* and a, DOI: 10.1097/MEG.0000000000002034 1 DOI: Shivaram P. Singh , Shivaram P. c play important roles in regulating host defense Cytokines play important roles in regulating Cytokines are cellu- and maintenance of homeostasis [5]. through cells on other their activity exert that products lar Their mechanisms [6]. paracrine and endocrine autocrine, they can control a host and activity is receptor-mediated dif- proliferation, such as cellular of biologic functions, responses [5,6]. activation and inflammatory ferentiation, Cytokines in viral infections Toll-like includes The artillery against invading viruses the RIG-I-like receptors Nod-like receptors and receptors, pattern recognition recep- which can identify specific [7], by attack an launch and surface viral the on tors a major role in inducing Cytokines play production [8]. activating by cells virus-infected of killing and apoptosis of site the to them recruiting and cells specific immune interferons (IFNs) Of the different cytokines, damage [9]. provide the most robust response against viral injury [10]. IFNs can induce the release of a number of proteins and IFN-induced peptides that have an antiviral action [11]. player in a key to be found has been chemokine CXCL10 mounting an antiviral response in several viral-mediated respiratory syndromes [12]. and cytokines Coronaviruses Many of the systemic manifestations in severe acute res- could be explained by an piratory syndrome (SARS) immune-mediated response rather than a direct viral a In animal models of murine hepatitis virus, injury [13]. disease manifestations corre- widely studied coronavirus, rather than viral replication lated with immune response, B-cells) mice T- and (lacking While immunodeficient [14]. the addition of virus-spe- do not develop demyelination, The timing T-cells causes demyelination to occur [15]. cific Cytokines , Fuad Z . Aloor

c,d,e Department of d Division of f , Negin Hajizadeh b,c * f, , Sonali Narain a Department of Medicine, Donald and Barbara Zucker School of c

Review Department of Medicine, Division of Rheumatology, Northwell Health, Department of Medicine, Division of Rheumatology, Copyright © 2020 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited. Copyright © 2020 Wolters b

Liver dysfunction manifesting as elevated aminotransferase levels has been a common feature of coronavirus of coronavirus has been a common feature as elevated aminotransferase levels Liver dysfunction manifesting it has been However, in COVID-19 infection is unclear. infection. The mechanism of liver injury disease-2019 (COVID-19) multiorgan storm-related cytokine and dysfunction immune virus, the of effects cytopathic direct of result a be to hypothesized of injury due to medications used in the management and idiosyncratic drug-induced liver injury damage, hypoxia-reperfusion of COVID-19 is cytokine storm, of liver injury in the setting the pathophysiology regarding hypothesis COVID-19. The favored we have review, In the current of cytokines. response leading to hyperproduction inflammatory an aberrant and unabated Eur J literature. reported liver injury based on the pathophysiologic mechanisms of cytokine-induced summarized the potential Hepatol XXX: 00–00 Gastroenterol Kluwer Health, Inc. All rights reserved. Copyright © 2020 Wolters Division of Pulmonary, Critical Care and Sleep Medicine, Department of and Sleep Medicine, Critical Care Division of Pulmonary, Department of Gastroenterology, S.C.B. Medical College, Cuttack, Department of Gastroenterology, Kluwer Health, Inc. All rights reserved. 0954-691X Copyright © 2020 Wolters Sanjaya K. Satapathy Introduction corona- syndrome, Despite being primarily a respiratory has been found to cause virus disease-2019 (COVID-19) In a study involv- significant multiorgan dysfunction [1,2]. more than half of the ing 5700 patients with COVID-19, levels patients presented with elevated aminotransferase infection of liver injury in COVID-19 mechanism The [3]. of direct cytopathic has been postulated to be a result imbalance and cytokine immune effects of the virus, hypoxia-reperfusion multiorgan damage, storm-related liver injury dysfunction and idiosyncratic drug-induced of COVID- due to medications used for the management review is to consolidate The focus of the current 19 [4]. as cytokine surge implications of our understanding of the reference to evidence a cause of hepatic injury with special in the context of COVID-19.

disease-2019 (COVID-19): untangling the knots untangling (COVID-19): disease-2019 Prajna Anirvan Cytokine-induced liver injury in coronavirus in coronavirus injury liver Cytokine-induced Information Services, Institute of Health, Innovations and Outcomes Research, Information Services, Institute of Health, Innovations and Outcomes Research, Northwell Health, New Hyde Park, Feinstein Institutes for Medical Research, Medicine, Northwell Health, Manhasset, New York, USA and Medicine, Northwell Health, Manhasset, New York, India, Medicine at Hofstra/Northwell, Northwell Health, Hempstead, Hepatology at Sandra Atlas Bass Centre for Liver Diseases and Transplantation, Liver Diseases and Transplantation, for Hepatology at Sandra Atlas Bass Centre Barbara and Zucker School of Medicine/Northwell Health, Manhasset, New USA York, DM, Donald and Barbara MBBS, MD, to Sanjaya K. Satapathy, Correspondence Zucker School of Medicine at Hofstra/ Northwell Health, 400 Community Drive, 11030, USA Manhasset, New York +516 562 5010; 562 fax: +516 2688;Tel: e-mail: [email protected] Sanjaya K. Satapathy contributed equally to the Singh and Dr. Shivaram P. *Dr. writing of this article. Received 27 September 2020 Accepted 23 November 2020 e a European Journal of Gastroenterology & Hepatology 2020, XXX:00–00 Journal of Gastroenterology European syndrome, disease-2019, cytokine release coronavirus Keywords: transaminitis endotheliitis, liver injury, Manhasset,

