Next Article in Journal
Exploring Potential Determinants of Mosquito Net Ownership and Use in Somalia: A Cross-Sectional Study
Previous Article in Journal
Psychophysiological Adaptations to Yoga Practice in Overweight and Obese Individuals: A Topical Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Nutritional Support in Acute Liver Failure

by
Ludovico Abenavoli
1,*,
Valentina Maurizi
2,
Luigi Boccuto
3,
Arianna Di Berardino
4,
Nena Giostra
4,
Pierangelo Santori
5,
Maria Laura Scarcella
6,
Anna Caterina Procopio
1,
Carlo Rasetti
4,5 and
Emidio Scarpellini
7
1
Department of Health Sciences, University “Magna Græcia”, 88100 Catanzaro, Italy
2
Internal Medicine Residency Program, Università Politecnica delle Marche, 60121 Ancona, Italy
3
Healthcare Genetics and Genomics Doctoral Program, School of Nursing, College of Behavioral, Social and Health Sciences, Clemson University, Clemson, SC 29634, USA
4
Clinical Nutrition and Internal Medicine Unit, “Madonna del Soccorso” General Hospital, 63074 San Benedetto del Tronto, Italy
5
Hepatology and Internal Medicine Unit, “Madonna del Soccorso” General Hospital, 63074 San Benedetto del Tronto, Italy
6
Anesthesia, Intensive Care and Nutritional Science, Azienda Ospedaliera “Santa Maria”, Via Tristano di Joannuccio, 05100 Terni, Italy
7
T.A.R.G.I.D., Gasthuisberg University Hospital, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
*
Author to whom correspondence should be addressed.
Diseases 2022, 10(4), 108; https://doi.org/10.3390/diseases10040108
Submission received: 5 September 2022 / Revised: 14 October 2022 / Accepted: 16 November 2022 / Published: 18 November 2022
(This article belongs to the Section Gastroenterology)

Abstract

:
Acute liver failure (ALF) presents with an acute abnormality of liver blood tests in an individual without underlying chronic liver disease. The clinical course leads to the development of coagulopathy and hepatic encephalopathy. The role of nutrition in its prevention and treatment remains uncertain. We aimed to review literature data on the concept of ALF and the role of nutrition in its treatment and prevention, considering the impact of gut microbiota dysbiosis and eubiosis. We conducted a review of the literature on the main medical databases using the following keywords and acronyms and their associations: liver failure, nutrition, branched-chain amino acids, gut microbiota, dysbiosis, and probiotics. Upon their arrival at the emergency department, an early, accurate nutritional assessment is crucial for individuals with ALF. Branched-chain amino acids (BCAAs), stable euglycemia maintenance, and moderate caloric support are crucial for this subset of patients. An excessive protein load must be avoided because it worsens hepatic encephalopathy. Preclinical evidence supports future probiotics use for ALF treatment/prevention. Nutritional support and treatment for ALF are crucial steps against patient morbidity and mortality. BCAAs and euglycemia remain the mainstay of nutritional treatment of ALF. Gut dysbiosis re-modulation has an emerging and natural-history changing impact on ALF.

1. Introduction

Acute liver failure (ALF) is a rare and challenging condition involving very often the emergency department (ER) team and, therefore, either the intensive care unit (ICU) or the mild-intensity hepatology clinic [1]. Typically, patients present with unexplained symptoms/signs of hepatic encephalopathy and coagulopathy in the blood test analyses [2]. It is also worth mentioning that the patients can be adults or children [3]. Importantly, these patients presenting with desperate clinical conditions do not have a pre-existing liver disease and/or cirrhosis [4,5]. Anamnestic investigation can help physicians recognize the aetiologic agent of acute liver injury (ALI) [6]. For example, the use and—very frequently—abuse of medications such as the over-the-counter acetaminophen, whose cut-off dosage is narrow and variable among people, is one of the most common etiologies of ALI leading to ALF. The prognosis of ALF can be extremely poor and require liver transplantation [7,8]. Malnutrition is a common condition in ALF and is an independent prognostic factor for morbidity and mortality. The deficiency of vitamins, minerals, glucose, and amino acids must be assessed and eventually addressed very early in the frame of patient management [9]. However, liver inflammation and consequent destruction open up the tight junctions maintaining a “vigil” intestinal permeability and release damage-associated molecular patterns (DAMPs), such as Gram-negative lipopolysaccharide, which can pass through the portal vein and perpetuate liver damage until necrosis via oxidative stress. This is an example of a disrupted gut–liver axis and is the pathophysiological loop explaining the pathogenesis of several chronic liver diseases such as liver steatosis (NAFLD), non-alcoholic steatohepatitis (NASH), liver cirrhosis, and hepatocellular carcinoma [10]. Within this pathophysiological model, an emerging actor is gaining increasing attention among researchers: gut microbiota, whose qualitative and quantitative composition is crucial for host homeostasis [11]. Its dysbiosis is responsible for several pathologic conditions involving liver inflammation and fibrosis, including ALF. For these reasons, we aimed to review literature data regarding the definition of ALF and the role of nutrition in its treatment and prevention, with a special focus on gut microbiota dysbiosis and its pathophysiological impact and potential therapeutic application.

2. Materials and Methods

We conducted a PubMed and Medline search for original articles, reviews, meta-analyses, and case series using the following keywords, their acronyms, and associations: liver failure, nutrition, branched-chain amino acids, gut microbiota, dysbiosis, and probiotics. When appropriate, we also included preliminary evidence from abstracts belonging to main national and international gastroenterological meetings. The papers found from the above-mentioned sources were reviewed by two of the authors (ES and VM) according to PRISMA guidelines [12]. The last Medline search was conducted on 30 June 2022.
In detail, we found 262 manuscripts matching our search: 52 were clinical trials/original articles, and of these, 24 were randomized clinical trials (RCT)s, in addition to 107 reviews of the scientific literature, 20 of them systematic reviews and/or metanalyses. In detail, we included data from 74 papers: 20 original articles (including in vitro and animal studies); 12 RCTs; 11 systematic reviews of the scientific literature, excluding those not updated; and 9 metanalyses.

