Gastroenterology

Gastroenterology

Volume 128, Issue 5, May 2005, Pages 1405-1415
Gastroenterology

Basic-liver, pancreas, and biliary tract
Cystathionine β Synthase Deficiency Promotes Oxidative Stress, Fibrosis, and Steatosis in Mice Liver

https://doi.org/10.1053/j.gastro.2005.02.034Get rights and content

Background & Aims: Cystathionine β-synthase (CBS) deficiency causes severe hyperhomocysteinemia, which confers diverse clinical manifestations, notably liver disease. To investigate this aspect of hyperhomocysteinemia, we performed a thorough investigation of liver pathology in CBS-deficient mice, a murine model of severe hyperhomocysteinemia. Methods: The degree of liver injury and inflammation was assessed by histologic examination, by measurements of products of lipid peroxidation, and by formation of carbonyl groups on protein as a measure for the occurrence of protein oxidation. Analysis of profibrogenic, proinflammatory factors and cell apoptosis was performed by Western blots, real-time quantitative reverse-transcription polymerase chain reaction, caspase-3 activity, DNA laddering, and TUNEL assay. Results: Histologic evaluation of liver specimens of 8- to 32-week-old CBS-deficient mice showed that CBS-deficient mice develop inflammation, fibrosis, and hepatic steatosis, concomitant with an enhanced expression of tissue inhibitor of metalloproteinase-1, α-smooth muscle actin, pro(α)1 collagen type I, transforming growth factor-β1, and proinflammatory cytokines. Moreover, even if the proapoptotic protein Bax was dominantly expressed and Bcl-2 was down-regulated, caspase-3 was not activated, DNA laddering was not detected, and number of positive TUNEL cells was not increased in liver of CBS-deficient mice compared with wild-type mice. Conclusions: The results show that hyperhomocysteinemia in liver of CBS-deficient mice promotes oxidative stress, which may cause mitochondrial damage in association with activation of hepatic stellate cells, leading to liver injury. The absence of caspase-3 activation, DNA fragmentation, and TUNEL-positive cells shows that protective signals may counteract apoptotic signals in liver of CBS-deficient mice.

Section snippets

Animals and Experimental Protocols

Heterozygous CBS-deficient (CBS +/−) mice were generously donated by Dr. N. Maeda (Department of Pathology, University of North Carolina, Chapel Hill, NC) and maintained in a controlled environment with access to unlimited food and water. All animals received human care according to the criteria outlined in the guide for the Care and Use of Laboratory Animals. CBS +/− mice were bred to obtain homozygous CBS-deficient (CBS −/−) mice. This produced CBS −/− and wild-type (CBS +/+) mice from the

Total Plasma and Hepatic Homocysteine Levels in CBS-Deficient Mice

Plasma tHcy concentration was 50-fold higher in CBS −/− mice, which is considered as a murine model of severe hyperhomocysteinemia,6 than that in CBS +/+ mice (205 ± 86 vs. 3.9 ± 0.9 μmol/L, respectively; P < .0001 by Student t test [n = 4 for each]). The mean hepatic concentration of tHcy in CBS −/− mice was approximately 20-fold higher than that in CBS +/+ mice (tHcy concentrations were 6.44 ± 3.86 vs. 0.34 ± 0.14 nmol/mg of cellular protein, respectively; P < .001 by Student t test [n = 5

Discussion

Despite clinical evidence for the toxicity of excess Hcy, the specific cytopathologic mechanisms of hepatocellular injury because of hyperhomocysteinemia have not been fully explored. The present study provides evidence that hyperhomocysteinemia because of CBS deficiency in mice was associated with increased hepatic oxidative stress. Enhanced lipid peroxidation seems to be a key feature in the liver injury observed in CBS-deficient mice. The process of lipid peroxidation generates numerous

References (57)

  • S.F. Choumenkovitch et al.