Downloaded from https://journals.lww.com/eurojgh by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCywCX1AWnYQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC4/OAVpDDa8K2+Ya6H515kE= on 01/15/2021 LWW Downloaded from https://journals.lww.com/eurojgh by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCywCX1AWnYQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC4/OAVpDDa8K2+Ya6H515kE= on 01/15/2021 2 European Journal of Gastroenterology & Hepatology xxx 2020 • Volume XXX • Number XXX of elevations in inflammatory cytokines and CRP corre- mortality [31]. The presence of increased levels of IL-1 lated with progression of pulmonary infiltrates of SARS beta (IL-1ß), IFN-γ and CXCL10 in COVID-19 patients patients [16]. In Middle Eastern respiratory syndrome implicate a potential T-helper-1 (Th1) cell-mediated (MERS) patients too, increase in serum levels of CXCL10 response [32]. When initial immune responses fail to con- correlated with disease severity [17]. trol viral replication, the virus may induce target cells to The role of cytokines as potentially important mediators release overwhelming amounts of cytokines and thus lead of the pathophysiology of COVID-19 has been established to pathological lesions [33]. In a randomized study com- with the demonstration that proinflammatory cytokines paring the neutralizing antibody Ly-Cov555 to placebo, and chemokines – tumor necrosis factor-alfa (TNFα) and the persistence of a high viral load at Day 7 was associated IFN-gamma (IFNγ), CXCL10 and CXCL8 – were higher with more severe forms of COVID-19, which is consist- in the plasma of COVID-19 patients compared to healthy ent with this assertion [34]. In patients of COVID-19 with controls [18]. Circulating concentrations of CXCL10, comorbidities, such as mellitus and factors like CCL2 and TNFα were also found to be significantly higher reduced T-cell function, diminished viral clearance and a in patients requiring admission to intensive care units [18]. state of chronic ultimately culminate in the Inflammatory markers such as C-reactive protein (CRP), genesis of a cytokine storm [35]. lactate dehydrogenase (LDH) and procalcitonin have been Activated T-cells release IFN and TNF-α, which, in found to be frequently elevated in COVID-19 patients turn, cause activation of macrophages and dendritic cells. [19]. Many of the early studies from China and Italy have These cells produce large amounts of IL-6 which, in a kind reported that elevated ferritin, CRP, LDH, interleukin 6 (IL- of vicious cycle, perpetuates the inappropriate immune 6) and D-dimer correlated with severe illness and increased response. In the early phase, TNF-α, IL-1ß, IL-8 and mono- mortality [20]. In a retrospective cohort study, severe acute cyte chemoattractant protein-1 appear which is followed liver injury was found to be significantly associated with by a sustained increase in IL-6 [6]. IL-6 contributes to elevated inflammatory markers including ferritin and IL-6 the characteristic symptoms of cytokine storm-increased [21]. In another retrospective, single-center study of 148 vascular permeability and coagulopathy [29]. Thus, the patients with COVID-19 from China, higher levels of pro- surge of massive amounts of pro-inflammatory cytokines calcitonin and CRP were found in patients presenting with [IFNs, ILs, TNFα and Transforming Growth Factor β abnormal liver function tests [22]. This appears quite sim- (TGFβ)] and chemokines such as CXCL10, CXCL8 and ilar to studies in SARS and MERS where IL, TNF-α and CCL5 generates and sustains the aberrant systemic inflam- endotoxins were higher in patients with liver injury com- matory response [18]. These events may be followed by pared to those with preserved liver function, [23,24] indi- an anti-inflammatory response in which there is a rise in cating that in addition to the association of cytokine storm IL-10 with concurrent downregulation of neutrophil and with disease outcomes, cytokine release could possibly also lymphocyte function – an event termed ‘immunoparalysis’ induce and propagate hepatic dysfunction. [36]. Although apparently favorable, immunoparalysis is associated with poor prognosis [37]. In COVID-19, continuously high levels of CXCL10 Cytokine release syndrome- pathophysiology and CCL7 were found to be associated with increased Cytokine storm or cytokine release syndrome (CRS) can be viral load, loss of lung function, lung injury and a fatal defined as an aberrant, exaggerated and unabated systemic outcome [38]. Upregulation of genes involved in apoptosis inflammatory response precipitated by infection, drugs or and p53 signaling have also been observed in COVID 19 as an allogenic response to foreign tissue resulting in the – a likely explanation of lymphopenia commonly encoun- overproduction of pro-inflammatory cytokines, leading to tered in these patients [39]. TNF-α-mediated apoptosis multiorgan dysfunction [25]. Cytokine storms have been of lymphocytes has been documented in several studies associated with a number of diseases, including the avian [40,41]. Lymphopenia, in turn, has been associated with H5N1 virus infection [26]. Cytokine storm is poorer outcomes in COVID-19 patients [42]. CRP values usually heralded by local inflammation that spills over to ≥20 mg/L and a lymphocyte count <1.1 × 109/L have been the systemic circulation [6]. Local inflammation involves found to be independent risk factors for liver injury in vasodilatation, increased vascular permeability and recruit- COVID-19 [42]. Lymphopenia can cause an unrestrained ment of inflammatory mediators to the site of involvement expression of the immune response leading to organ dam- aimed at generating a host response. Dysregulation of this age [43]. This might be one mechanism of how lymphope- immune response causes increased production of cytokines nia in COVID-19 may upregulate inflammatory mediators and chemokines, which in turn, act as chemoattractants for such as IL-6, IL-2 and IFN-γ and not only cause pulmonary other inflammatory cells, resulting in exaggerated immune injury, but also involve extra-pulmonary organs such as response with consequent systemic manifestations [27]. the liver [44]. It is quite interesting to note that lymphope- IL-6, IL-10 and IFN-γ are the primary cytokines that are nia, despite being an effect of COVID-19-triggered apop- consistently found to be elevated in the serum of patients tosis, can perpetuate an exaggerated immune response. with CRS [28,29]. Moreover, the differential expression of Therefore, the cytokine storm represents the extremes of chemokines, such as CXCL9, CXCL10 and CXCL11, has immune system dysfunction and dysregulation that leads been found to regulate T-cell function and infiltration in to systemic complications and unfavorable outcomes. the periphery during inflammation [30]. In COVID-19, similar to MERS CoV and SARS CoV Clinical manifestations of cytokine storm infections, the key immune-mediated manifestation of end-organ damage is the acute respiratory distress syn- Cytokine storm can present as a spectrum of illness drome (ARDS), which is associated with high short-term ranging from mild flu-like symptoms to life-threatening