3. Results

3.1. Acute Liver Failure: Definition and Clinical “Scenarios”

ALF is a rare syndrome characterized by an acute abnormality of liver blood tests, due to a primary liver insult, in subjects without underlying chronic liver disease. ALF’s main features are coagulopathy and hepatic encephalopathy. Of importance, patients who develop coagulopathy without encephalopathy have an ALI [13]. From a wider point of view, ALF clinical presentation includes non-specific mild symptoms (e.g., fatigue, malaise, anorexia, nausea, and/or vomiting) and, rising in gravity, abdominal pain, jaundice and itch, ascites, mental confusion, agitation, and coma because of cerebral edema. The development of multiorgan failure (MOF) is not uncommon. In detail, hepatic encephalopathy manifestations range from changes in behavior to coma. They have five grades according to the West Haven classification [14]. At grade 0, patients have no changes in personality/behavior; there is just mild loss of memory, concentration, and coordination. Asterixis is the hallmark of grade I, together with deranged orientation and sleep disturbances (hypersomnia or insomnia); patients start to fail neuropsychological tests also because of decreased attention. These patients can also show mood alterations (both depression and euphoria). In grade II, lethargy and/or apathy are characteristic symptoms, although personality changes toward irritability can also be present. In grade III, severe confusion until reversible sleeping and gross disorientation are present. In the fourth grade, there is a coma [15,16]. Intriguingly, “overt” encephalopathy is another syndromic association, difficult to diagnose, because it is characterized by marked alteration of mental status, asterixis, hyperreflexia, and hypertonicity that can easily evolve to coma and exitus [3,4,5,17]. Importantly, since initial mental impairment may be subtle, the identification of hepatic encephalopathy in a patient without subsiding cirrhosis and/or preexisting liver disease requires an accurate differential diagnosis. Coagulopathy, a criterion required for diagnosis, is defined by a prolongation of prothrombin time or prolongation of the international normalized ratio (INR) >1,5 [18]. The incidence of ALF ranges from one case per million people per year to about 2000–3000 cases per year in the USA [19]. However, in developing countries, the incidence is significantly higher [20]. Acute liver failure can result from a wide variety of causes (Table 1).
Acute viral hepatic damage remains the most common cause of ALF worldwide. However, it has sensibly declined in the European Union because of direct antiviral drugs used for HCV-related infections and HBV large-scale vaccination. Thus, drug-induced liver injury (DILI) is the main cause of ALF (e.g., from acetaminophen intoxication) in European countries. Moreover, autoimmune hepatitis, Budd–Chiari syndrome, and/or Wilson disease may have an acute onset with coagulopathy and encephalopathy and should be considered as causes of ALF in the absence of overt fibrosis and/or cirrhosis [21]. From a clinical point of view, O’Grady classified ALF as hyperacute, acute, and subacute according to encephalopathy onset when it develops within 7 days, 8–28 days, or 5–26 weeks, respectively, upon jaundice appearance [22]. Intriguingly, this classification was built up to be associated with the prognostic state. However, the associations reflected mainly the underlying causes as determinants of patient outcomes. For example, hyperacute presentation is more often related to acetaminophen or ischemic liver damage. Both of these causes can result in cerebral edema and/or extrahepatic MOF. On the other hand, they have a good outcome after medical treatment vs. acute or subacute ALF. The latter have a poorer outcome without a liver transplantation attempt. Indeed, the survival rate of patients with ALF has dramatically improved in the last few decades: it has gone from 20% to more than 60%. This radical change can be explained by the combination of the emerging practice of multidisciplinary medical teams and by the growing use of emergency liver transplantation (ELT) when needed [23]. As the medical management of patients with ALF in the ER, ICU, and/or mild intensity unit also requires the early treatment of the underlying causes of this condition and general supportive measures, appropriate nutritional support is crucial for patients’ morbidity and mortality.

