TNF-α and HGF.

Blockade of MSC-derived IL-10 and TNF-α abolished the inhibitory effects of MSC on HSC proliferation and collagen synthesis; MSC-derived HGF was responsible for the marked induction of HSC apoptosis as determined by antibody neutralization studies. IL-6 secretion from activated HSCs induced IL-10 secretion from MSC, suggesting a dynamic response of MSC to HSCs in the microenvironment 25, 58. Consistent with their result, our data found that indirect coculture of activated HSCs with MSCs decreased the production of TGF-1 (24.7%) and IL-6 (8.4%).

Conversely, coculture of activated HSCs with MSCs increased the production of HGF (33.5%) and IL-10 (31.6%). HSC viability was decreased by 66% and apoptosis was increased by 43% with direct coculture of activated HSCs with MSCs. These results suggest that BM-MSCs inhibit the proliferative and fibrogenic function of activated HSCs in a paracrine manner and as a function of BM-MSC number. This inhibition could be caused by BM-MSC derived IL-10 which acted synergistically. The secretion of IL-10 by MSCs was found to be a dynamic response to IL-6 secretion by

75

activated HSCs. Furthermore, BM-MSCs induced apoptosis in activated HSCs that is, in part, mediated by HGF. These results support the hypothesis that the therapeutic effect of BM-MSCs may be due to paracrine factors that modulate the proliferation, viability, and function of resident HSCs. Immunomodulation of HSCs by BM-MSC soluble factors provides the mechanistic evidence that BM-MSCs can exert a protective role through paracrine signaling to liver HSCs.

Our pre-clinical study has confirmed the effects of BM-MSCs on hepatic fibrosis, which probably are related with the inhibition of hepatic HSC activation and TGF-β1/Smad signaling pathways.

Furthermore, we investigated the antifibrotic effect of BM-MSCs on alcoholic cirrhosis as a phase II in clinical study. In clinical study, after autologous BM-MSCs injection, histological improvements were observed in 6 of 11 patients (54.5%). The Child-Pugh score improved in ten patients (90.9%), and the expressions of TGF-1, collagen-1 and α-SMA were significantly decreased (p<0.05). Importantly, no significant complications or side effects were observed during this study. These results indicate that BM-MSCs therapy has a potential as an antifibrotic treatment in cirrhosis and a bridging therapy for liver transplantation in advanced cirrhosis with hepatic insufficiency. Current antiviral therapies can improve the hepatic fibrosis in patients with B or C virus-related cirrhosis, but the only possible treatment for alcoholic cirrhosis is alcohol abstinence. Therefore, our results suggest a novel strategy for the treatment of alcoholic cirrhosis.

There is a clear histological variability of severity within cirrhosis classified as F4 by the METAVIR system. Cirrhosis is currently considered to be potentially

76

reversible if the cause of the injury is removed. The lack of subclassification within cirrhosis can be problematic when assessing the antifibrotic effect of agents such as antiviral drugs. For instance, even though antifibrotic therapy leads to improvement of hepatic fibrosis from F4C to F4A in the Laennec system, the lack of change under the conventional METAVIR system will lead to the false conclusion that treatment is ineffective. Hence, further histological subclassification of cirrhosis is required 4, 30. In the present study, we applied the new Laennec fibrosis scoring system to provide a more detailed classification of F4 cirrhosis.

The quality of interventional studies of alcoholic liver disease patients is crucially dependent on monitoring the alcohol intake 59, 60. In the present study we did our best to monitor the subjects’ alcohol reintake and alcohol abstinence, using serologic liver function tests including serum AST/ALT ratio and γ-glutamyl transpeptidase, and mean corpus volume in complete blood counts. Furthermore, alcohol intake was monitored every week by phone calls and in monthly face to face interviews with patients and their family members. This was effective in preventing alcohol reintake in most of the participants due to the formation of a good rapport.

The present study was subject to some limitations. First, this pilot study was designed and performed as an open-label single arm study without control. Indeed, lack of a control group is certainly a major limitation of the present study. However, repeating biopsies in control group would be ethically difficult if no active treatment is being offered. Nevertheless, future prospective controlled studies would be necessary to validate the beneficial effect of MSCs on hepatic fibrosis shown in the present study. Although being an open-label study, the pathologist was blinded to the

77

clinical data and the kind of therapy used, which enabled us to be confident about the accuracy of our histological data.