Copyright © 2020 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited. Cytokine-induced liver injury in coronavirus disease-2019 Anirvan et al. www.eurojgh.com 3 manifestations such as hyperpyrexia, shock requiring vas- mention that angiotensin converting enzyme 2 (ACE2)- opressor support, disseminated intravascular coagulation mediated entry of SARS CoV-2 leads to ACE2 receptor and multiorgan dysfunction [45]. Respiratory involve- shedding [57]. ACE2 downregulation has been linked to ment may progress to ARDS [45]. In addition, renal injury, vascular dysfunction through the induction of a state of cytopenias, coagulopathy and cardiac dysfunction may NO deficiency and oxidative stress [58]. Suppression of occur [45]. Neurological symptoms, ranging from mild superoxide production markedly alleviates influenza A confusion to seizures, may occur [46]. Cytokine storm can virus-induced lung injury and virus replication, irrespec- sometimes, in severe cases, resemble macrophage activa- tive of the infecting strain [59]. Preliminary studies sug- tion syndrome or hemophagocytic lymphohistiocytosis gest that alpha-lipoic acid treatment might be able to (HLH) with similar biochemical abnormalities [47]. improve the 30-day survival rate of patients with critically ill COVID-19, indicating a role of ROS generation in the pathophysiology of COVID-19 [60]. Pulmonary capillary Liver injury in the setting of cytokine storm leak syndrome secondary to SARS-CoV-2 virus infection Hepatic involvement in patients in critical care units with and development of noncardiogenic pulmonary edema or other insults resulting in cytokine surge usually has recently been documented [61]. Thus, increased vas- manifests either transaminitis, predominantly with AST cular permeability secondary to cytokine release, hypoper- elevation and uncommonly with mild hyperbilirubine- fusion, endothelial dysfunction, ROS generation and NO mia with or without alkaline phosphatase elevation. In a deficiency underlie the pathophysiology of liver dysfunc- study on 148 patients of COVID-19, more than one-third tion due to systemic effects of the cytokine storm. of patients admitted to the hospital were found to have the abnormal liver function, manifested chiefly as derange- Cytokine storm and renin-angiotensin-aldosterone system ments in alanine aminotransferase (ALT) (18.2%) and aspartate aminotransferase (AST) (21.6%), which was Involvement of the renin-angiotensin-aldosterone sys- associated with a longer duration of hospital stay [22]. In tem (RAAS) has been observed in the setting of hyper- addition, patients with abnormal liver function had higher cytokinemia [62]. TNF-α-mediated lung injury requires levels of procalcitonin and CRP [22]. Further research is the mediation of angiotensin II [63]. Angiotensin II has needed to understand the pathophysiology of cytokine-in- been linked to inflammation, tissue injury and oxidative duced hepatic injury in the light of the complex interplay stress [64]. Angiotensin II can induce inflammation via the between the offending cytokines, the systemic adverse angiotensin 1 receptor (AT1) [64]. ACE2 downregulation effects of cytokine release, the local injury affected and the has been linked to thrombosis through multiple mecha- hepatic response to such injury. nisms [65]. Loss of ACE 2 has been shown to shift the RAAS axis away from the anti-inflammatory angiotensin Liver injury due to systemic effects of cytokine release (1–7)/angiotensin (1–9) pathways and augment the proin- flammatory angiotensin II–AT1 pathway with deleterious The liver is extremely sensitive to disturbances of systemic consequences [66]. Imbalance of the RAAS has also been homoeostasis and is affected through multiple mecha- linked to induction of thrombosis through the expression nisms, which include both vascular and immune-mediated of plasminogen activator inhibitor-1 [67], bradykinin pathways [48]. We describe here the major mechanisms accumulation [68] and angiotensin IV [69] and aldoster- of cytokine-induced systemic effects that contribute to one-mediated [70] impairment of fibrinolysis. The pres- hepatic injury. ence of local RAAS in the liver [71] and an imbalance between the various components of RAAS, including the Cytokine-induced vascular dysfunction, hypoperfusion canonical and noncanonical axes, may play a pivotal role and oxidative stress in hepatocyte inflammation and injury [72]. Cytokines are known to cause adhesion of immune cells to endothelial cells, thereby increasing vascular permea- Local effects of cytokine release bility [49]. TNF-α can induce endothelial dysfunction in The liver, although classically believed to be an immuno- pulmonary arteries in response to thrombin [50]. TNF- privileged site [73], is replete with inflammatory ticking α also impairs endothelium-dependent and nitric oxide bombs, comprising of a heterogeneous milieu of inflamma- (NO)-mediated vasodilation in various vascular beds as tory components [74,75]. Dysregulated cytokine release evidenced in experimental models [51]. Cytokines could can elicit an acute phase response in the hepatocytes [76]. also potentially lead to increased generation of reactive An overview of local injury in the liver and the hepatic oxygen species (ROS) which, in turn, may cause cellular response to such an injury is described below. injury and death [52]. This excess activation of cytokines causing endothelial dysfunction and ROS generation can Kupffer cells, endothelial dysfunction, hepatocyte damage result in vascular leakage and and microthrombosis [53]. Capillary leak syndrome can, by itself, cause alveolar edema and hypoxia, and can produce edema in different In the liver, Kupffer cells, neutrophils, hepatocytes and organs, such as the brain, heart, liver and kidneys [54]. liver sinusoidal endothelial cells (LSECs) mediate the This is corroborated by studies demonstrating a reduction hepatic response to cytokine release [77]. Hepatocellular of microcirculatory blood flow in the liver to 50% of base- damage can trigger Kupffer cell activation [78]. Kupffer line during hypodynamic septic shock [55]. cells release cytokines, ROS and NO which induce LSEC NO deficiency in vascular endothelium may contribute and can perpetuate hepatocyte injury [77]. TNF-α can to the pathophysiology of COVID-19 [56]. It needs no directly stimulate hepatocytes to produce IL-6, which, in