3.2. Nutritional Support in Acute Liver Failure

Recent ESPEN guidelines outlined a strong relationship between liver diseases and nutritional status [24]. In detail, in ALF patients there is a dramatic loss of hepatocellular function and subsequent MOF. From a metabolic point of view, there is a severe impairment of carbohydrate, protein, and lipid metabolism. In detail, these patients present with reduced glucose synthesis and lactate elimination by the liver, protein catabolism associated with hyperaminoacidemia, and hyperammonemia [9]. Malnutrition has a variable prevalence in ALF patients, also according to its hyperacute, acute, or subacute course. Indeed, it is an independent risk factor for morbidity and mortality, such as in chronic liver diseases. More in detail, typical factors of ALF malnutrition depend on fat malabsorption and impaired gastric emptying [25]. Nutritional support in liver failure should optimize caloric, macronutrient, and micronutrient supplementation. In addition, nutritional support should save the remaining liver function with improved metabolic reserve. Altogether, these can reduce the risk of infection and improve the overall performance of patients in general and of those candidates for liver transplantation in particular [10,26]. Sarcopenia can be prevented just after an accurate assessment of nutritional status (e.g., bioimpedance, indirect calorimetry): intake of calories, protein, and essential amino acids has to be assessed and has to be maintained to stabilize muscle mass. Thus, although branched-chain amino acid (BCAA) supplementation is crucial for the management of hepatic encephalopathy and sarcopenia, L-leucine has been described as the main amino acid involved in protein turnover and is able to reverse the proteolytic process towards protein synthesis [27]. L-leucine belongs to the BCAA group (valine, leucine, and isoleucine). Almost 40% of the amino acids needed by mammals are BCAAs. Moreover, these BCAAs altogether or leucine alone can sustain protein synthesis and can inhibit protein breakdown [28].
From a wider hepatologic perspective, in healthy people in general and in patients with liver cirrhosis specifically, BCAA supplementation promotes anabolic pathways and reduces cachexia, prevents and/or treats hepatic encephalopathy, alleviates fatigue during exercise, promotes wound repair, and stimulates insulin production [29,30]. Going in more detail, hydroxy-beta-methyl butyrate (HMB) is an active metabolite of leucine, with anti-catabolic properties, used in several randomized controlled trials (RCTs) in patients with liver cirrhosis against sarcopenia and fat accumulation in skeletal muscles [31,32,33]. In addition, a systematic review and consequent meta-analysis of RCTs showed HMB and/or nutritional supplements containing HMB to increase skeletal muscle mass and promote its strength and hypertrophy/hyperplasia [34]. From a molecular point of view, the interaction of leucine and HMB activates the mTOR metabolic pathway, increasing protein translation through the phosphorylation of eukaryotic initiation factor 4E binding protein 1 (4E-BP1) and ribosomal protein S6 kinase (S6K), reaching muscle hyperplasia [35,36,37].
Within a detailed ALF focus, according to the European Association for the Study of the Liver, recommended nutritional support in liver failure, regarding BCAAs, the suggested doses are L-leucine 2500 mg, L-isoleucine 1250 mg, L-valine 1250 mg, HMB 1500 mg (namely, the therapeutic dosage), vitamin D 40 mcg (therapeutic dosage) or 1600 I.U., and vitamin K 60 and 400 micrograms. For prebiotics and B-vitamin complex, the recommended doses are about 25 milligrams of vitamin B1, B2, B6, and niacin; 400 to 800 micrograms of folic acid; and 25 micrograms of vitamin B12.
Enteral feeding is the preferred choice vs. parenteral nutrition in these patients. Oral nutrition should be encouraged but low-consciousness status due to rapid progressive hepatic encephalopathy or anorexia may impair this method of nutrient administration. Thus, nasogastric tube insertion is recommended, considering the risk of aspiration. For example, if delayed gastric emptying is present, feeding tubes can be placed post-pylorically. In addition, if a small-bowel failure occurs (e.g., due to ileus mechanical obstruction or ischemia), there is a higher rate of gut bacterial translocation and sepsis. Both events can be reduced by enteral feeding. Similarly to other ICU patients, total parenteral nutrition initiation is not recommended 5–7 days after ICU admission [38,39]. In the rare cases in which the parenteral route of administration has to be used, lipid emulsions are safe. Interestingly, SMOF lipids (SMOF) are newer preparations containing ω-3 and medium-chain triglycerides with higher patient performance vs. soy-based lipids [40]. Indeed, fat metabolism is regulated by active mitochondrial function, avoiding their potential accumulation and further liver damage [41]. Unfortunately, solid evidence in patients with ALF is lacking. Lipidic metabolism (triglycerides target >3 mmol/L or 265 mg/dL) has to be controlled along with creatinine kinase [40,41]. Caloric goals for enteral feeding in patients with ALF are driven by increased resting energy expenditure (between 18% and 30%) [42]. Hypoglycemia is a recurrent condition in subjects with ALF, who may develop hepatic glycolysis and impaired gluconeogenesis. These processes are concomitant with a hyperinsulinemic state and rapidly consumed glycogen reserves [43]. Thus, accurate glycemia monitoring should be performed and the administration of glucose at 1.5–2 g/kg/day is recommended in the ICU. Furthermore, both enteral and parenteral nutrition prevent hypoglycemia [44]. In detail, enteral nutrition should be started as soon as possible in both pediatric and adult patients with ALF. However, severe shock and/or gut dysfunction suggest going for parenteral nutrition. On the other hand, hyperglycemia must be avoided, as it can favor intracranial hypertension (ICH). Optimal blood-glucose levels are between 150 and 180 mg/dL [45]. Interestingly, hypertonic sodium chloride can be used to reach hypernatremia (e.g., values between 145 and 155 mmEq/L) to further reduce the incidence of ICH and consequent cerebral edema [40]. It is worth considering potential thiamin and vitamin B12 deficiencies check in patients with ALF suspected of alcohol/drug abuse. Patients may have also fat-soluble vitamins (e.g., A, D, E, and K) deficiency. In particular, the prolongation of clotting tests (namely, prolonged INR, partial thromboplastin time, and abnormal factor V) can be a feature of vitamin K deficiency. However, we must note that patients with ALF have a more frequent procoagulant tendency, which results in a hypercoagulable state [46,47]. Thus, there is no recommendation for vitamin K level dosing in these patients. Vitamin D is not only crucial for bone integrity and the prevention of osteoporosis but also modulates intestinal permeability and inflammatory response to pathogens [48]. Therefore, it can be supplemented for patients with ALF with low levels of vitamin D [49]. In addition, zinc—a microelement crucial for our immune system homeostasis—is also a cofactor for ammonia conversion to urea. Thus, zinc deficiency is strictly linked to hepatic encephalopathy [50]. Conversely, patients with ALF derived from undiagnosed Wilson disease can present with copper-driven toxicity [51]. The hydration of individuals with ALF is important in terms of hemodynamic state stability. Indeed, the hemodynamic state of patients with ALF is very similar to that of cirrhotic and/or septic patients: there is low systemic vascular resistance. However, a subject with ALF has less pronounced portal hypertension compared to cirrhotic patients. Thus, there is an urgent need to maintain the adequate perfusion of the brain and kidney to avoid cerebral edema and renal failure [52]. In particular, more patients with ALF need to maintain a cerebral perfusion pressure of 60–80 mmHg through intravenous fluid and vasopressor administration. The final goal is to reach a mean arterial pressure of at least 75 mm Hg [45,53]. Renal replacement therapy is indicated to reverse intravascular volume overload, acidosis, and electrolyte abnormalities with a significant reduction in mortality [45,54].

3.3. Gut Microbiota and Acute Liver Failure: A Potential Biologic Weapon Reversing the Syndrome

The human intestine hosts over 100 trillion microbes, prevalently bacteria. There are also viruses, fungi, archaea, and protozoa [55]. Gut microbiota participates in nutrient absorption and fermentation, modulate intestinal permeability (IP), and are implicated in host metabolism (e.g., carbohydrates absorption and processing, proteins putrefaction, bile acids formation, insulin sensitivity) and the modulation of mucosal and systemic immunity [56].
In general, some evidence from the literature suggests the degree of acute liver damage is inversely correlated with gut microbiota dysbiosis reduction. The latter is directly correlated with liver inflammation via the development of a gut–liver axis through healthy and disease-affected life [57,58]. Furthermore, the gut microbiome contains genes encoding anti-inflammatory upstream signals typical of ALF. This results in the regulation of the gene expression of MYC activation during ALF and counteracts liver injury [59].
Data on gut dysbiosis related to ALF development can be extracted from several reports on ALI. In detail, changes in the intestinal microbial composition circadian rhythm can affect acetaminophen-induced ALI and related ALF [57]. Indeed, 16S rRNA gene sequencing showed rhythm daily changes to be associated with changes in gut microbiota abundance: the ratio of Firmicutes/Bacteroides was significantly reduced at night. Accordingly, acetaminophen hepatotoxicity was more severe at night vs. in the morning. This finding can be explained by the increased abundance of the 1-phenyl-1,2-propanedione (PPD) consuming glutathione (GSH) in hepatocytes. Further, the oral gavage of acetaminophen weakens the intestinal mucosal barrier function (increasing “intestinal permeability”), allowing abundant pathogenic antigens to enter the liver, with worsening liver inflammation and damage [60]. Thus, acute liver damage can lead to gut dysbiosis, and this, in turn, can start and endure the “gut–liver axis” through intestinal tight-junction dysregulation, impaired intestinal permeability, and the passage of PAMPs and DAMPs that hit hepatocytes. This vicious circle is virtually infinite [10,11]. On the other hand, we cannot exclude the possibility that gut dysbiosis can cause a predisposition to acute liver damage and subsequent ALF.
Indeed, a crucial recent work by Schneider and co-workers shed a clear light on the role of gut microbiota in chemical-induced ALI leading to ALF. Subjects with intestinal dysbiosis are more sensitive to acetaminophen ALI. Moreover, within a cohort of 500,000 participants in the British Biobank, proton pump inhibitors (PPI) or long-term antibiotic use were associated with gut dysbiosis whose risk of ALF was significantly higher than those with gut microbiota eubiosis. In parallel, Nlrp6−/− mice (an intestinal gut dysbiotic model) also showed that gut dysbiosis is associated with the same ALI vs. wild-type mice. Even more, interestingly, this hepatic damage was reproduced after fecal bacteria transplantation [61]. Several animal studies have shown that probiotic administration can improve chemical-induced liver injury. For example, Lactobacillus salivarius LI01 and Pediococcus pentosaceus LI05 significantly showed lower levels of transaminases in ALF rats. In addition, they can prevent jaundice occurrence and, interestingly, improve the histological abnormalities typical of ALF within the liver [62]. More, interestingly this probiotic mixture can finely change the composition of the cecal microbiome [63,64]. On the other hand, from a metabolic point of view, gut microbiota metabolites (e.g., 1-phenyl-1,2-propanedione) can also aggravate liver injury through the consumption of hepatic glutathione and ultimately facilitate hepatic oxidative damage [22]. More in detail, bile acids metabolically produced by gut microbiota (namely, hexanoic acid, trigeminal, 1-hexadecanol, campesterol, d-lactose, and lithocholic acid) show a significant alteration in ALF rats. Thus, they can pathologically affect the rate of liver damage (Table 2) [65].