    In the cystathionine β-synthase knockout mouse, elevations in total plasma homocysteine increase tissue S-adenosylhomocysteine, but responses of S-adenosylmethionine and DNA methylation are tissue specific

    J Nutr

    (2002)
  • K. Namekata et al.

    Abnormal lipid metabolism in cystathionine β-synthase-deficient mice, an animal model for hyperhomocysteinemia

    J Biol Chem

    (2004)
  • Z. Wu et al.

    Structure and functions of human oxysterol 7-hydroxylase cDNAs and gene CYP7B1

    J Lipid Res

    (1999)
  • M.D. Weltman et al.

    Increased hepatocyte CYP2E1 expression in a rat nutritional model of hepatic steatosis with inflammation

    Gastroenterology

    (1996)
  • L. Torres et al.

    Induction of TIMP-1 expression in rat hepatic stellate cells and hepatocytesa new role for homocysteine in liver fibrosis

    Biochim Biophys Acta

    (1999)
  • A.E. Reid

    Nonalcoholic steatohepatitis

    Gastroenterology

    (2001)
  • N. Nieto et al.

    Stimulation and proliferation of primary rat hepatic stellate cells by cytochrome P450 2E1-derived reactive oxygen species

    Hepatology

    (2002)
  • K.R. Feingold et al.

    Paraoxonase activity in the serum and hepatic mRNA levels decrease during the acute phase response

    Atherosclerosis

    (1998)
  • G.S. Hossain et al.

    TDAG51 is induced by homocysteine, promotes detachment-mediated programmed cell death, and contributes to the development of atherosclerosis in hyperhomocysteinemia

    J Biol Chem

    (2003)
  • G. Sass et al.

    Heme oxygenase-1 and its reaction product, carbon monoxide, prevent inflammation-related apoptotic liver damage in mice

    Hepatology

    (2003)
  • F.R. Murphy et al.

    Inhibition of apoptosis of activated hepatic stellate cells by tissue inhibitor of metalloproteinase-1 is mediated via effects on matrix metalloproteinase inhibitionimplications for reversibility of liver fibrosis

    J Biol Chem

    (2002)
  • J. George et al.

    Lipid peroxidation, stellate cell activation and hepatic fibrogenesis in a rat model of chronic steatohepatitis

    J Hepatol

    (2003)
  • A. Bosy-Westphal et al.

    Increased plasma homocysteine in liver cirrhosis

    Hepatol Res

    (2001)
  • M.A. Avila et al.

    Reduced mRNA abundance of the main enzymes involved in methionine metabolism in human liver cirrhosis and hepatocellular carcinoma

    J Hepatol

    (2000)
  • G. Varela-Moreiras et al.

    Carbon tetrachloride-induced hepatic injury is associated with global DNA hypomethylation and homocysteinemiaeffect of S-adenosylmethionine treatment

    Hepatology

    (1995)
  • S.H. Mudd et al.

    Disorders of transsulfuration

  • J. Selhub

    Homocysteine metabolism

    Annu Rev Nutr

    (1999)
  • G.N. Welch et al.

    Homocysteine and atherothrombosis

    N Engl J Med

    (1998)
  • Cited by (156)

    • Developmental and Inherited Liver Disease

      2023, MacSween's Pathology of the Liver, Eighth Edition
    View all citing articles on Scopus

    Supported by the European Union Grant (QLRT-2001-00816), a grant “vin et santé” and the Fondation de France, a fellowship from the Fondation pour la Recherche Médicale (FRM) and the GEHT-ISTH 2001 (to K.R.), and a fellowship from the Société Française d’Hypertension Artérielle (SFHTA; to J.N.).

    1

    K.R. and J.N. contributed equally to this paper.

    2

    E.B.’s current address is EA 2526, Laboratoire de Biochimie et Génétique Moléculaire; Faculté des Sciences et Technologies, Université de la Réunion, 15, Avenue René Cassin, BP 7151; 97715 Saint Denis Messag Cedex 09, La réunion, France.

    View full text