Copyright © 2020 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited. 4 European Journal of Gastroenterology & Hepatology xxx 2020 • Volume XXX • Number XXX turn, increases caspase-3 and leads to hepatocellular apop- classically described in immunodeficiency syndromes, an tosis [79]. NO generated by activated neutrophils can lead acquired form induced by offending agents – viruses, bac- to peroxynitrite formation, a potent ROS that can cause teria and malignancies – has been increasingly described mitochondrial damage with consequent hepatocellular [93]. The pathophysiology of acquired HLH probably necrosis and apoptosis [77]. This interplay between sinu- involves suppression of T-cell function by viral infections soidal endothelial cells, hepatic macrophages and leuko- [94]. Hepatic injury in HLH is mainly due to cytokine cytes leads to a redistribution of intrahepatic blood flow storm, which results due to impaired natural killer/cyto- causing decreased sinusoidal perfusion [80]. Endothelial toxic T lymphocyte function [95]. Macrophage-derived dysfunction, neutrophil invasion and microthrombi for- IL-2, IFN-γ and TNF-α have been found to produce mation further enhance liver tissue ischemia and damage porto-sinusoidal inflammation that leads to transamini- [77]. Liver biopsy specimen of a patient with COVID-19 tis, sinusoidal dilatation and congestion and Kupffer cell revealed microvesicular steatosis along with lobular and hyperplasia [95]. Interestingly, it has been postulated based portal activity, probably implying viral-mediated injury or on robust evidence that severe COVID-19-associated a possibility of drug-induced liver injury [81]. However, pneumonia patients may exhibit several features of sys- in a study carried out by Liu et al. [82], autopsy of the temic hyperinflammation designated as secondary HLH liver of COVID-19 patients revealed hepatomegaly with [96]. The inflammatory response in COVID-19 pneumo- hepatocyte degeneration accompanied by lobular focal nia is significant, but does not seem to be as pronounced necrosis and neutrophil infiltration, portal infiltration of as that found in HLH. Nevertheless, data suggest that sec- lymphocytes and , and congestion of hepatic ondary HLH may play a crucial role in the pathogenesis sinuses with microthrombosis. In another autopsy case of severe COVID-19. A recent study showed that hemo- series, thrombosis was found in multiple organs and was phagocytosis is a common finding in the bone marrow of present even after anticoagulation in some cases with deceased patients with severe COVID-19 [97]. the liver showing accumulation of platelet-fibrin micro- The laboratory profile of severe COVID-19 pneumonia thrombi in the hepatic sinusoids [83]. Recent consensus patients have revealed highly elevated CRP and ferritin statements on the treatment of coagulation dysfunction levels, which are central to the diagnosis of HLH [98]. in COVID-19 [84] have highlighted the microvascular The presence of abnormal liver function tests and coagu- damage due to intense inflammation occurring in differ- lopathy may imply the diagnosis of secondary HLH in a ent organs. This manifests as extensive microthrombosis subset of COVID-19 patients [99]. Elevation of particu- in the pulmonary circulation [85] and could possibly, in lar cytokines, such as IL-1β, IL-2, IL-6, TNF and CCL2, our opinion, involve other organs, such as the liver. ACE2 has been observed in COVID-19 patients which is similar receptors, through which SARS-CoV-2 gains access to the to that in secondary HLH [100]. The pathophysiology of host cell, are also expressed by endothelial cells [86]. Viral cytokine-induced liver dysfunction is illustrated in Fig. 1. elements are indeed present within endothelial cells with evidence of endothelial and inflammatory cell death [2]. Pattern of liver injury in cytokine storm These findings suggest that SARS-CoV-2 infection induces endotheliitis in several organs as a direct consequence Hypercytokinemia-induced liver injury can be either of viral involvement and aberrant host inflammatory hypoxic/hepatocellular injury manifesting as transaminitis response. Histopathological data on the liver in patients or cholestatic injury presenting with hyperbilirubinemia. with COVID-19 are emerging [87], and the occurrence Sepsis is an important promoter of liver injury causing of hepatic microthrombosis could be a potential mecha- both hyperbilirubinemia and hypoxic hepatitis [101,102]. nism of liver injury. In fact, there is now emerging evi- A recent clinical trial involving 312 patients with sep- dence to support anticoagulation as a treatment strategy sis reported 20% prevalence of acute liver failure [103]. for COVID-19 [88]. Patients with chemotherapy induced cytokine storm and Visualization of hepatobiliary excretory function those with acquired HLH also develop transaminitis and in experimental models of sepsis, where high levels of hyperbilirubinemia [104–106]. To differentiate between cytokines were generated, revealed severe impairment the terms ‘liver dysfunction’ and ‘liver failure’, the use of hepatocellular transport, specifically at the canalicu- of organ dysfunction scores have been advocated [107]. lar pole [89]. Release of pro-inflammatory cytokines by There is no single marker or a combination of markers Kupffer cells could lead to downregulation of transporters that reflects the entire spectrum of hepatic dysfunction or involved in bile flow [90]. In the setting of COVID-19, could predict a mortality rate of over 50% [108]. Skeletal however, the cholestatic pattern of liver injury is uncom- muscles, heart, lung, brain, kidney and other organs may mon [91]. Most patients present with elevated ami- release AST, and so serum ALT may better reflect hepatic notransferase levels with a predominant AST elevation dysfunction [109]. However, it is generally accepted that [3]. As such, liver injury related to COVID-19 although during liver injury, hepatocellular permeability is increased akin to sepsis with significant proinflammatory cytokine with consequent release of AST and ALT into the plasma production may have alternate explanations. [109]. Metanalytic studies have shown that liver dysfunc- Acquired hemophagocytic lymphohistiocytosis and liver tion in COVID-19 is not uncommon [110]. In a study by Chen et al., out of 99 cases diagnosed with COVID-19, injury 43.4% had increased serum levels of ALT and AST [111]. HLH is a catastrophic hyperinflammatory syndrome In another study from Wuhan, among 305 patients with occurring in genetically susceptible individuals resulting COVID-19, 39.1% had transaminitis or hyperbilirubine- from an exaggerated immune response [92]. Although mia at admission [112]. The mean levels of serum ALT,

Copyright © 2020 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited. Cytokine-induced liver injury in coronavirus disease-2019 Anirvan et al. www.eurojgh.com 5

Fig. 1. The figure illustrates the pathophysiologic mechanisms of liver injury in COVID-19.

AST and bilirubin were also significantly higher in patients Acknowledgements with more severe disease [113]. In a meta-analysis of 20 S.K.S. and S.P.S. conceptualized, wrote, reviewed and retrospective studies, higher serum levels of AST and ALT edited the article. P.A. prepared the original draft. S.N., were found to be associated with a significant increase in N.H. and F.Z. reviewed and edited the article. F.Z. is the severity of COVID-19 infection [114]. Additionally, responsible for figure illustration. All authors participated AST-dominant aminotransferase elevation has been found in providing important intellectual inputs, participated in to be common in COVID-19, mirrors disease severity and critical revision of the article and approved the final ver- appears to reflect true hepatic injury [115]. sion of the manuscript. This work was partially supported by a grant from the Kalinga Gastroenterology Foundation, Conclusion Cuttack, India. The pathogenesis of cytokine-mediated liver injury in the setting of COVID-19 is poorly understood, but sev- Conflicts of interest eral intriguing observations point towards the complex S.K.S. has served as a speaker for Intercept, Alexion, pathophysiological process mediated by inflammation, Dova, as an advisory board member for Gilead, Intercept, altered coagulation and RAAS activation culminating Bayer, and has received research funding from Gilead, in microvascular insult, hepatocyte damage and per- Biotest, Genfit, Conatus, Intercept, Shire, Exact Sciences, petuation of inflammation. Although transaminitis has Eananta, Dova, Bayer. S.K.S. is an employee of Northwell been reported in more than half of the patients, hyper- Health. For the remaining authors, there are no conflicts bilirubinemia and elevated alkaline phosphatase are of interest. uncommon. Liver injury probably is, perhaps, a part of dysregulated immune response in COVID-19 [116]. References Therapeutic strategies should target the immune system, RAAS and coagulation pathways synergistically. A crit- 1 Zaim S, Chong JH, Sankaranarayanan V, Harky A. COVID-19 and mul- ti-organ response. Curr Probl Cardiol 2020; 45:100618. ical appraisal of hepatic injury in the setting of CRSs 2 Varga Z, Flammer AJ, Steiger P, Haberecker M, Andermatt R, would be crucial to our understanding of liver dysfunc- Zinkernagel AS, et al. Endothelial cell infection and endotheliitis in tion in COVID-19. COVID-19. Lancet 2020; 395:1417–1418.