4. Conclusions

Nutritional support is crucial in the prevention and management of ALF patients. Specifically, amino acid, mineral, vitamin, and glucose supplementation is necessary to reduce mortality and the grade of encephalopathy. Therefore, tailored enteral nutrition is advised for the period of hospitalization. Gut microbiota modulation is an appealing preventive and therapeutic option for ALF. This suggestion is based on the evidence of gut dysbiosis involvement in liver damage initiation and perpetuation via the altered gut–liver axis. Future RCTs are needed to confirm these in vitro and animal evidence.

Author Contributions

Conceptualization, E.S. and L.A.; methodology, V.M., A.D.B., C.R. and P.S.; software, A.C.P.; validation, E.S., L.A., A.C.P. and M.L.S.; formal analysis, E.S. and N.G.; investigation, E.S., V.M. and N.G.; data curation, E.S. and L.A.; writing—original draft preparation, E.S., L.A., V.M. and L.B.; writing—review and editing, E.S., L.A., P.S. and A.C.P.; visualization, A.D.B. and V.M.; supervision, E.S., L.A. and C.R.; project administration, E.S. and L.A. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Ecker, M.E.; Paparoupa, M.; Sostmann, B.; Weissenborn, K.; Schuppert, F. Hepatic Encephalopathy Is Not Always due to Liver Cirrhosis. Case Rep. Gastroenterol. 2022, 16, 313–319. [Google Scholar] [CrossRef] [PubMed]
  2. Çalışkan, A.R.; Harputluoğlu, M. Acute liver failure in adults. Turk. J. Gastroenterol. 2019, 30, 938–939. [Google Scholar] [CrossRef] [PubMed]
  3. Berardi, G.; Tuckfield, L.; DelVecchio, M.T.; Aronoff, S. Differential Diagnosis of Acute Liver Failure in Children: A Systematic Review. Pediatr. Gastroenterol. Hepatol. Nutr. 2020, 23, 501–510. [Google Scholar] [CrossRef] [PubMed]
  4. Bazerbachi, F.; Haffar, S. Acute fulminant vs. acute-on-chronic liver failure in hepatitis E: Diagnostic implications. Infect. Dis. 2015, 47, 112. [Google Scholar] [CrossRef]
  5. Gustot, T.; Moreau, R. Acute-on-chronic liver failure vs. traditional acute decompensation of cirrhosis. J. Hepatol. 2018, 69, 1384–1393. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Sanabria-Cabrera, J.; Tabbai, S.; Niu, H.; Alvarez-Alvarez, I.; Licata, A.; Björnsson, E.; Andrade, R.J.; Lucena, M.I. N-Acetylcysteine for the Management of Non-Acetaminophen Drug-Induced Liver Injury in Adults: A Systematic Review. Front. Pharmacol. 2022, 13, 876868. [Google Scholar] [CrossRef]
  7. Shingina, A.; Vutien, P.; Uleryk, E.; Shah, P.S.; Renner, E.; Bhat, M.; Tinmouth, J.; Kim, J. Long-term Outcomes of Pediatric Living Versus Deceased Donor Liver Transplantation Recipients: A Systematic Review and Meta-analysis. Liver Transpl. 2022, 28, 437–453. [Google Scholar] [CrossRef]
  8. Shingina, A.; Ziogas, I.A.; Vutien, P.; Uleryk, E.; Shah, P.S.; Renner, E.; Bhat, M.; Tinmouth, J.; Kim, J. Adult-to-adult living donor liver transplantation in acute liver failure. Transpl. Rev. 2022, 36, 100691. [Google Scholar] [CrossRef]
  9. Koretz, R.L.; Avenell, A.; Lipman, T.O.; Braunschweig, C.L.; Milne, A.C. Does enteral nutrition affect clinical outcome? A systematic review of the randomized trials. Am. J. Gastroenterol. 2007, 102, 412–429. [Google Scholar] [CrossRef]
  10. Ni, Y.; Lu, M.; Xu, Y.; Wang, Q.; Gu, X.; Li, Y.; Zhuang, T.; Xia, C.; Zhang, T.; Gou, X.J.; et al. The Role of Gut Microbiota-Bile Acids Axis in the Progression of Non-alcoholic Fatty Liver Disease. Front. Microbiol. 2022, 13, 908011. [Google Scholar] [CrossRef]
  11. Rodriguez-Diaz, C.; Taminiau, B.; García-García, A.; Cueto, A.; Robles-Díaz, M.; Ortega-Alonso, A.; Martín-Reyes, F.; Daube, G.; Sanabria-Cabrera, J.; Jimenez-Perez, M.; et al. Microbiota diversity in nonalcoholic fatty liver disease and in drug-induced liver injury. Pharmacol. Res. 2022, 182, 106348. [Google Scholar] [CrossRef] [PubMed]
  12. Liberati, A.; Altman, D.G.; Tetzlaff, J.; Mulrow, C.; Gøtzsche, P.C.; Ioannidis, J.P.; Clarke, M.; Devereaux, P.J.; Kleijnen, J.; Moher, D. The PRISMA statement for reporting systematic reviews and meta-analyses of studies that evaluate health care interventions: Explanation and elaboration. PLoS Med. 2009, 6, e1000100. [Google Scholar] [CrossRef] [PubMed]
  13. European Association for the Study of the Liver; Wendon, J.; Cordoba, J.; Dhawan, A.; Larsen, F.S.; Manns, M.; Samuel, D.; Simpson, K.J.; Yaron, I.; Bernardi, M. EASL Clinical Practical Guidelines on the management of acute (fulminant) liver failure. J. Hepatol. 2017, 66, 1047–1081. [Google Scholar] [CrossRef] [PubMed]
  14. Weissenborn, K. Hepatic Encephalopathy: Definition, Clinical Grading and Diagnostic Principles. Drugs 2019, 79, 5–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Poordad, F.F. Review article: The burden of hepatic encephalopathy. Aliment. Pharm. 2007, 25, 3–9. [Google Scholar] [CrossRef]
  16. Hadjihambi, A.; Arias, N.; Sheikh, M.; Jalan, R. Hepatic encephalopathy: A critical current review. Hepatol. Int. 2018, 12, 135–147. [Google Scholar] [CrossRef] [Green Version]