Second, like all biopsy studies there were potential limitations regarding liver biopsy sampling, and in particular sampling error. In the present study, our criteria of excluding biopsy specimens smaller than 15 mm should have minimized the magnitude of any sampling error. In addition, we incorporated other methods to assess hepatic fibrosis in this study to improve the validity of our morphometric analysis and measurement of TGF-1, collagen-1, and α-SMA by quantitative real-time PCR of the biopsied liver tissues 31.

This is the first study to determine the effect of BM-MSCs on hepatic fibrosis in patients with AC. The obtained results support the approval of this class of agents as a therapy for hepatic fibrosis.

78

Ⅴ. CONCLUSION

This study has confirmed the significant inhibitory effect of bone marrow-derived mesenchymal stem cells on TAA-induced hepatic fibrosis in rats, which probably correlates with the inhibition of hepatic HSC activation and TGF-β1/Smad sign aling pathways. Furthermore, BM-MSCs therapy in alcoholic cirrhosis induces a histological and quantitative improvement of hepatic fibrosis. Therefore, the obtained results support the approval of this class of agents as a therapy for hepatic fibrosis.

In conclusion, BM-MSC therapy leads to the improvement of hepatic fibrosis, and so may provide a new strategy for antifibrosis therapy in near future.

79

REFERENCE

1. Bolondi L, Gramantieri L. . From liver cirrhosis to HCC. Intern Emerg Med 2011; 6 Suppl 1: 93-8.

2. Iredale JP.Cirrhosis: new research provides a basis for rational and targeted treatments. BMJ 2003; 327(7407): 143-7.

3. Kim MY, Baik SK, Yea CJ, et al. Hepatic venous pressure gradient can predict the development of hepatocellular carcinoma and hyponatremia in decompensated alcoholic cirrhosis. Eur J Gastroenterol Hepatol 2009;

21(11): 1241-6.

4. Kim MY, Suk KT, Baik SK, et al. Hepatic vein arrival time as assessed by contrast-enhanced ultrasonography is useful for the assessment of portal hypertension in compensated cirrhosis. Hepatology 2012; 56(3):1053-62.

5. Lee SS, Shin HS, Kim HJ, et al. Analysis of prognostic factors and 5-year survival rate in patients with hepatocellular carcinoma: a single-center experience. Korean J Hepatol 2012; 18(1): 48-55.

6. Koichi Matsuzaki. Smad phosphoisoform signals in acute and chronic liver injury: similarities and differences between epithelial and mesenchymal cells.Cell Tissue Res 2012; 347:225–243.

80

Current status and future development. World J Hepatol 2011; 3(8): 215-8.

10. Strom SC, Bruzzone P, Cai H,et al. Hepatocyte transplantation: clinical experience and potential for future use. Cell Transplant 2006; 15 Suppl 1:

S105-10.

11. Strom S, Fisher R. Hepatocyte transplantation: new possibilities for therapy.

Gastroenterology 2003; 124(2): 568-71.

12. Kakinuma S, Nakauchi H, Watanabe M. Hepatic stem/progenitor cells and stem-cell transplantation for the treatment of liver disease. J Gastroenterol 2009; 44(3): 167-72.

13. Kharaziha P, Hellström PM, Noorinayer B, et al. Improvement of liver function in liver cirrhosis patients after autologous mesenchymal stem cell injection: a phase I-II clinical trial. Eur J Gastroenterol Hepatol 2009;

21(10): 1199-205.

14. Cho KA, Lim GW, Joo SY, et al. Transplantation of bone marrow cells reduces CCl4 -induced liver fibrosis in mice. Liver Int 2011; 31(7): 932-9.

81

15. Sakaida I, Terai S, Yamamoto N, et al. Transplantation of bone marrow cells reduces CCl4-induced liver fibrosis in mice. Hepatology 2004; 40(6): 1304-11.

16. Hardjo M, Miyazaki M, Sakaguchi M, et al. Suppression of carbon tetrachloride-induced liver fibrosis by transplantation of a clonal mesenchymal stem cell line derived from rat bone marrow. Cell transplant 2009; 18(1): 89-99.

17. Higashiyama R, Inagaki Y, Hong YY, et al. Bone marrow-derived cells express matrix metalloproteinases and contribute to regression of liver fibrosis in mice. Hepatology 2007; 45(1): 213-22.