Copyright © 2020 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited. 6 European Journal of Gastroenterology & Hepatology xxx 2020 • Volume XXX • Number XXX

3 Richardson S, Hirsch JS, Narasimhan M, Crawford JM, McGinn T, protective effects and biphasic changes in blood cytokine levels. Eur J Davidson KW, et al. Presenting characteristics, comorbidities, and Immunol 1993; 23:2209–2216. outcomes among 5700 patients hospitalized with COVID-19 in the 29 Tanaka T, Narazaki M, Kishimoto T. Immunotherapeutic implications of New York City Area. JAMA 2020; 323:2052–2059. IL-6 blockade for cytokine storm. Immunotherapy 2016; 8:959–970. 4 Garrido I, Liberal R, Macedo G. Review article: COVID-19 and liver 30 Groom JR, Luster AD. CXCR3 ligands: redundant, collaborative and disease—what we know on 1st May 2020. Aliment Pharmacol Ther antagonistic functions. Immunol Cell Biol 2011; 89:207–215. 2020; 52:267–275. 31 Zangrillo A, Beretta L, Scandroglio AM, Monti G, Fominskiy E, Colombo 5 Cohen MC, Cohen S. Cytokine function: a study in biologic diversity. S, et al.; COVID-BioB Study Group. Characteristics, treatment, Am J Clin Pathol 1996; 105:589–598. outcomes and cause of death of invasively ventilated patients with 6 Tisoncik JR, Korth MJ, Simmons CP, Farrar J, Martin TR, Katze MG. COVID-19 ARDS in Milan, Italy. Crit Care Resusc 2020; 22:200–211. Into the eye of the cytokine storm. Microbiol Mol Biol Rev 2012; 32 Coperchini F, Chiovato L, Croce L, Magri F, Rotondi M. The cytokine 76:16–32. storm in COVID-19: an overview of the involvement of the chemok- 7 Kawai T, Akira S. The roles of TLRs, RLRs and NLRs in pathogen ine/chemokine-receptor system. Cytokine Growth Factor Rev 2020; recognition. Int Immunol 2009; 21:317–337. 53:25–32. 8 Kumar H, Kawai T, Akira S. Pathogen recognition by the innate 33 Sun Y, Jin C, Zhan F, Wang X, Liang M, Zhang Q, et al. Host cytokine immune system. Int Rev Immunol 2011; 30:16–34. storm is associated with disease severity of severe fever with throm- 9 Kimura H, Yoshizumi M, Ishii H, Oishi K, Ryo A. Cytokine production bocytopenia syndrome. J Infect Dis 2012; 206:1085–1094. and signaling pathways in respiratory virus infection. Front Microbiol 34 Chen P, Nirula A, Heller B, Gottlieb RL, Boscia J, Morris J, et al. SARS- 2013; 4:276. CoV-2 neutralizing antibody LY-CoV555 in outpatients with Covid-19. 10 Qin L, Hu CP, Feng JT, Xia Q. Activation of lymphocytes induced by New England J Med 2020. doi: 10.1056/NEJMoa2029849. [Epub bronchial epithelial cells with prolonged RSV infection. PLoS One ahead of print] 2011; 6:e27113. 35 Azar WS, Njeim R, Fares AH, Azar NS, Azar ST, El Sayed M, Eid AA. 11 Borden EC, Sen GC, Uze G, Silverman RH, Ransohoff RM, Foster GR, COVID-19 and diabetes mellitus: how one pandemic worsens the Stark GR. Interferons at age 50: past, current and future impact on other. Rev Endocr Metab Disord 2020; 21:451–463. biomedicine. Nat Rev Drug Discov 2007; 6:975–990. 36 Cohen J. The immunopathogenesis of sepsis. Nature 2002; 12 Quint JK, Donaldson GC, Goldring JJ, Baghai-Ravary R, Hurst JR, 420:885–891. Wedzicha JA. Serum IP-10 as a biomarker of human rhinovirus infec- 37 Monneret G, Lepape A, Voirin N, Bohé J, Venet F, Debard AL, et tion at exacerbation of COPD. Chest 2010; 137:812–822. al. Persisting low human leukocyte antigen-DR expres- 13 Peiris J, Chu C, Cheng V, Chan KS, Hung IF, Poon LL, et al. Clinical sion predicts mortality in septic shock. Intensive Care Med 2006; progression and viral load in a community outbreak of coronavi- 32:1175–1183. rus-associated SARS pneumonia: a prospective study. Lancet. 2003; 38 Yang Y, Shen C, Li J, Yuan J, Yang M, Wang F, et al. Exuberant 361:1767–1772. elevation of IP-10, MCP-3 and IL-1ra during SARS-CoV-2 infec- 14 Fehr AR, Perlman S. Coronaviruses: an overview of their replication tion is associated with disease severity and fatal outcome. medRxiv and pathogenesis. Methods Mol Biol. 2015; 1282:1–23. 2020:2020.03.02.20029975. doi:10.1101/2020.03.02.20029975 15 Wang FI, Stohlman SA, Fleming JO. Demyelination induced by murine 39 Xiong Y, Liu Y, Cao L, Wang D, Guo M, Jiang A, et al. Transcriptomic hepatitis virus JHM strain (MHV-4) is immunologically mediated. J characteristics of bronchoalveolar lavage fluid and peripheral blood Neuroimmunol 1990; 30:31–41. mononuclear cells in COVID-19 patients. Emerg Microbes Infect 2020; 16 Sheng WH, Chiang BL, Chang SC, Ho HN, Wang JT, Chen YC, et 9:761–770. al. Clinical manifestations and inflammatory cytokine responses 40 Liao YC, Liang WG, Chen FW, Hsu JH, Yang JJ, Chang MS. IL-19 in patients with severe acute respiratory syndrome. J Formos Med induces production of IL-6 and TNF-alpha and results in cell apoptosis Assoc 2005; 104:715–723. through TNF-alpha. J Immunol 2002; 169:4288–4297. 