Spatial Reconstruction of the Early Hepatic Transcriptomic Landscape after an Acetaminophen Overdose using Single-cell RNA Sequencing.

An acetaminophen (APAP) overdose is the most common cause of acute liver failure in the United States. A hallmark characteristic of APAP hepatotoxicity is centrilobular necrosis. General, innate mechanisms such as lower amounts of GSH and higher Cyp2e1 expression in pericentral (PC) hepatocytes are known to contribute to the differences in susceptibility to cell injury between periportal (PP) hepatocytes and PC hepatocytes. While a sequence of molecular events involving formation of the reactive metabolite N-acetyl-p-benzoquinone imine, GSH depletion, oxidative stress, and JNK activation define the early cell stress trajectory following APAP exposure, their activation in PC vs PP hepatocytes is not well characterized. By using single-cell RNA sequencing, we provide the first reconstruction of the early transcriptomic APAP liver lobule after validation of our methodology using human liver scRNA-seq data. Two hours after APAP treatment, we find that PP hepatocytes progress along the APAP stress axis to oxidative stress, before resolving injury due to innate and adaptive mechanisms. However, PC hepatocytes continue along this stress axis as indicated by activation of mitogen-activated protein kinase (MAPK) genes, which is absent in PP hepatocytes. We also identify a population of glutamine synthetase enriched PC hepatocytes in close proximity to the central vein, where a stepwise induction of a stress program culminated in cell death. Collectively, these findings elucidate a molecular sequence of events distinguishing the differential response to APAP exposure between PP and PC hepatocytes and identify a subset of uniquely susceptible PC hepatocytes.

[1]  H. Jaeschke,et al.  Mitochondrial Protein Adduct and Superoxide Generation are Prerequisites for Early Activation of c-Jun N-terminal Kinase within the Cytosol after an Acetaminophen Overdose in Mice. , 2020, Toxicology letters.

[2]  B. Sosa-Pineda,et al.  Metabolic and non-metabolic liver zonation is established non-synchronously and requires sinusoidal Wnts , 2020, eLife.

[3]  Bret Duffin,et al.  Reproducibility across single-cell RNA-seq protocols for spatial ordering analysis , 2019, PloS one.

[4]  Dominic Grün,et al.  A Human Liver Cell Atlas reveals Heterogeneity and Epithelial Progenitors , 2019, Nature.

[5]  Fabian J Theis,et al.  Current best practices in single‐cell RNA‐seq analysis: a tutorial , 2019, Molecular systems biology.

[6]  Alireza Hadj Khodabakhshi,et al.  Metascape provides a biologist-oriented resource for the analysis of systems-level datasets , 2019, Nature Communications.

[7]  S. Itzkovitz,et al.  Spatial sorting enables comprehensive characterization of liver zonation , 2019, Nature Metabolism.

[8]  H. Jaeschke,et al.  Mito-tempo protects against acute liver injury but induces limited secondary apoptosis during the late phase of acetaminophen hepatotoxicity , 2018, Archives of Toxicology.

[9]  Abdel-latif Seddek,et al.  Hepatic zonation of toxic metabolite formation: perspectives of matrix-assisted laser desorption mass spectrometry imaging , 2018, Archives of Toxicology.

[10]  L. Kuepfer,et al.  Spatio-temporal visualization of the distribution of acetaminophen as well as its metabolites and adducts in mouse livers by MALDI MSI , 2018, Archives of Toxicology.

[11]  H. Jaeschke,et al.  Mitochondrial dysfunction as a mechanism of drug-induced hepatotoxicity: current understanding and future perspectives , 2018, Journal of clinical and translational research.

[12]  Paul Hoffman,et al.  Integrating single-cell transcriptomic data across different conditions, technologies, and species , 2018, Nature Biotechnology.

[13]  H. Jaeschke,et al.  Mechanisms of acetaminophen hepatotoxicity and their translation to the human pathophysiology , 2017, Journal of clinical and translational research.

[14]  H. Jaeschke,et al.  Mitochondria-targeted antioxidant Mito-Tempo protects against acetaminophen hepatotoxicity , 2016, Archives of Toxicology.

[15]  I. Amit,et al.  Single-cell spatial reconstruction reveals global division of labor in the mammalian liver , 2016, Nature.