18. Mormone E, George J, Nieto N. Molecular pathogenesis of hepatic fibrosis and current therapeutic approaches. Chem Biol Interac. 2011; 193(3): 225-31.

19. Pai M, Zacharoulis D, Milicevic MN, et al. Autologous infusion of expanded mobilized adult bone marrow-derived CD34+ cells into patients with alcoholic liver cirrhosis. Am J Gastroenterol 2008; 103(8): 1952-8.

20. Rockey D C. Current and future anti-fibrotic therapies for chronic liver disease. Clin Liver Dis 2008; 12(4):939-62.

21. Sakaida I, Terai S, Nishina H, et al. Development of cell therapy using autologous bone marrow cells for liver cirrhosis. Med Mol Morphol 2005:

38(4):197-202.

82

22. Terai S, Ishikawa T, Omori K, et al. Improved liver function in patients with liver cirrhosis after autologous bone marrow cell infusion therapy. Stem Cells 2006; 24(10): 2292-98.

23. am Esch JS 2nd, Knoefel WT, Klein M, et al. Portal application of autologous CD133+ bone marrow cells to the liver: a novel concept to support hepatic regeneration. Stem Cells 2005; 23(4): 463-70.

24. van Poll D, Parekkadan B, Cho CH, et al. Mesenchymal stem cell-derived molecules directly modulate hepatocellular death and regeneration in vitro and in vivo. Hepatology 2008; 47(5):1634-43.

25. Parekkadan B, van Poll D, Megeed Z Immunomodulation of activated hepatic stellate cells by mesenchymal stem cells. Biochem Biophys Res Commun 2007; 16;363(2):247-52.

26. Noda S, Masumi S, Moriyama M , et al. Population of hepatic macrophages and response of perfused liver to platelet-activating factor during production of thioacetamide-induced cirrhosis in rats. Hepatology 1996; 24(2):412-8.

27. Constantinou MA, Theocharis SE, Mikros E. Application of metabonomics on an experimental model of fibrosis and cirrhosis induced by thioacetamide in rats. Toxicol Appl Pharmacol 2007; 218(1):11-9

28. Hu Y, Liao L, Wang Q,et al. Isolation and identification of mesenchymal stem cells from human fetal pancreas. J Lab Clin Med 2003; 141(5): 342-9.

83

29. Martin JY, Dean DD, Cochran DL, et al. Proliferation, differentiation, and protein synthesis of human osteoblast-like cells (MG63) cultured on previously used titanium surfaces. Clin Oral Implants Res 1996; 7(1): 27-37.

30. Kim MY, Cho MY, Baik SK, et al. Histological subclassification of cirrhosis using the Laennec fibrosis scoring system correlates with clinical stage and grade of portal hypertension. J Hepatol 2011; 55(5): 1004-9.

31. Calvaruso V, Burroughs AK, Standish R, et al. Computer-assisted image

33. Veldt BJ, Lainé F, Guillygomarc'h A, et al. Indication of liver transplantation in severe alcoholic liver cirrhosis: quantitative evaluation and optimal timing.

J Hepatol 2002; 36(1): 93-8.

34. Wanless IR, Sweeney G, Dhillon AP, et al. Lack of progressive hepatic fibrosis during long-term therapy with deferiprone in subjects with transfusion-dependent beta-thalassemia. Blood 2002; 100(5): 1566-9.

35. Suk KT, Baik SK, Yoon JH, et al. Revision and update on clinical practice guideline for liver cirrhosis. Korean J Hepatol 2012; 18(1): 1-21.

84

36. Peng L, Xie DY, Lin BL, et al. Autologous bone marrow mesenchymal stem cell transplantation in liver failure patients caused by hepatitis B: short-term and long-term outcomes. Hepatology 2011; 54(3): 820-8.

37. Hatakeyama Y, Ono T, Sato N et al. Usefulness of the tactile sensor for estimating the degree of liver fibrosis and the DNA synthesis activity of remnant liver cells after partial hepatectomy. Fukushima J Med Sci 2002;

48(2):93-101.

38. Müller A, Machnik F, Zimmermann T et al. Thioacetamide-induced cirrhosis-like liver lesions in rats-usefulness and reliability of this animal model. Exp Pathol 1988; 34(4):229-36.