17 Kim ES, Choe PG, Park WB, Oh HS, Kim EJ, Nam EY, et al. Clinical 41 Aggarwal S, Gollapudi S, Gupta S. Increased TNF-alpha-induced progression and cytokine profiles of middle east respiratory syndrome apoptosis in lymphocytes from aged humans: changes in TNF-alpha coronavirus infection. J Korean Med Sci 2016; 31:1717–1725. receptor expression and activation of caspases. J Immunol 1999; 18 Huang C, Wang Y, Li X, Ren L, Zhao J, Hu Y, et al. Clinical features of 162:2154–2161. patients infected with 2019 novel coronavirus in Wuhan, China. Lancet 42 Li L, Li S, Xu M, Yu P, Zheng S, Duan Z, et al. Risk factors related 2020; 395:497–506. to hepatic injury in patients with corona virus disease 2019. medRxiv 19 Henry BM, Benoit SW, de Oliveira MHS, Hsieh WC, Benoit J, Ballout 2020:2020.02.28.20028514. doi:10.1101/2020.02.28.20028514 RA, et al. Laboratory abnormalities in children with mild and severe 43 Kim KD, Zhao J, Auh S, Yang X, Du P, Tang H, Fu YX. Adaptive coronavirus disease 2019 (COVID-19): a pooled analysis and review. immune cells temper initial innate responses. Nat Med 2007; 13:1248– Clin Biochem 2020; 81:1–8. 1252. 20 Wu C, Chen X, Cai Y, Xia J, Zhou X, Xu S, et al. Risk factors associated 44 Liu J, Li S, Liu J, Liang B, Wang X, Wang H et al. Longitudinal with acute respiratory distress syndrome and death in patients with characteristics of lymphocyte responses and cytokine profiles in coronavirus disease 2019 pneumonia in Wuhan, China. JAMA Intern the peripheral blood of SARS-CoV-2 infected patients. MedRxiv Med 2020; 180:934–943. 2020:2020.02.16.20023671. doi:10.1101/2020.02.16.20023671 21 Phipps MM, Barraza LH, LaSota ED, Sobieszczyk ME, Pereira MR, 45 Shimabukuro-Vornhagen A, Gödel P, Subklewe M, Stemmler HJ, Zheng EX, et al. Acute liver injury in Covid-19: prevalence and asso- Schlößer HA, Schlaak M, et al. Cytokine release syndrome. J ciation with clinical outcomes in a large US cohort. Hepatology 2020. Immunother Cancer 2018; 6:56. doi: 10.1002/hep.31404. [Epub ahead of print] 46 Neelapu SS, Tummala S, Kebriaei P, Wierda W, Gutierrez C, Locke 22 Fan Z, Chen L, Li J, Cheng X, Yang J, Tian C, et al. Clinical features FL, et al. Chimeric antigen receptor T-cell therapy - assessment and of COVID-19-related liver functional abnormality. Clin Gastroenterol management of toxicities. Nat Rev Clin Oncol 2018; 15:47–62. Hepatol 2020; 18:1561–1566. 47 Kantarjian H, Stein A, Gökbuget N, Fielding AK, Schuh AC, Ribera 23 Wong CK, Lam CW, Wu AK, Ip WK, Lee NL, Chan IH, et al. Plasma JM, et al. Blinatumomab versus chemotherapy for advanced acute inflammatory cytokines and chemokines in severe acute respiratory lymphoblastic leukemia. N Engl J Med. 2017; 376:836–847. syndrome. Clin Exp Immunol 2004; 136:95–103. 48 Edwards L, Wanless IR. Mechanisms of liver involvement in systemic 24 Mahallawi WH, Khabour OF, Zhang Q, Makhdoum HM, Suliman BA. disease. Best Pract Res Clin Gastroenterol 2013; 27:471–483. MERS-CoV infection in humans is associated with a pro-inflammatory 49 Sprague AH, Khalil RA. Inflammatory cytokines in vascular dysfunction Th1 and Th17 cytokine profile. Cytokine 2018; 104:8–13. and vascular disease. Biochem Pharmacol 2009; 78:539–552. 25 Teijaro JR. Cytokine storms in infectious diseases. Semin 50 Paria BC, Vogel SM, Ahmmed GU, Alamgir S, Shroff J, Malik AB, Immunopathol 2017; 39:501–503. Tiruppathi C. Tumor necrosis factor-alpha-induced TRPC1 expression 2+ 26 Yuen K, Wong S. Human infection by avian influenza A 5H N1. Hong amplifies store-operated Ca influx and endothelial permeability. Am Kong Med J 2005; 11:189–199. J Physiol Lung Cell Mol Physiol 2004; 287:L1303–L1313. 27 Ye Q, Wang B, Mao J. The pathogenesis and treatment of the 51 Gao X, Belmadani S, Picchi A, Xu X, Potter BJ, Tewari-Singh N, et `Cytokine Storm’ in COVID-19. J Infect 2020; 80:607–613. al. Tumor necrosis factor-alpha induces endothelial dysfunction in 28 Matthys P, Dillen C, Proost P, Heremans H, Van Damme J, Billiau Lepr(db) mice. Circulation 2007; 115:245–254. A. Modification of the anti-CD3-induced cytokine release syndrome 52 Yang D, Elner SG, Bian ZM, Till GO, Petty HR, Elner VM. Pro- by anti-interferon-gamma or anti-interleukin-6 antibody treatment: inflammatory cytokines increase reactive oxygen species through

