Systems Genetics of Liver Fibrosis: Identification of Fibrogenic and Expression Quantitative Trait Loci in the BXD Murine Reference Population

The progression of liver fibrosis in response to chronic injury varies considerably among individual patients. The underlying genetics is highly complex due to large numbers of potential genes, environmental factors and cell types involved. Here, we provide the first toxicogenomic analysis of liver fibrosis induced by carbon tetrachloride in the murine ‘genetic reference panel’ of recombinant inbred BXD lines. Our aim was to define the core of risk genes and gene interaction networks that control fibrosis progression. Liver fibrosis phenotypes and gene expression profiles were determined in 35 BXD lines. Quantitative trait locus (QTL) analysis identified seven genomic loci influencing fibrosis phenotypes (pQTLs) with genome-wide significance on chromosomes 4, 5, 7, 12, and 17. Stepwise refinement was based on expression QTL mapping with stringent selection criteria, reducing the number of 1,351 candidate genes located in the pQTLs to a final list of 11 cis-regulated genes. Our findings demonstrate that the BXD reference population represents a powerful experimental resource for shortlisting the genes within a regulatory network that determine the liver's vulnerability to chronic injury.

[1]  F. Lammert,et al.  Lith genes control mucin accumulation, cholesterol crystallization, and gallstone formation in A/J and AKR/J inbred mice , 2002, Hepatology.

[2]  B. McMahon,et al.  Chronic hepatitis B , 2001, Hepatology.

[3]  D. Houlihan,et al.  Pioglitazone, vitamin E, or placebo for nonalcoholic steatohepatitis. , 2010, The New England journal of medicine.

[4]  F. Tacke,et al.  The fractalkine receptor CX3CR1 is involved in liver fibrosis due to chronic hepatitis C infection. , 2008, Journal of Hepatology.

[5]  Z. Kutalik,et al.  Genome-wide association study identifies variants associated with progression of liver fibrosis from HCV infection. , 2012, Gastroenterology.

[6]  Ivan Rusyn,et al.  Genome‐level analysis of genetic regulation of liver gene expression networks , 2007, Hepatology.

[7]  J. Pfeilschifter,et al.  CXCL10 promotes liver fibrosis by prevention of NK cell mediated hepatic stellate cell inactivation. , 2010, Journal of autoimmunity.

[8]  B. Spear,et al.  The mouse alpha-albumin (afamin) promoter is differentially regulated by hepatocyte nuclear factor 1α and hepatocyte nuclear factor 1β. , 2011, DNA and cell biology.

[9]  F. Lammert,et al.  Genetic determinants in hepatic fibrosis: from experimental models to fibrogenic gene signatures in humans. , 2008, Clinics in liver disease.

[10]  Elizabeth S. Pietras,et al.  Chronic Hepatitis: An Update on Terminology and Reporting , 1996 .

[11]  Robert W. Williams,et al.  Systems Genetics of Metabolism: The Use of the BXD Murine Reference Panel for Multiscalar Integration of Traits , 2012, Cell.

[12]  R. Bataller,et al.  Amendment history : Corrigendum ( April 2005 ) Liver fibrosis Ramón Bataller , 2018 .

[13]  R. Schwabe,et al.  TLR4 enhances TGF-beta signaling and hepatic fibrosis. , 2007, Nature medicine.

[14]  Wei Wang,et al.  The polymorphism architecture of mouse genetic resources elucidated using genome-wide resequencing data: implications for QTL discovery and systems genetics , 2007, Mammalian Genome.

[15]  S. Kojima,et al.  Impaired liver regeneration in mice by lipopolysaccharide via TNF-alpha/kallikrein-mediated activation of latent TGF-beta. , 2002, Gastroenterology.

[16]  S. Ryu,et al.  Afamin secreted from nonresorbing osteoclasts acts as a chemokine for preosteoblasts via the Akt-signaling pathway. , 2012, Bone.

[17]  V. N. Finelli,et al.  A simple method to determine nanogram levels of 4-hydroxyproline in biological tissues. , 1981, Analytical biochemistry.

[18]  M. Hørder,et al.  International Federation of Clinical Chemistry (IFCC) Scientific Committee, Analytical Section: approved recommendation (1985) on IFCC methods for the measurement of catalytic concentration of enzymes. Part 2. IFCC method for aspartate aminotransferase (L-aspartate: 2-oxoglutarate aminotransferase, , 1986, Journal of clinical chemistry and clinical biochemistry. Zeitschrift fur klinische Chemie und klinische Biochemie.

[19]  F. Lammert,et al.  Genome-wide analysis of hepatic fibrosis in inbred mice identifies the susceptibility locus Hfib1 on chromosome 15. , 2002, Gastroenterology.

[20]  R. Doerge,et al.  Empirical threshold values for quantitative trait mapping. , 1994, Genetics.

[21]  Albert-László Barabási,et al.  Genetic Dissection of Transcriptional Regulation in Budding Yeast , 2002 .

[22]  B. Motta,et al.  Recombinant PNPLA3 protein shows triglyceride hydrolase activity and its I148M mutation results in loss of function. , 2014, Biochimica et biophysica acta.

[23]  Klaus Schughart,et al.  QTLminer: identifying genes regulating quantitative traits , 2010, BMC Bioinformatics.

[24]  S. Sookoian,et al.  Meta‐analysis of the influence of I148M variant of patatin‐like phospholipase domain containing 3 gene (PNPLA3) on the susceptibility and histological severity of nonalcoholic fatty liver disease , 2011, Hepatology.

[25]  Florian Kronenberg,et al.  Characterization of the vitamin E-binding properties of human plasma afamin. , 2002, Biochemistry.