  17. Neff, G.; Zachry, W., III. Systematic Review of the Economic Burden of Overt Hepatic Encephalopathy and Pharmacoeconomic Impact of Rifaximin. Pharmacoeconomics 2018, 36, 809–822. [Google Scholar] [CrossRef] [Green Version]
  18. Wlodzimirow, K.A.; Eslami, S.; Abu-Hanna, A.; Nieuwoudt, M.; Chamuleau, R.A. Systematic review: Acute liver failure - one disease, more than 40 definitions. Aliment. Pharm. 2012, 35, 1245–1256. [Google Scholar] [CrossRef]
  19. Stravitz, R.T.; Lee, W.M. Acute liver failure. Lancet 2019, 394, 869–881. [Google Scholar] [CrossRef]
  20. Vento, S.; Cainelli, F. Acute liver failure. Lancet 2020, 395, 1833. [Google Scholar] [CrossRef]
  21. Bernal, W.; Wendon, J. Acute liver failure. N. Engl. J. Med. 2013, 369, 2525–2534. [Google Scholar] [CrossRef] [PubMed]
  22. Lai, W.K.; Murphy, N. Management of acute liver failure. CEACCP 2004, 4, 40–43. [Google Scholar] [CrossRef] [Green Version]
  23. Arshad, M.A.; Murphy, N.; Bangash, M.N. Acute liver failure. Clin. Med. 2020, 20, 505–508. [Google Scholar] [CrossRef] [PubMed]
  24. Bischoff, S.C.; Bernal, W.; Dasarathy, S.; Merli, M.; Plank, L.D.; Schütz, T.; Plauth, M. ESPEN practical guideline: Clinical nutrition in liver disease. Clin. Nutr. 2020, 39, 3533–3562. [Google Scholar] [CrossRef] [PubMed]
  25. Kappus, M.R. Acute Hepatic Failure and Nutrition. Nutr. Clin. Pract. 2020, 35, 30–35. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Dai, X.; Gao, B.; Zhang, X.X.; Li, J.; Jiang, W.T. Value of the controlling nutritional status score and psoas muscle thickness per height in predicting prognosis in liver transplantation. World J. Clin. Cases 2021, 9, 10871–10883. [Google Scholar] [CrossRef]
  27. Dos Santos, A.L.S.; Anastácio, L.R. The impact of L-chain amino acids and L-leucine on malnutrition, sarcopenia and other outcomes in patients with chronic liver disease. Expert Rev. Gastroenterol. Hepatol. 2021, 15, 181–194. [Google Scholar] [CrossRef]
  28. Ismaiel, A.; Bucsa, C.; Farcas, A.; Leucuta, D.C.; Popa, S.L.; Dumitrascu, D.L. Effects of Brancheded-Chain Amino Acids on Parameters Evaluating Sarcopenia in Liver Cirrhosis: Systematic Review and Meta-Analysis. Front. Nutr. 2022, 9, 749969. [Google Scholar] [CrossRef]
  29. Holecek, M. Brancheded-chain amino acids in health and disease: Metabolism, alterations in blood plasma and as supplements. Nutr. Metab. 2018, 15, 33. [Google Scholar] [CrossRef] [Green Version]
  30. Mohta, S.; Anand, A.; Sharma, S.; Qamar, S.; Agarwal, S.; Gunjan, D.; Singh, N.; Madhusudhan, K.S.; Pandey, R.M.; Saraya, A. Randomised clinical trial: Effect of adding brancheded chain amino acids to exercise and standard-of-care on muscle mass in cirrhotic patients with sarcopenia. Hepatol. Int. 2022, 16, 680–690. [Google Scholar] [CrossRef]
  31. Espina, S.; Sanz-Paris, A.; Gonzalez-Irazabal, Y.; Pérez-Matute, P.; Andrade, F.; Garcia-Rodriguez, B.; Carpéné, C.; Zakaroff, A.; Bernal-Monterde, V.; Fuentes-Olmo, J.; et al. Randomized Clinical Trial: Effects of β-Hydroxy-β-Methylbutyrate (HMB)-Enriched vs. HMB-Free Oral Nutritional Supplementation in Malnourished Cirrhotic Patients. Nutrients 2022, 14, 2344. [Google Scholar] [CrossRef] [PubMed]
  32. Hernández-Conde, M.; Llop, E.; Gómez-Pimpollo, L.; Fernández Carrillo, C.; Rodríguez, L.; Van Den Brule, E.; Perelló, C.; López-Gómez, M.; Abad, J.; Martínez-Porras, J.L.; et al. Adding Brancheded-Chain Amino Acids to an Enhanced Standard-of-Care Treatment Improves Muscle Mass of Cirrhotic Patients With Sarcopenia: A Placebo-Controlled Trial. Am. J. Gastroenterol. 2021, 116, 2241–2249. [Google Scholar] [CrossRef] [PubMed]
  33. Kitajima, Y.; Takahashi, H.; Akiyama, T.; Murayama, K.; Iwane, S.; Kuwashiro, T.; Tanaka, K.; Kawazoe, S.; Ono, N.; Eguchi, T.; et al. Supplementation with brancheded-chain amino acids ameliorates hypoalbuminemia, prevents sarcopenia, and reduces fat accumulation in the skeletal muscles of patients with liver cirrhosis. J. Gastroenterol. 2018, 53, 427–437. [Google Scholar] [CrossRef] [PubMed]
  34. Bear, D.E.; Langan, A.; Dimidi, E.; Wandrag, L.; Harridge, S.D.R.; Hart, N.; Connolly, B.; Whelan, K. β-Hydroxy-β-methylbutyrate and its impact on skeletal muscle mass and physical function in clinical practice: A systematic review and meta-analysis. Am. J. Clin. Nutr. 2019, 109, 1119–1132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Hara, K.; Maruki, Y.; Long, X.; Yoshino, K.; Oshiro, N.; Hidayat, S.; Tokunaga, C.; Avruch, J.; Yonezawa, K. Raptor, a target binding partner of rapamycin (TOR), mediates mTOR action. Cell 2002, 110, 177–189. [Google Scholar] [CrossRef] [Green Version]
  36. Gallagher, P.; Richmond, S.; Dudley, K.; Prewitt, M.; Gandy, N.; Kudrna, B.; Touchberry, C. Interaction of resistance exercise and BCAA supplementation on phosphorylation of Akt and p70 s6 kinase in human skeletal muscle. PHASEB J. 2007, 21, A1206. [Google Scholar] [CrossRef]