Copyright © 2020 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited. Cytokine-induced liver injury in coronavirus disease-2019 Anirvan et al. www.eurojgh.com 7

mitochondria and NADPH oxidase in cultured RPE cells. Exp Eye Res 78 Nguyen-Lefebvre AT, Horuzsko A. Kupffer cell metabolism and func- 2007; 85:462–472. tion. J Enzymol Metab 2015; 1:101. https://www.ncbi.nlm.nih.gov/ 53 Chihara R, Nakamoto H, Arima H, Moriwaki K, Kanno Y, Sugahara S, pmc/articles/PMC4771376/. [Accessed 23 July 2020]. et al. Systemic capillary leak syndrome. Intern Med 2002; 41:953–956. 79 Jaeschke H, Farhood A, Cai SX, Tseng BY, Bajt ML. Protection against 54 Duan CY, Zhang J, Wu HL, Li T, Liu LM. Regulatory mechanisms, TNF-induced liver parenchymal cell apoptosis during endotoxemia prophylaxis and treatment of vascular leakage following severe trauma by a novel caspase inhibitor in mice. Toxicol Appl Pharmacol 2000; and shock. Mil Med Res 2017; 4:11. 169:77–83. 55 Hiltebrand LB, Krejci V, Banic A, Erni D, Wheatley AM, Sigurdsson 80 Spapen H. Liver perfusion in sepsis, septic shock, and multiorgan fail- GH. Dynamic study of the distribution of microcirculatory blood flow ure. Anat Rec (Hoboken) 2008; 291:714–720. in multiple splanchnic organs in septic shock. Crit Care Med 2000; 81 Xu Z, Shi L, Wang Y, Zhang J, Huang L, Zhang C, et al. Pathological 28:3233–3241. findings of COVID-19 associated with acute respiratory distress syn- 56 Green SJ. Covid-19 accelerates endothelial dysfunction and nitric drome. Lancet Respir Med 2020; 8:420–422. oxide deficiency. Microbes Infect 2020; 22:149–150. 82 Liu Q, Wang RS, Qu GQ, Wang YY, Liu P, Zhu YZ, et al. Gross exam- 57 Hoffmann M, Kleine-Weber H, Schroeder S, Krüger N, Herrler T, ination report of a COVID-19 death autopsy. Fa Yi Xue Za Zhi 2020; Erichsen S, et al. SARS-CoV-2 cell entry depends on ACE2 and 36:21–23. TMPRSS2 and is blocked by a clinically proven protease inhibitor. Cell 83 Rapkiewicz AV, Mai X, Carsons SE, Pittaluga S, Kleiner DE, Berger 2020; 181:271–280.e8. JS, et al. Megakaryocytes and platelet-fibrin thrombi character- 58 Rabelo LA, Todiras M, Nunes-Souza V, Qadri F, Szijártó IA, Gollasch ize multi-organ thrombosis at autopsy in COVID-19: a case series. M, et al. Genetic deletion of ACE2 induces vascular dysfunction in Eclinicalmedicine 2020; 24:100434. c57bl/6 mice: role of nitric oxide imbalance and oxidative stress. PLoS 84 Song JC, Wang G, Zhang W, Zhang Y, Li WQ, Zhou Z; People’s One 2016; 11:e0150255. Liberation Army Professional Committee of Critical Care Medicine, 59 Vlahos R, Stambas J, Selemidis S. Suppressing production of reactive Chinese Society on Thrombosis and Haemostasis. Chinese expert oxygen species (ROS) for influenza A virus therapy. Trends Pharmacol consensus on diagnosis and treatment of coagulation dysfunction in Sci 2012; 33:3–8. COVID-19. Mil Med Res 2020; 7:19. 60 Zhong M, Sun A, Xiao T, Yao G, Sang L, Zheng X, et al. A rand- 85 Tian S, Hu W, Niu L, Liu H, Xu H, Xiao S-Y. Pulmonary pathology omized, single-blind, group sequential, active-controlled study to of early-phase 2019 novel coronavirus (COVID-19) pneumonia in two evaluate the clinical efficacy and safety ofα -Lipoic acid for critically patients with lung cancer. J Thorac Oncol 2020; 15:700–704. ill patients with coronavirus disease 2019 (COVID-19). medRxiv 86 Ferrario CM, Jessup J, Chappell MC, Averill DB, Brosnihan KB, Tallant 2020:2020.04.15.20066266. doi:10.1101/2020.04.15.20066266 EA, et al. Effect of angiotensin-converting enzyme inhibition and angi- 61 Bahloul M, Ketata W, Lahyeni D, Mayoufi H, Kotti A, Smaoui F,et al. otensin ii receptor blockers on cardiac angiotensin-converting enzyme Pulmonary capillary leak syndrome following COVID-19 virus infection. 2. Circulation 2005; 111:2605–2610. J Med Virol 2020. 87 Salerno M, Sessa F, Piscopo A, Montana A, Torrisi M, Patanè F, et 62 Genctoy G, Altun B, Kiykim AA, Arici M, Erdem Y, Cağlarg M, et al. al. No autopsies on COVID-19 deaths: a missed opportunity and the TNF alpha-308 genotype and renin-angiotensin system in hemodial- lockdown of science. J Clin Med 2020; 9:1472. ysis patients: an effect on inflammatory cytokine levels? Artif Organs 88 Hippensteel JA, LaRiviere WB, Colbert JF, Langouët-Astrié CJ, 2005; 29:174–178. Schmidt EP. Heparin as a therapy for COVID-19: current evidence 63 Wang R, Alam G, Zagariya A, Gidea C, Pinillos H, Lalude O, et al. and future possibilities. Am J Physiol Lung Cell Mol Physiol 2020; Apoptosis of lung epithelial cells in response to TNF-alpha requires 319:L211–L217. angiotensin II generation de novo. J Cell Physiol 2000; 185:253–259. 89 Gonnert FA, Recknagel P, Hilger I, Claus RA, Bauer M, Kortgen A. 64 Benigni A, Cassis P, Remuzzi G. Angiotensin II revisited: new roles Hepatic excretory function in sepsis: implications from biophotonic in inflammation, and aging. EMBO Mol Med 2010; analysis of transcellular xenobiotic transport in a rodent model. Crit 2:247–257. Care 2013; 17:R67. 65 Ahmed S, Anirvan P. Reply to Rheumatologists’ perspective on coro- 90 Bhogal HK, Sanyal AJ. The molecular pathogenesis of cholestasis in navirus disease 19: is heparin the dark horse for COVID-19? Clin sepsis. Front Biosci (Elite Ed) 2013; 5:87–96. Rheumatol 2020; 39:2099–2100. 91 Omrani-Nava V, Maleki I, Ahmadi A, et al. Evaluation of hepatic 66 Wang K, Gheblawi M, Oudit GY. Angiotensin converting enzyme 2: a enzymes changes and association with prognosis in COVID-19 double-edged sword. Circulation 2020; 142:426–428. patients. Hepat Mon 2020; 20. 67 Vaughan DE, Lazos SA, Tong K. Angiotensin II regulates the expres- 92 Rosado FG, Kim AS. Hemophagocytic lymphohistiocytosis: an update sion of plasminogen activator inhibitor-1 in cultured endothelial cells. A on diagnosis and pathogenesis. Am J Clin Pathol 2013; 139:713–727. potential link between the renin-angiotensin system and thrombosis. J 93 Kleynberg RL, Schiller GJ. Secondary hemophagocytic lymphohis- Clin Invest 1995; 95:995–1001. tiocytosis in adults: an update on diagnosis and therapy. Clin Adv 68 Imai Y, Kuba K, Ohto-Nakanishi T, Penninger JM. Angiotensin- Hematol Oncol 2012; 10:726–732. converting enzyme 2 (ACE2) in disease pathogenesis. Circ J 2010; 94 Janka G. Hemophagocytic lymphohistiocytosis: when the immune 74:405–410. system runs amok. Klin Padiatr 2009; 221:278–285. 69 Numaguchi Y, Ishii M, Kubota R, Morita Y, Yamamoto K, Matsushita 95 Padhi S, Sarangi R, Patra S, Samal SC. Hepatic involvement in hemo- T, et al. Ablation of angiotensin IV receptor attenuates hypofibrinolysis phagocytic lymphohistiocytosis. In: Streba CT, Vere CC, Rogoveanu via PAI-1 downregulation and reduces occlusive arterial thrombosis. I, Tripodi V, Lucangioli S, editors. Hepatitis A and Other Associated Arterioscler Thromb Vasc Biol 2009; 29:2102–2108. Hepatobiliary Diseases. Rijeka: IntechOpen; 2020. doi: 10.5772/ 70 Gromotowicz-Poplawska A, Marcinczyk N, Misztal T, Golaszewska A, intechopen.90238. Aleksiejczuk M, Rusak T, Chabielska E. Rapid effects of aldosterone 96 McGonagle D, Sharif K, O’Regan A, Bridgewood C. The role of on platelets, coagulation, and fibrinolysis lead to experimental throm- cytokines including interleukin-6 in COVID-19 induced pneumonia and bosis augmentation. Vascul Pharmacol 2019; 122–123:106598. macrophage activation syndrome-like disease. Autoimmun Rev 2020; 71 Simões e Silva AC, Miranda AS, Rocha NP, Teixeira AL. Renin angi- 19:102537. otensin system in liver diseases: friend or foe? World J Gastroenterol 97 Swoboda J, Wittschieber D, Sanft J, Kleemann S, Elschner S, Ihle 2017; 23:3396–3406. H, et al. Bone marrow haemophagocytosis designate severe SARS- 72 Taskin E, Guven C. Local renin-angiotensin system at liver and cross- CoV-2 infections. Histopathology 2020. talk with hepatic diseases. Renin-Angiotensin System - Past, Present 98 Rosário C, Zandman-Goddard G, Meyron-Holtz EG, D’Cruz DP, and Future 2017. Shoenfeld Y. The hyperferritinemic syndrome: macrophage activation 73 Jahr H, Wolff H. [The liver–an immunologically privileged organ?]. syndrome, Still’s disease, septic shock and catastrophic antiphospho- Allerg Immunol (Leipz) 1989; 35:155–166. lipid syndrome. BMC Med 2013; 11:1–11. 74 Robinson MW, Harmon C, O’Farrelly C. Liver immunology and its role in 99 Bracaglia C, Prencipe G, De Benedetti F. Macrophage activation inflammation and homeostasis. Cell Mol Immunol 2016; 13:267–276. syndrome: different mechanisms leading to a one clinical syndrome. 75 Gao B. Basic liver immunology. Cell Mol Immunol 2016; 13:265–266. Pediatr Rheumatol Online J 2017; 15:5. 76 Moshage H. Cytokines and the hepatic acute phase response. J 100 Wan S, Yi Q, Fan S, Lv J, Zhang X, Guo L, et al. Characteristics of lym- Pathol 1997; 181:257–266. phocyte subsets and cytokines in peripheral blood of 123 hospitalized 77 Wang D, Yin Y, Yao Y. Advances in sepsis-associated liver dysfunction. patients with 2019 novel coronavirus pneumonia (NCP). Hematology Burns Trauma 2014; 2:97–105. 2020.