[26]  Robert W. Williams,et al.  Complex trait analysis of gene expression uncovers polygenic and pleiotropic networks that modulate nervous system function , 2005, Nature Genetics.

[27]  W. G. Hill,et al.  Heritability in the genomics era — concepts and misconceptions , 2008, Nature Reviews Genetics.

[28]  P. Tontonoz,et al.  Liver X receptor signaling is a determinant of stellate cell activation and susceptibility to fibrotic liver disease. , 2011, Gastroenterology.

[29]  Udo Hoffmann,et al.  Genome-Wide Association Analysis Identifies Variants Associated with Nonalcoholic Fatty Liver Disease That Have Distinct Effects on Metabolic Traits , 2011, PLoS genetics.

[30]  B. Taylor,et al.  Genetic Analysis of Resistance to Cadmium-Induced Testicular Damage in Mice , 1973, Proceedings of the Society for Experimental Biology and Medicine. Society for Experimental Biology and Medicine.

[31]  A. El-Karef,et al.  Expression of large tenascin-C splice variants by hepatic stellate cells/myofibroblasts in chronic hepatitis C. , 2007, Journal of hepatology.

[32]  F. Tacke,et al.  Antifibrotic effects of CXCL9 and its receptor CXCR3 in livers of mice and humans. , 2009, Gastroenterology.

[33]  S. Werner,et al.  Skin wounds and severed nerves heal normally in mice lacking tenascin-C. , 1996, Proceedings of the National Academy of Sciences of the United States of America.

[34]  Eric E. Schadt,et al.  Moving toward a system genetics view of disease , 2007, Mammalian Genome.

[35]  C. Haley,et al.  A simple regression method for mapping quantitative trait loci in line crosses using flanking markers , 1992, Heredity.

[36]  L. Kruglyak,et al.  Genetic Dissection of Transcriptional Regulation in Budding Yeast , 2002, Science.

[37]  H. Bergmeyer IFCC methods for the measurement of catalytic concentrations of enzymes: Part 3. IFCC method for alanine aminotransferase (L-alanine: 2-oxoglutarate aminotransferase, EC 2.6.1.2)☆ , 1980 .

[38]  Robert W. Williams,et al.  WebQTL - Web-based complex trait analysis , 2003, Neuroinformatics.

[39]  T. Sakakura,et al.  Tenascin‐C upregulates matrix metalloproteinase‐9 in breast cancer cells: Direct and synergistic effects with transforming growth factor β1 , 2003, International journal of cancer.

[40]  M. Hørder,et al.  International Federation of Clinical Chemistry (IFCC) Scientific Committee, Analytical Section: approved recommendation (1985) on IFCC methods for the measurement of catalytic concentration of enzymes. Part 3. IFCC method for alanine aminotransferase (L-alanine: 2-oxoglutarate aminotransferase, EC 2 , 1986, Journal of clinical chemistry and clinical biochemistry. Zeitschrift fur klinische Chemie und klinische Biochemie.

[41]  S. Kojima,et al.  Impaired liver regeneration in mice by lipopolysaccharide via TNF-α/kallikrein–mediated activation of latent TGF-β , 2002 .

[42]  P. Shannon,et al.  Cytoscape: a software environment for integrated models of biomolecular interaction networks. , 2003, Genome research.

[43]  P. Bedossa,et al.  Natural history of liver fibrosis progression in patients with chronic hepatitis C , 1997, The Lancet.

[44]  Zimmerman Hj,et al.  Morphologic spectrum of drug-induced hepatic disease. , 1995 .

[45]  Robert W. Williams Animal Models in Biomedical Research , 2006 .

[46]  Marcin Krawczyk,et al.  Genome-wide association studies and genetic risk assessment of liver diseases , 2010, Nature Reviews Gastroenterology &Hepatology.

[47]  I. Leclercq,et al.  Pro-oxidant-mediated hepatic fibrosis and effects of antioxidant intervention in murine dietary steatohepatitis. , 2009, International journal of molecular medicine.

[48]  S. Friedman,et al.  Pathogenesis of liver fibrosis. , 2011, Annual review of pathology.

[49]  Robert W. Williams,et al.  A new set of BXD recombinant inbred lines from advanced intercross populations in mice , 2004, BMC Genetics.

[50]  Tom H. Pringle,et al.  The human genome browser at UCSC. , 2002, Genome research.

[51]  K. Broman,et al.  Review of statistical methods for QTL mapping in experimental crosses. , 2001, Lab animal.

[52]  Magalie S Leduc,et al.  Using bioinformatics and systems genetics to dissect HDL-cholesterol genetics in an MRL/MpJ × SM/J intercross[S] , 2012, Journal of Lipid Research.

[53]  J. Schwarzbauer,et al.  Meet the Tenascins: Multifunctional and Mysterious* , 2005, Journal of Biological Chemistry.

[54]  Jörg Köhl,et al.  Complement factor 5 is a quantitative trait gene that modifies liver fibrogenesis in mice and humans , 2005, Nature Genetics.

[55]  E. Chesler,et al.  Heritability, correlations and in silico mapping of locomotor behavior and neurochemistry in inbred strains of mice , 2004, Genes, brain, and behavior.

[56]  F. Lammert,et al.  Variant adiponutrin (PNPLA3) represents a common fibrosis risk gene: non-invasive elastography-based study in chronic liver disease. , 2011, Journal of hepatology.

[57]  M. Kashiwagi,et al.  Tenascin-C is an endogenous activator of Toll-like receptor 4 that is essential for maintaining inflammation in arthritic joint disease , 2009, Nature Medicine.