  37. Shen, W.H.; Boyle, D.W.; Wisniowski, P.; Bade, A.; Liechty, E.A. Insulin and IGF-I stimulate eukaryotic initiation factor 4F complex formation and protein synthesis in C2C12 myotubes independent of external amino acid availability. J. Endocrinol. 2005, 185, 275–289. [Google Scholar] [CrossRef] [Green Version]
  38. Casaer, M.P.; Mesotten, D.; Hermans, G.; Wouters, P.J.; Schetz, M.; Meyfroidt, G.; Van Cromphaut, S.; Ingels, C.; Meersseman, P.; Muller, J.; et al. Early versus late parenteral nutrition in critically ill adults. N. Engl. J. Med. 2011, 365, 506–517. [Google Scholar] [CrossRef] [Green Version]
  39. Sakusic, A.; Sabov, M.; McCambridge, A.J.; Rabinstein, A.A.; Singh, T.D.; Mukesh, K.; Kashani, K.B.; Cook, D.; Gajic, O. Features of Adult Hyperammonemia Not Due to Liver Failure in the ICU. Crit. Care Med. 2018, 46, e897–e903. [Google Scholar] [CrossRef]
  40. Osowska, S.; Kunecki, M.; Sobocki, J.; Tokarczyk, J.; Majewska, K.; Omidi, M.; Radkowski, M.; Fisk, H.L.; Calder, P.C. Effect of changing the lipid component of home parenteral nutrition in adults. Clin. Nutr. 2019, 38, 1355–1361. [Google Scholar] [CrossRef]
  41. Adelusi, O.B.; Ramachandran, A.; Lemasters, J.J.; Jaeschke, H. The role of Iron in lipid peroxidation and protein nitration during acetaminophen-induced liver injury in mice. Toxicol. Appl. Pharm. 2022, 445, 116043. [Google Scholar] [CrossRef] [PubMed]
  42. Walsh, T.S.; Wigmore, S.J.; Hopton, P.; Richardson, R.; Lee, A. Energy expenditure in acetaminophen-induced fulminant hepatic failure. Crit. Care Med. 2000, 28, 649–654. [Google Scholar] [CrossRef] [PubMed]
  43. Oldenbeuving, G.; McDonald, J.R.; Goodwin, M.L.; Sayilir, R.; Reijngoud, D.J.; Gladden, L.B.; Nijsten, M.W. A patient with acute liver failure and extreme hypoglycaemia with lactic acidosis who was not in a coma: Causes and consequences of lactate-protected hypoglycaemia. Anaesth. Intensive Care 2014, 42, 507–511. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Alsaleem, M.; Saadeh, L.; Kumar, V.H.S.; Wilding, G.E.; Miller, L.; Mathew, B. Continued enteral feeding is beneficial in hypoglycemic infants admitted to intensive care for parenteral dextrose therapy. Glob. Pediatr. Health 2019, 6, 2333794X19857415. [Google Scholar] [CrossRef]
  45. Kramer, A.H.; Roberts, D.J.; Zygun, D.A. Optimal glycemic control in neurocritical care patients: A systematic review and meta-analysis. Crit. Care 2012, 16, R203. [Google Scholar] [CrossRef] [Green Version]
  46. Hugenholtz, G.C.; Adelmeijer, J.; Meijers, J.C.; Porte, R.J.; Stravitz, R.T.; Lisman, T. An unbalance between von Willebrand factor and ADAMTS13 in acute liver failure: Implications for hemostasis and clinical outcome. Hepatology 2013, 58, 752–761. [Google Scholar] [CrossRef]
  47. Lisman, T.; Bakhtiari, K.; Adelmeijer, J.; Meijers, J.C.; Porte, R.J.; Stravitz, R.T. Intact thrombin generation and decreased fibrinolytic capacity in patients with acute liver injury or acute liver failure. J. Thromb. Haemost. 2012, 10, 1312–1319. [Google Scholar] [CrossRef]
  48. Valle, M.; Mitchell, P.L.; Pilon, G.; Varin, T.; Hénault, L.; Rolin, J.; McLeod, R.; Gill, T.; Richard, D.; Vohl, M.C.; et al. Salmon peptides limit obesity-associated metabolic disorders by modulating a gut-liver axis in vitamin D-deficient mice. Obesity 2021, 29, 1635–1649. [Google Scholar] [CrossRef]
  49. Grama, A.; Burac, L.; Aldea, C.O.; Bulata, B.; Delean, D.; Samasca, G.; Abrudan, C.; Sirbe, C.; Pop, T.L. Vitamin D-Binding Protein (Gc-Globulin) in Acute Liver Failure in Children. Diagnostics 2020, 10, 278. [Google Scholar] [CrossRef]
  50. Janyajirawong, R.; Vilaichone, R.K.; Sethasine, S. Efficacy of Zinc Supplement in Minimal hepatic Encephalopathy: A prospective, Randomized Controlled Study (Zinc-MHE Trial). Asian Pac. J. Cancer Prev. 2021, 22, 2879–2887. [Google Scholar] [CrossRef]
  51. Pop, T.L.; Grama, A.; Stefanescu, A.C.; Willheim, C.; Ferenci, P. Acute liver failure with hemolytic anemia in children with Wilson’s disease: Genotype-phenotype correlations? World J. Hepatol. 2021, 13, 1428–1438. [Google Scholar] [CrossRef] [PubMed]
  52. Montrief, T.; Koyfman, A.; Long, B. Acute liver failure: A review for emergency physicians. Am. J. Emerg. Med. 2019, 37, 329–337. [Google Scholar] [CrossRef] [PubMed]
  53. Stravitz, R.T.; Kramer, D.J. Management of acute liver failure. Nat. Rev. Gastroenterol. Hepatol. 2009, 6, 542–553. [Google Scholar] [CrossRef] [PubMed]
  54. Cardoso, F.S.; Gottfried, M.; Tujios, S.; Olson, J.C.; Karvellas, C.J.; Group USALFS. Continuous renal replacement therapy is associated with reduced serum ammonia levels and mortality in acute liver failure. Hepatology 2018, 67, 711–720. [Google Scholar] [CrossRef] [Green Version]