Copyright © 2020 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited. 8 European Journal of Gastroenterology & Hepatology xxx 2020 • Volume XXX • Number XXX

101 Fuhrmann V, Kneidinger N, Herkner H, Heinz G, Nikfardjam M, Bojic A, 108 Marshall JC, Cook DJ, Christou NV, Bernard GR, Sprung CL, Sibbald et al. Hypoxic hepatitis: underlying conditions and risk factors for mor- WJ. Multiple organ dysfunction score: a reliable descriptor of a com- tality in critically ill patients. Intensive Care Med 2009; 35:1397–1405. plex clinical outcome. Crit Care Med 1995; 23:1638–1652. 102 Kramer L, Jordan B, Druml W, Bauer P, Metnitz PG; Austrian 109 Lescot T, Karvellas C, Beaussier M, Magder S. Acquired liver injury in Epidemiologic Study on Intensive Care, ASDI Study Group. Incidence the intensive care unit. Anesthesiology 2012; 117:898–904. and prognosis of early hepatic dysfunction in critically ill patients–a 110 Mao R, Qiu Y, He J-S, Tan JY, Li XH, Liang J, et al. Manifestations prospective multicenter study. Crit Care Med 2007; 35:1099– and prognosis of gastrointestinal and liver involvement in patients 1104. with COVID-19: a systematic review and meta-analysis. Lancet 103 Bakker J, Grover R, McLuckie A, Holzapfel L, Andersson J, Lodato Gastroenterol Hepatol 2020; 5:667–678. R, et al.; Glaxo Wellcome International Septic Shock Study Group. 111 Chen N, Zhou M, Dong X, Qu J, Gong F, Han Y, et al. Epidemiological Administration of the nitric oxide synthase inhibitor NG-methyl-L- and clinical characteristics of 99 cases of 2019 novel coronavi- arginine hydrochloride (546C88) by intravenous infusion for up to 72 rus pneumonia in Wuhan, China: a descriptive study. Lancet 2020; hours can promote the resolution of shock in patients with severe sep- 395:507–513. sis: results of a randomized, double-blind, placebo-controlled multi- 112 Dan F, Jingdong M, Jialun G, et al. Manifestations of Digestive system in center study (study no. 144-002). Crit Care Med 2004; 32:1–12. hospitalized patients with novel coronavirus pneumonia in Wuhan, China: 104 Nägele V, Kratzer A, Zugmaier G, Holland C, Hijazi Y, Topp MS, a single-center, descriptive study. Chin J Dig 2020; 40:E005–E005. et al. Changes in clinical laboratory parameters and pharma- 113 Liu C, Jiang ZC, Shao CX, Zhang HG, Yue HM, Chen ZH, et al. codynamic markers in response to blinatumomab treatment of [Preliminary study of the relationship between novel coronavirus pneu- patients with relapsed/refractory ALL. Exp Hematol Oncol 2017; monia and liver function damage: a multicenter study]. Zhonghua Gan 6:14. Zang Bing Za Zhi 2020; 28:107–111. 105 Lutfi F, Patel A, Becker D, Shahid M, Shah K. Hemophagocytic lym- 114 Parohan M, Yaghoubi S, Seraj A. Liver injury is associated with phohistiocytosis (HLH) presenting as fever of unknown origin and severe Coronavirus disease 2019 (COVID-19) infection: a systematic acute liver failure. IDCases 2018; 14:e00413. review and meta-analysis of retrospective studies. Hepatol Res 2020; 106 Lin S, Li Y, Long J, Liu Q, Yang F, He Y. Acute liver failure caused 50:924–935. by hemophagocytic lymphohistiocytosis in adults: a case report and 115 Bloom PP, Meyerowitz EA, Reinus Z, Daidone M, Gustafson J, Kim review of the literature. Medicine (Baltimore) 2016; 95:e5431. AY, et al. Liver biochemistries in hospitalized patients with COVID-19. 107 Brun-Buisson C, Meshaka P, Pinton P, Vallet B; EPISEPSIS Study Hepatology 2020. Group. EPISEPSIS: a reappraisal of the epidemiology and outcome 116 Thomson SJ, Cowan ML, Johnston I, Musa S, Grounds M, Rahman of severe sepsis in French intensive care units. Intensive Care Med. TM. ‘Liver function tests’ on the intensive care unit: a prospective, 2004; 30:580–588. observational study. Intensive Care Med 2009; 35:1406–1411.

Copyright © 2020 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.