[16]  H. Jaeschke,et al.  Oxidative stress during acetaminophen hepatotoxicity: Sources, pathophysiological role and therapeutic potential , 2016, Redox biology.

[17]  R. Stewart,et al.  Single-cell RNA-seq reveals novel regulators of human embryonic stem cell differentiation to definitive endoderm , 2016, Genome Biology.

[18]  William M. Lee,et al.  Outcomes in Adults With Acute Liver Failure Between 1998 and 2013 , 2016, Annals of Internal Medicine.

[19]  Mitchell R. McGill,et al.  Mitochondrial protein adducts formation and mitochondrial dysfunction during N-acetyl-m-aminophenol (AMAP)-induced hepatotoxicity in primary human hepatocytes. , 2015, Toxicology and applied pharmacology.

[20]  Mitchell R. McGill,et al.  Lower susceptibility of female mice to acetaminophen hepatotoxicity: Role of mitochondrial glutathione, oxidant stress and c-jun N-terminal kinase. , 2014, Toxicology and applied pharmacology.

[21]  Hartmut Jaeschke,et al.  Plasma and liver acetaminophen-protein adduct levels in mice after acetaminophen treatment: dose-response, mechanisms, and clinical implications. , 2013, Toxicology and applied pharmacology.

[22]  F. Gonzalez,et al.  Peroxisome proliferator‐activated receptor alpha induction of uncoupling protein 2 protects against acetaminophen‐induced liver toxicity , 2012, Hepatology.

[23]  T. Quinn,et al.  Endogenous interleukin-4 regulates glutathione synthesis following acetaminophen-induced liver injury in mice. , 2012, Chemical research in toxicology.

[24]  L. MacMillan-Crow,et al.  Acetaminophen-Induced Hepatotoxicity in Mice Occurs with Inhibition of Activity and Nitration of Mitochondrial Manganese Superoxide Dismutase , 2011, Journal of Pharmacology and Experimental Therapeutics.

[25]  H. Jaeschke,et al.  The impact of partial manganese superoxide dismutase (SOD2)-deficiency on mitochondrial oxidant stress, DNA fragmentation and liver injury during acetaminophen hepatotoxicity. , 2011, Toxicology and applied pharmacology.

[26]  H. Jaeschke,et al.  c-Jun N-terminal kinase modulates oxidant stress and peroxynitrite formation independent of inducible nitric oxide synthase in acetaminophen hepatotoxicity. , 2010, Toxicology and applied pharmacology.

[27]  C. Zwingmann,et al.  Novel mechanisms of protection against acetaminophen hepatotoxicity in mice by glutathione and N‐acetylcysteine , 2010, Hepatology.

[28]  Neil Kaplowitz,et al.  Role of JNK Translocation to Mitochondria Leading to Inhibition of Mitochondria Bioenergetics in Acetaminophen-induced Liver Injury* , 2008, Journal of Biological Chemistry.

[29]  K. Jungermann,et al.  Functional Hepatocellular Heterogeneity , 2007, Hepatology.

[30]  M. Bonin,et al.  Differential gene expression in periportal and perivenous mouse hepatocytes , 2006, The FEBS journal.

[31]  N. Kaplowitz,et al.  c-Jun N-terminal kinase plays a major role in murine acetaminophen hepatotoxicity. , 2006, Gastroenterology.

[32]  H. Jaeschke,et al.  Peroxynitrite-Induced Mitochondrial and Endonuclease-Mediated Nuclear DNA Damage in Acetaminophen Hepatotoxicity , 2005, Journal of Pharmacology and Experimental Therapeutics.

[33]  H. Jaeschke,et al.  Acetaminophen-induced oxidant stress and cell injury in cultured mouse hepatocytes: protection by N-acetyl cysteine. , 2004, Toxicological sciences : an official journal of the Society of Toxicology.

[34]  L. James,et al.  Effect of N-acetylcysteine on acetaminophen toxicity in mice: relationship to reactive nitrogen and cytokine formation. , 2003, Toxicological sciences : an official journal of the Society of Toxicology.