39. Li X, Benjamin IS, Alexander B. Reproducible production of thioacetamide-induced macronodular cirrhosis in the rat with no mortality. J Hepatol 2002;

36(4):488-93.

40. Abdel Aziz MT, Atta HM, Mahfouz S et al. Therapeutic potential of bone marrow-derived mesenchymal stem cells on experimental liver fibrosis. Clin Biochem 2007; 40(12):893-9.

41. Breitkopf K, Sawitza I, Gressner AM. Characterization of intracellular pathways leading to coinduction of thrombospondin-1 and TGF-beta1 expression in rat hepatic stellate cells. Growth Factors 2005; 23(2):77-85.

42. Wrana JL. Transforming growth factor-beta signaling and cirrhosis.

Hepatology 1999; 29(6):1909-10.

85

43. Friedman SL. Seminars in medicine of the Beth Israel Hospital, Boston. The cellular basis of hepatic fibrosis. Mechanisms and treatment strategies. N Engl J Med 1993; 328(25):1828-35.

44. Campbell JS, Hughes SD, Gilbertson DG, et al. Platelet-derived growth factor C induces liver fibrosis, steatosis, and hepatocellular carcinoma. Proc Natl Acad Sci U S A. 2005; 102(9):3389-94.

45. Friedman SL. Mechanisms of hepatic fibrogenesis. Gastroenterology 2008;

134(6):1655-69.

46. Inagaki Y, Okazaki I. Emerging insights into Transforming growth factor beta Smad signal in hepatic fibrogenesis. Gut 2007; 56(2):284-92.

47. Meindl-Beinker NM, Dooley S. Transforming growth factor-beta and hepatocyte transdifferentiation in liver fibrogenesis. J Gastroenterol Hepatol 2008; 23 Suppl 1:S122-7.

48. Grotendorst GR, Rahmanie H, Duncan MR. Combinatorial signaling pathways determine fibroblast proliferation and myofibroblast differentiation.

FASEB J 2004; 18(3):469-79.

49. Moreno M, Gonzalo T, Kok RJ, et al. Reduction of advanced liver fibrosis by short-term targeted delivery of an angiotensin receptor blocker to hepatic stellate cells in rats. Hepatology 2010; 51(3):942-52.

50. Le Moine O, Louis H, Sermon F, et al. Interleukin-10 and liver diseases.

Acta Gastroenterol Belg 1999; 62(1):1-8.

86

51. Thompson K, Maltby J, Fallowfield J, et al. Interleukin-10 expression and function in experimental murine liver inflammation and fibrosis. Hepatology 1998; 28(6):1597-606.

52. Hung KS, Lee TH, Chou WY, et al. Interleukin-10 gene therapy reverses thioacetamide-induced liver fibrosis in mice. Biochem Biophys Res Commun 2005; 336(1):324-31.

53. Safadi R, Ohta M, Alvarez CE, et al. Immune stimulation of hepatic fibrogenesis by CD8 cells and attenuation by transgenic interleukin-10 from hepatocytes. Gastroenterology 2004; 127(3):870-82.

54. Nakamura T, Nishizawa T, Hagiya M, et al. Molecular cloning and expression of human hepatocyte growth factor. Nature 1989; 342(6248):440-3.

55. Michalopoulos GK, DeFrances MC. Liver regeneration. Science 1997;

276(5309):60-6.

56. Ueki T, Kaneda Y, Tsutsui H, et al. Hepatocyte growth factor gene therapy of liver cirrhosis in rats. Nat Med 1999; 5(2):226-30.

57. Matsuda Y, Matsumoto K, Ichida T, et al. Hepatocyte growth factor suppresses the onset of liver cirrhosis and abrogates lethal hepatic dysfunction in rats. J Biochem 1995; 118(3):643-9.

87

58. Chen X, Li Y, Wang L, Katakowski M, et al. Ischemic rat brain extracts induce human marrow stromal cell growth factor production.

Neuropathology 2002; 22(4):275-9.

59. Gao B, Bataller R. Alcoholic liver disease: pathogenesis and new therapeutic targets. Gastroenterology 2011; 141(5): 1572-85.

60. Kim MY, Cho MY, Baik SK, et al. Beneficial effects of candesartan, an

60. Kim MY, Cho MY, Baik SK, et al. Beneficial effects of candesartan, an

관련 문서