  55. Scarpellini, E.; Fagoonee, S.; Rinninella, E.; Rasetti, C.; Aquila, I.; Larussa, T.; Ricci, P.; Luzza, F.; Abenavoli, L. Gut Microbiota and Liver Interaction through Immune System Cross-Talk: A Comprehensive Review at the Time of the SARS-CoV-2 Pandemic. J. Clin. Med. 2020, 9, 2488. [Google Scholar] [CrossRef]
  56. Maynard, C.L.; Elson, C.O.; Hatton, R.D.; Weaver, C.T. Reciprocal interactions of the intestinal microbiota and immune system. Nature 2012, 489, 231–241. [Google Scholar] [CrossRef] [Green Version]
  57. Gong, S.; Lan, T.; Zeng, L.; Luo, H.; Yang, X.; Li, N.; Hen, X.; Liu, Z.; Li, R.; Win, S.; et al. Gut microbiota mediates diurnal variation of acetaminophen induced acute liver injury in mice. J. Hepatol. 2018, 69, 51–59. [Google Scholar] [CrossRef]
  58. Castillo-Dela Cruz, P.; Wanek, A.G.; Kumar, P.; An, X.; Elsegeiny, W.; Horne, W.; Fitch, A.; Burr, A.H.P.; Gopalakrishna, K.P.; Chen, K.; et al. Intestinal IL-17R Signaling Constrains IL-18-Driven Liver Inflammation by the Regulation of Microbiome-Derived Products. Cell Rep. 2019, 29, 2270–2283. [Google Scholar] [CrossRef] [Green Version]
  59. Kolodziejczyk, A.A.; Federici, S.; Zmora, N.; Mohapatra, G.; Dori-Bachash, M.; Hornstein, S.; Leshem, A.; Reuveni, D.; Zigmond, E.; Tobar, A.; et al. Acute liver failure is regulated by MYC- and microbiome-dependent programs. Nat. Med. 2020, 26, 1899–1911. [Google Scholar] [CrossRef]
  60. Niu, M.; Luo, Z.; Gong, S.; Win, S.; Kaplowitz, N.; Jiang, Y.; Chen, P. Intestinal Epithelial Chemokine (C-C Motif) Ligand 7 Overexpression Enhances Acetaminophen-Induced Hepatotoxicity in Mice. Am. J. Pathol. 2020, 190, 57–67. [Google Scholar] [CrossRef]
  61. Schneider, K.M.; Elfers, C.; Ghallab, A.; Schneider, C.V.; Galvez, E.J.C.; Mohs, A.; Gui, W.; Candles, L.S.; Wirtz, T.H.; Zuehlke, S.; et al. Intestinal Dysbiosis Amplifies Acetaminophen-Induced Acute Liver Injury. Cell. Mol. Gastroenterol. Hepatol. 2021, 11, 909–933. [Google Scholar] [CrossRef] [PubMed]
  62. Lv, L.X.; Hu, X.J.; Qian, G.R.; Zhang, H.; Lu, H.F.; Zheng, B.W.; Jiang, L.; Li, L.J. Administration of Lactobacillus salivarius LI01 or Pediococcus pentosaceus LI05 improves acute liver injury induced by D-galactosamine in rats. Appl. Microbiol. Biotechnol. 2014, 98, 5619–5632. [Google Scholar] [CrossRef] [PubMed]
  63. Shi, D.; Lv, L.; Fang, D.; Wu, W.; Hu, C.; Xu, L.; Chen, Y.; Guo, J.; Hu, X.; Li, A.; et al. Administration of Lactobacillus salivarius LI01 or Pediococcus pentosaceus LI05 prevents CCl4-induced liver cirrhosis by protecting the intestinal barrier in rats. Sci. Rep. 2017, 7, 6927. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Zhuge, A.; Li, B.; Yuan, Y.; Lv, L.; Li, Y.; Wu, J.; Yang, L.; Bian, X.; Wang, K.; Wang, Q.; et al. Lactobacillus salivarius LI01 encapsulated in alginate-pectin microgels ameliorates D-galactosamine-induced acute liver injury in rats. Appl. Microbiol. Biotechnol. 2020, 104, 7437–7455. [Google Scholar] [CrossRef] [PubMed]
  65. Jiang, H.; Yan, R.; Wang, K.; Wang, Q.; Chen, X.; Chen, L.; Li, L.; Lv, L. Lactobacillus Reuteri DSM 17938 Alleviates D-Galactosamine-Induced Liver Failure in Rats. BioMed. Pharm. 2021, 133, 111000. [Google Scholar] [CrossRef]
  66. Saeedi, B.J.; Liu, K.H.; Owens, J.A.; Hunter-Chang, S.; Camacho, M.C.; Eboka, R.U.; Chandrasekharan, B.; Baker, N.F.; Darby, T.M.; Robinson, B.S.; et al. Gut-Resident Lactobacilli Activate Hepatic Nrf2 and Protect Against Oxidative Liver Injury. Cell. Metab. 2020, 31, 956–968.e5. [Google Scholar] [CrossRef]
  67. Neag, M.A.; Catinean, A.; Muntean, D.M.; Pop, M.R.; Bocsan, C.I.; Botan, E.C.; Buzoianu, A.D. Probiotic Bacillus Spores Protect Against Acetaminophen Induced Acute Liver Injury in Rats. Nutrients 2020, 12, 632. [Google Scholar] [CrossRef] [Green Version]
  68. Li, Y.; Lv, L.; Ye, J.; Fang, D.; Shi, D.; Wu, W.; Wang, Q.; Wu, J.; Yang, L.; Bian, X.; et al. Bifidobacterium adolescentis CGMCC 15058 alleviates liver injury, enhances the intestinal barrier and modifies the gut microbiota in D-galactosamine-treated rats. Appl. Microbiol. Biotechnol. 2019, 103, 375–393. [Google Scholar] [CrossRef]
  69. Li, Y.T.; Ye, J.Z.; Lv, L.X.; Xu, H.; Yang, L.Y.; Jiang, X.W.; Wu, W.R.; Shi, D.; Fang, D.Q.; Bian, X.Y.; et al. Pretreatment with Bacillus cereus Preserves Against D-Galactosamine-Induced Liver Injury in a Rat Model. Front. Microbiol. 2019, 10, 1751. [Google Scholar] [CrossRef] [Green Version]
  70. Grander, C.; Adolph, T.E.; Wieser, V.; Lowe, P.; Wrzosek, L.; Gyongyosi, B.; Ward, D.V.; Grabherr, F.; Gerner, R.R.; Pfister, A.; et al. Recovery of ethanol-induced Akkermansia muciniphila depletion ameliorates alcoholic liver disease. Gut 2018, 67, 891–901. [Google Scholar] [CrossRef]
  71. Wu, W.; Lv, L.; Shi, D.; Ye, J.; Fang, D.; Guo, F.; Li, Y.; He, X.; Li, L. Protective Effect of Akkermansia muciniphila against Immune-Mediated Liver Injury in a Mouse Model. Front. Microbiol. 2017, 8, 1804. [Google Scholar] [CrossRef] [PubMed]