[35]  H. Jaeschke,et al.  Peroxynitrite Is a Critical Mediator of Acetaminophen Hepatotoxicity in Murine Livers: Protection by Glutathione , 2002, Journal of Pharmacology and Experimental Therapeutics.

[36]  J. Hinson,et al.  Vascular and hepatocellular peroxynitrite formation during acetaminophen toxicity: role of mitochondrial oxidant stress. , 2001, Toxicological sciences : an official journal of the Society of Toxicology.

[37]  Hans van Dam,et al.  Distinct roles of Jun : Fos and Jun : ATF dimers in oncogenesis , 2001, Oncogene.

[38]  J. Corton,et al.  Peroxisome proliferator-activated receptor alpha-null mice lack resistance to acetaminophen hepatotoxicity following clofibrate exposure. , 2000, Toxicological sciences : an official journal of the Society of Toxicology.

[39]  J. Carbone,et al.  The PPAR activator docosahexaenoic acid prevents acetaminophen hepatotoxicity in male CD-1 mice. , 1999, Journal of toxicology and environmental health. Part A.

[40]  C. V. Smith,et al.  Inhibition of carbamyl phosphate synthetase-I and glutamine synthetase by hepatotoxic doses of acetaminophen in mice. , 1997, Toxicology and applied pharmacology.

[41]  Steven D. Cohen,et al.  Protection against Acetaminophen Hepatotoxicity by a Single Dose of Clofibrate: Effects on Selective Protein Arylation and Glutathione Depletion , 1996 .

[42]  Steven D. Cohen,et al.  Identification of the mouse liver 44-kDa acetaminophen-binding protein as a subunit of glutamine synthetase. , 1995, Toxicology and applied pharmacology.

[43]  P. Moldéus,et al.  Zonation of acetaminophen metabolism and cytochrome P450 2E1-mediated toxicity studied in isolated periportal and perivenous hepatocytes. , 1993, Biochemical pharmacology.

[44]  I. Calder,et al.  Peroxisome proliferators protect against paracetamol hepatotoxicity in mice. , 1992, Biochemical pharmacology.

[45]  A. Moorman,et al.  Hepatocellular heterogeneity in ammonia metabolism: demonstration of limited colocalization of carbamoylphosphate synthetase and glutamine synthetase. , 1991, European journal of cell biology.

[46]  T J Bucci,et al.  Immunohistochemical localization and quantification of the 3-(cystein-S-yl)-acetaminophen protein adduct in acetaminophen hepatotoxicity. , 1991, The American journal of pathology.

[47]  M. Tirmenstein,et al.  Subcellular binding and effects on calcium homeostasis produced by acetaminophen and a nonhepatotoxic regioisomer, 3'-hydroxyacetanilide, in mouse liver. , 1989, The Journal of biological chemistry.

[48]  K. Lindros,et al.  Glutathione replenishment capacity is lower in isolated perivenous than in periportal hepatocytes. , 1988, The Biochemical journal.

[49]  A. Y. Lu,et al.  N-acetyl-p-benzoquinone imine: a cytochrome P-450-mediated oxidation product of acetaminophen. , 1984, Proceedings of the National Academy of Sciences of the United States of America.

[50]  R. Gebhardt,et al.  Heterogeneous distribution of glutamine synthetase among rat liver parenchymal cells in situ and in primary culture. , 1983, The EMBO journal.

[51]  B B Brodie,et al.  Acetaminophen-induced hepatic necrosis. IV. Protective role of glutathione. , 1973, The Journal of pharmacology and experimental therapeutics.

[52]  B. Brodie,et al.  ACETAMINOPHEN-INDUCED HEPATIC NECROSIS. III. CYTOCHROME P-450-MEDIATED COVALENT BINDING IN VITRO , 1973 .

[53]  A. Loud A QUANTITATIVE STEREOLOGICAL DESCRIPTION OF THE ULTRASTRUCTURE OF NORMAL RAT LIVER PARENCHYMAL CELLS , 1968, The Journal of cell biology.

[54]  K. Kaneda,et al.  Liver microvascular architecture: an insight into the pathophysiology of portal hypertension. , 1999, Seminars in liver disease.

[55]  R. Gebhardt Metabolic zonation of the liver: regulation and implications for liver function. , 1992, Pharmacology & therapeutics.