  72. Yu, L.; Zhao, X.K.; Cheng, M.L.; Yang, G.Z.; Wang, B.; Liu, H.J.; Hu, Y.X.; Zhu, L.L.; Zhang, S.; Xiao, Z.W.; et al. Saccharomyces boulardii Administration Changes Gut Microbiota and Attenuates D-Galactosamine-Induced Liver Injury. Sci. Rep. 2017, 7, 1359. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Wang, Y.; Xie, J.; Li, Y.; Dong, S.; Liu, H.; Chen, J.; Wang, Y.; Zhao, S.; Zhang, Y.; Zhang, H. Probiotic Lactobacillus casei Zhang reduces pro-inflammatory cytokine production and hepatic inflammation in a rat model of acute liver failure. Eur. J. Nutr. 2016, 55, 821–831. [Google Scholar] [CrossRef] [PubMed]
  74. Wang, Y.; Liu, Y.; Sidhu, A.; Ma, Z.; McClain, C.; Feng, W. Lactobacillus rhamnosus GG culture supernatant ameliorates acute alcohol-induced intestinal permeability and liver injury. Am. J. Physiol. Gastrointest. Liver Physiol. 2012, 303, G32–G41. [Google Scholar] [CrossRef] [PubMed]
Table 1. Main causes of ALF.
Table 1. Main causes of ALF.
Cause of ALF
Acetaminophen
Other drugs
Acute viral hepatitis
Acute-onset autoimmune hepatitis
Wilson disease
Acute ischemia
Budd–Chiari syndrome
Veno-occlusive disease
Acute fatty liver of pregnancy
Cancer liver infiltration
Hepatectomy
Toxins
Sepsis
Heat stroke
Hemophagocytic lymphohistiocytosis
Table 2. Preclinical use of probiotics in ALF models.
Table 2. Preclinical use of probiotics in ALF models.
Probiotic Used and DoseAnimal ModelLiver DamageEffectsRef.
Lactobacillus reuteri DSM 17,938 (oral gavage with 3 × 109 CFU daily
for one week)
Sprague-Dawley ratsD-GaIN (1.1 g/kg of body weight)
intraperitoneally injected
↓ dysbiosis;
↓ transcription of inflammatory
factors in hepatocytes
[65]
Lactobacillus rhamnosus GG (oral
gavage with 2 × 108 CFU/100 μL HBSS
daily for 2 weeks)
Germ-free C57BL/6 miceAcetaminophen (300 mg/kg
of body weight) oral gavage
Production of 5-MIAA with the activation of Nrf2 within the liver to protect against drug-induced
oxidative stress
[66]
MegaSporeBioticTM (MSB) (orally 1
× 109 CFU/rat daily for 12 days)
Charles River Wistar white male ratsAcetaminophen (2 g/kg of body weight) oral gavage↓ pro-inflammatory
cytokines production, ↓ hepatocyte necrosis
[67]
Bifidobacterium adolescents
CGMCC15058 (gavage 3 × 109 CFU/mL PBS daily for 2 weeks)
Germ-free Sprague–Dawley
(SD) rats
D-GaIN (1.1 g/kg of body weight) intraperitoneal injection↓ levels of mTOR and
inflammatory cytokines;
↑ levels of anti-inflammatory
cytokines (e.g., interleukin-10)
[68]
Bacillus cereus (gavage at 3 × 109 CFU/mL PBS daily for 2 weeks)Sprague–Dawley ratsD-GaIN (1.1 g/kg of body weight) intraperitoneal injection↓ inflammatory response;
improved intestinal permeability;
re-establishing of eubiota
[69]
Akkermansia muciniphila (oral
gavage 1.5 × 109 CFU/200 μL PBS daily for 2 days)
C57BL/6 miceAlcohol (6 g/kg of body weight) oral gavage↓ hepatic
injury, steatosis, and infiltration of MPO+ neutrophils
[70]
Akkermansia muciniphila (oral gavage
3 × 109 CFU/200 μL PBS daily for 2 weeks)
C57BL/6 miceCon A (15 mg/kg of body weight) injection through
the tail vein
↓ inflammatory cytokines production,
cytotoxic factors and hepatocellular
necrosis;
↑ gut microbiota diversity
[71]
Saccharomyces boulardii (gavaged
with 1 × 109 CFU/mL for 4 weeks)
BALB/c miceD-GaIN (200 mg/kg of body weight) intraperitoneally injectedBacteroidetes abundance;
↓ the abundance of Firmicutes and Proteobacteria
[72]
Lactobacillus casei Zhang (gavaged
with 109 CFU/daily for one month)
Wistar ratsLPS / D-GalN (50 μg/kg and 300 mg/kg of body weight, LPS and
D-GalN, respectively) intraperitoneal injection
Modulation of the TLR-MAPK-PPAR-g pathways leading to ↓↓ pro-inflammatory cytokine production and hepatic inflammation[73]
Lactobacillus rhamnosus GG (mixed
with drinking water approximately 109 CFU/daily for 5 days)
C57BL/6N miceAlcohol (6 g/kg
of body weight) oral gavage
Activating HIF signaling to:
↓ hepatic damage, impaired intestinal permeability, and levels of endotoxemia
[74]
D-GaIN, D-galactosamine; 5-MIAA, 5-methoxyindoleacetic acid; Con A, concanavalin A.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Abenavoli, L.; Maurizi, V.; Boccuto, L.; Di Berardino, A.; Giostra, N.; Santori, P.; Scarcella, M.L.; Procopio, A.C.; Rasetti, C.; Scarpellini, E. Nutritional Support in Acute Liver Failure. Diseases 2022, 10, 108. https://doi.org/10.3390/diseases10040108

AMA Style

Abenavoli L, Maurizi V, Boccuto L, Di Berardino A, Giostra N, Santori P, Scarcella ML, Procopio AC, Rasetti C, Scarpellini E. Nutritional Support in Acute Liver Failure. Diseases. 2022; 10(4):108. https://doi.org/10.3390/diseases10040108

Chicago/Turabian Style

Abenavoli, Ludovico, Valentina Maurizi, Luigi Boccuto, Arianna Di Berardino, Nena Giostra, Pierangelo Santori, Maria Laura Scarcella, Anna Caterina Procopio, Carlo Rasetti, and Emidio Scarpellini. 2022. "Nutritional Support in Acute Liver Failure" Diseases 10, no. 4: 108. https://doi.org/10.3390/diseases10040108

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop