EASL Clinical Practice Guidelines on nutrition in chronic liver disease 您所在的位置:网站首页 European Association for the Study of the Liver EASL Clinical Practice Guidelines on nutrition in chronic liver disease

EASL Clinical Practice Guidelines on nutrition in chronic liver disease

2024-07-16 02:06| 来源: 网络整理| 查看: 265

J Hepatol. Author manuscript; available in PMC 2020 Jan 1.Published in final edited form as:J Hepatol. 2019 Jan; 70(1): 172–193. Published online 2018 Aug 23. doi: 10.1016/j.jhep.2018.06.024PMCID: PMC6657019NIHMSID: NIHMS1526417PMID: 30144956EASL Clinical Practice Guidelines on nutrition in chronic liver diseaseEuropean Association for the Study of the Liver Author information Copyright and License information PMC DisclaimerEuropean Association for the Study of the Liver (EASL), The EASL Building – Home of Hepatology, 7 rue Daubin, CH 1203 Geneva, Switzerland.Corresponding author. Address: European Association for the Study of the Liver (EASL), The EASL Building – Home of Hepatology, 7 rue Daubin, CH 1203 Geneva, Switzerland. Tel.: +41 (0) 22 807 03 60; fax: +41 (0) 22 328 07 24. ue.eciffolsae@eciffolsaePMC Copyright notice Publisher's DisclaimerThe publisher's final edited version of this article is available at J HepatolAssociated DataSupplementary Materials1.NIHMS1526417-supplement-1.docx (67K)GUID: D16CE799-6405-470C-9F6E-81850B28F7D3Introduction

Malnutrition is frequently a burden in patients with liver cirrhosis, occurring in 20–50% of patients. The progression of malnutrition is associated with that of liver failure. While malnutrition may be less evident in patients with compensated cirrhosis it is easily recognisable in those with decompensated cirrhosis. Malnutrition has been reported in 20% of patients with compensated cirrhosis and in more than 50% of patients with decompensated liver disease(1). Both adipose tissue and muscle tissue can be depleted; female patients more frequently develop a depletion in fat deposits while males more rapidly lose muscle tissue(2)(1).

As detailed in these clinical practice guidelines (CPGs), malnutrition and muscle mass loss (sarcopenia), which has often been used as an equivalent of severe malnutrition (3), are associated with a higher rate of complications (4) such as susceptibility to infections (5), hepatic encephalopathy (HE) (6) and ascites (4), as well as being independent predictors of lower survival in cirrhosis (7, 8) and in patients undergoing liver transplantation (9). Given these observations, malnutrition and sarcopenia should be recognised as a complication of cirrhosis, which in turn worsens the prognosis of cirrhotic patients.

Whether malnutrition can be reversed in cirrhotic patients is controversial. Although there is general agreement about the need to improve the dietary intake of these patients, avoiding those limitations and restrictions that are not evidence based, amelioration of the nutritional status and muscle mass is not always achievable (10–12).

Although the term “malnutrition” refers both to deficiencies and to excesses in nutritional status, in the present CPGs “malnutrition” refers to “undernutrition”. More recently, in addition to undernutrition, overweight or obesity are increasingly observed in cirrhotic patients because of the increasing number of cirrhosis cases related to non-alcoholic steatohepatitis (NASH). Muscle mass depletion may also occur in these patients, but due to the coexistence of obesity, sarcopenia might be overlooked. Obesity and sarcopenic obesity may worsen the prognosis of patients with liver cirrhosis (13–15)(3).

No previous guidelines released by the European Association for the Study of Liver Disease (EASL) have dealt with nutrition in advanced liver disease and/or have evaluated the relationship between nutritional status and the clinical outcome of patients. Therefore, the EASL Governing Board has asked a panel of experts in the field of nutrition and hepatology to produce the present CPGs.

Methodology

The panel initially established the most relevant questions to answer, considering relevance, urgency and completeness of the topics to be covered. The main questions addressed were: How can nutritional problems be recognised? In which conditions are nutritional assessments recommended? What are the available methods of evaluation? What are the consequences of malnutrition and its correction? Different clinical scenarios have been considered with special attention paid to nutrition in HE and before and after liver transplantation. A section devoted to bone metabolism in chronic liver disease has also been included. Each expert took responsibility and made proposals for statements for a specific section of the guideline.

The literature search was performed in different databases (PubMed, Embase, Google Scholar, Scopus) and reference from papers identified. The initial key words were: “Nutrition” OR “Nutritional status” OR “Malnutrition” OR “Sarcopenia” AND “Liver cirrhosis” OR “Chronic liver Disease”. Further, more specific key words were also utilised: “nutritional assessment”, “nutrition risk”, “hepatic encephalopathy”, “osteoporosis”, “liver transplantation”) for each specific topic of the guideline. The selection of references was based on appropriateness of study design, number of patients, and publication in peer-reviewed journals. Original data were prioritised. The resulting literature database was made available to all members of the panel.

All recommendations were discussed and approved by all participants. The Committee met on two occasions during international meetings with experts who were available to participate, two ad hoc teleconferences also took place for discussion and voting.

The evidence and recommendations in these guidelines have been graded according to the Grading of Recommendations Assessment, Development and Evaluation (GRADE) system (16). The classifications and recommendations are therefore based on three categories: the source of evidence in levels I through III; the quality of evidence designated by high (A), moderate (B), or low quality (C); and the strength of recommendations classified as strong (1) or weak (2) (Table 1). All recommendations based on expert opinion because of the lack of available data were graded as C. The recommendations were peer-reviewed by external expert reviewers and approved by the EASL Governing Board.

Table 1.

Evidence quality according to the GRADE scoring system.

GradeEvidence IRandomized, controlled trials II-1Controlled trials without randomization II-2Cohort or case-control analytic studies II-3Multiple time series, dramatic uncontrolled experiments IIIOpinions of respected authorities, descriptive epidemiologyEvidence (quality)Description HighFurther research is very unlikely to change our confidence in the estimated effectA ModerateFurther research is likely to have an important impact on our confidence in the estimate effect and may change the estimateB LowFurther research is likely to have an important impact on our confidence in the estimate effect and is likely to change the estimate. Any change of estimate is uncertainCRecommendation StrongFactors influencing the strength of recommendation included the quality of evidence, presumed patient-important outcomes, and costs1 WeakVariability in preferences and values, or more uncertainty. Recommendation is made with less certainty, higher costs, or resource consumption2Open in a separate window

These guidelines are directed at consultant hepatologists, specialists in training, and general practitioners and refer specifically to adult patients with cirrhosis. Their purpose is to provide guidance on the best available evidence to deal with nutritional problems in patients with chronic liver disease. A few schemes were produced by the panel and are included in these guidelines to help with the management of nutritional problems in patients with liver cirrhosis.

For clarity, the terms and definitions used in the present CPGs are summarised (Box 1).

BOX:TERMINOLOGY UTILIZEDMalnutritionA nutrition-related disorder resulting from lack of intake or uptake of nutrition that leads to altered body composition (decreased fat free mass) and body cell mass leading to diminish physical and mental function and impaired clinical outcome from disease. In the present CPGs, we have used “malnutrition” as a synonymous of “undernutrition”.UndernutritionSynonym of malnutrition (see above)Muscle wastingThe active, progressive loss of muscle mass due to an underlying disease, ultimately leading to muscle atrophy. Most inflammatory diseases, malnutrition and increased catabolism induce muscle wasting.SarcopeniaA generalized reduction in muscle mass and function due to aging (primary sarcopenia), acute or chronic illness (secondary sarcopenia), including chronic liver disease.FrailtyLoss of functional, cognitive, and physiologic reserve leading to a vulnerable state. Frailty may be considered a form of nutrition-related disorderImmunonutritionUse of specific nutrients in an attempt to modulate the immune system (not necessarily in the presence of malnutrition) and function to improve health state. Examples include enteral nutritional formulas enriched with ω−3 fatty acids, arginine, glutamine and nucleotidesDeconditioningDeterioration of muscle functional capacity related to immobility and chronic debilitating diseaseOpen in a separate windowScreening and assessment for malnutrition and obesity in liver cirrhosis: Who, when and how

Given the worse prognosis associated with malnutrition, all patients with advanced chronic liver disease, and in particular patients with decompensated cirrhosis are advised to undergo a rapid nutritional screen. Those at risk of malnutrition should complete a more detailed nutritional assessment to confirm the presence and severity of malnutrition (17–19), in order to actively manage this complication.

Nutrition screening tools

Two simple criteria stratify patients at high risk of malnutrition: being underweight, defined as a body mass index (BMI) (kg.body weight [BW]/[height in metre]2) Open in a separate windowFigure 1Nutritional screening and assessment in patients with cirrhosis.

All patients should undergo a rapid screening of malnutrition using validated, accepted tools. A liver specific screening tool which takes into consideration fluid retention may be advisable (Royal Free Hospital Nutritional Prioritizing Tool (RFH-NPT). Patients found to be at high risk of malnutrition should undergo a detailed nutritional assessment, and based on the findings they should receive either supplementation or regular follow-up. †In a case of fluid retention, body weight should be corrected by evaluating the patient’s dry weight by post-paracentesis body weight or weight recorded before fluid retention if available, or by subtracting a percentage of weight based upon severity of ascites (mild, 5%; moderate, 10%; severe, 15%), with an additional 5% subtracted if bilateral pedal edema is present.

RecommendationsPerform a rapid nutritional screen in all patients with cirrhosis and complete a detailed assessment in those at risk of malnutrition, to confirm the presence and severity of malnutrition. (Grade II-2, B1)Assume risk for malnutrition to be high if BMI 30 kg/m2) consider the confounding effect of fluid retention. Estimate dry BW, even though the accuracy is low. (Grade II-2, B2)Always include an assessment of sarcopenia within the nutritional assessment. (Grade II-2, B1)Whenever CT scan has been performed, assess muscle mass on images by this method. Anthropometry, DEXA or BIA are possible alternatives, which also enable serial measurements. (Grade II-2, B1)Assess muscle function, in the clinical setting, with the most appropriate tool, such as handgrip strength and/or the short physical performance battery. (Grade II-2, B1)Assess dietary intake by trained personnel (ideally a dietician with knowledge of managing patients with liver disease) working as part of a team with the hepatologist. Assessment should include: quality and quantity of food and supplements, fluids, sodium in diet, number and timing of meals during the day and barriers to eating. (Grade II-2, B1)Nutritional management principles in patients with liver cirrhosis

Since malnutrition and sarcopenia are independent predictors of adverse clinical outcomes including survival (57),(58, 59), (17) (60) any nutritional approach in cirrhotic patients needs to follow some general principles of nutritional management.

Energy and protein requirements in cirrhosis

Cirrhosis is a state of accelerated starvation demonstrated by a rapid post absorptive physiology which is characterised by a reduction in the respiratory quotient(61, 62). The reduction in the respiratory quotient is the manifestation of a metabolic switch in the primary fuel from glucose to fatty acids. During this state of accelerated starvation, protein synthesis is decreased and gluconeogenesis from amino acids is increased, necessitating proteolysis, which contributes to sarcopenia. Gluconeogenesis is an energy-expensive procedure which may further increase resting energy expenditure (REE) in these patients. Accelerated starvation is aggravated by reduced dietary intake due to a variety of factors including dysgeusia, anorexia of chronic disease, salt restricted food that is not tasty, portal hypertension that contributes to impaired gut motility, decreased nutrient absorption and protein losing enteropathy (63–66). Additional factors that result in decreased dietary intake include inappropriate dietary protein restriction, hospitalisation with periods of fasting for diagnostic and therapeutic procedures, encephalopathy and gastrointestinal bleeding.

Energy supply needs to balance total energy expenditure (TEE), which includes REE, food-related thermogenesis and energy expenditure related to physical activity. TEE is measured ideally with doubly labelled water or in a respiratory chamber, but these methods are not feasible in the clinical setting. Physical activity is reduced in patients with decompensated cirrhosis and negligible when patients are hospitalised. In cirrhotic patients, TEE varies between 28 to 37.5 kcal/kg.BW/d. (63, 67–70). Some studies evaluated whether decompensated liver cirrhosis affected REE. One small longitudinal study suggested that ascites increases REE (71). However, a cross-sectional study found no difference in REE between patients with varying levels of liver disease severity and fluid retention (72–74). Measured REE may be higher than predicted, a situation termed hypermetabolism. However, hypermetabolism cannot be identified by clinical or laboratory parameters (75), the severity and the aetiology of liver cirrhosis and the presence of ascites (25). REE may be estimated by predictive formulae but these are inaccurate in advanced cirrhotic patients, and thus measurement by indirect calorimetry is advisable whenever possible (61, 62). The availability of the hand held calorimeter at the bedside is a possible alternative to determine a patient’s daily caloric needs (76).

The approach of most nutritional intervention studies in liver cirrhosis is to supply at least 35 kcal/kg.BW/d. The use of actual BW, corrected for ascites (see previous section), is considered safe. This can be achieved primarily by tailoring the oral dietary intake, even though this goal is frequently difficult to accomplish. The role of a nutrition support team has recently been underlined by a retrospective study showing that nutritional intervention, led by a multidisciplinary team, and in which cirrhotic patients participated in teaching sessions about the relevance of appropriate nutrition in chronic liver disease, was able to improve survival rates and quality of life(77).

Whether frequent feeding can help prevent accelerated starvation and the related proteolysis has also been extensively evaluated. Since the longest inter-meal duration is at night, strategies to shorten nocturnal fasting with a late evening snack have been explored, achieving an improvement in metabolic profile and quality of life, although muscle mass did not show consistent improvement(78). The adoption of a breakfast containing some proteins (79) and a late evening snack (80) to shorten the period of fasting are therefore recommended in cirrhotic patients.

Protein needs are based on the minimum protein intake required to maintain nitrogen balance. In alcoholic cirrhosis, nitrogen balance was achieved with intakes of 0.8 g/kg.BW/d (81). This cut-off was confirmed in studies wherein cirrhotic patients received diets with increasing protein content (70, 82). These studies also showed that cirrhotic patients are able to utilise up to 1.8 g/kg.BW/d of protein (70). In the past, there has been controversy about whether patients suffering from HE should undergo a transient restriction in protein intake, in order to limit the synthesis of ammonium and the deamination of protein to aromatic amino acids. However, normal to high protein intake does not precipitate HE (83),(84) and may even improve mental status (85), (86) (see paragraph on hepatic encephalopathy).

The recommended protein intake in patients with a diagnosis of liver cirrhosis is 1.2–1.5 g/kg.BW/d to prevent loss of muscle mass and reverse muscle loss in those who are sarcopenic. Indeed, sarcopenia, as previously stated, contributes to worse clinical outcomes, independent of the severity of liver disease (27, 63). Options for the treatment of sarcopenia will be discussed in the next section.

Short dietary advice for use when treating a cirrhotic patient at bedside or during an outpatient visit is provided (Table 2).

Table 2

Short, practical dietary advice for bedside or outpatient clinic use.

● Most of what you have heard/read on the relationship between food and the liver has limited scientific evidence to support it. Generally, healthy eating of a variety of foods is advisable to all patients.● Virtually no food other than alcohol does actually damage the liver and/or is genuinely contraindicated in patients with chronic liver disease.● In most patients with chronic liver disease, eating an adequate amount of calories and protein is much more important than avoiding specific types of food, so it is important that you have a good, varied diet that you enjoy.● You should try to split your food intake into 3 main meals (breakfast, lunch and dinner) and 3 snacks (mid-morning, mid-afternoon, late evening). The late-evening snack is the most important, as it covers the long interval between dinner and breakfast.● You should try to eat as much vegetables and fruit as you can. If you feel that this makes you feel bloated, and that it makes you eat less, please report to your doctor or dietician.● You should try not to add too much salt to your food. It may take some time to adjust, but it usually gets easier with time. However, if you keep feeling that this makes your food unpleasant to eat, and that it makes you eat less, please report to your doctor or dietician.● A limited proportion of patents with liver disease have a complication called hepatic encephalopathy, which may make them tolerate animal protein (meat) less well than vegetable protein (beans, peas etc) and dairy proteins. Before you make any changes to your protein intake, you should always ask your doctor or dietician. Please do not reduce your total protein intake as it is not advisable in cirrhosis.● Some patients with liver disease have other diseases, for example diabetes or overweight/obesity, which require dietary adjustments. Please remember to tell your doctor about all your illnesses and about any dietary advice you have already received from other doctors, nurses or dieticians.Open in a separate windowApproach to sarcopenia in patients with liver cirrhosisFactors related with sarcopenia in patients with cirrhosis

Skeletal muscle mass is the largest protein store in the body. A balance between skeletal muscle protein synthesis and breakdown is responsible for protein homeostasis (or proteostasis) that maintains skeletal muscle mass(66, 87, 88). In the past, whole body protein turnover studies have yielded conflicting results with unaltered, increased or decreased protein synthesis and breakdown in cirrhosis (3, 89). Skeletal muscle mass depends on a number of factors including age, gender and ethnicity in physiological states. The severity and aetiology of liver disease also affects muscle mass, with cholestatic and alcoholic liver disease leading to the most severe muscle loss independently of the severity of the underlying liver disease, although data on alcoholic liver disease are not consistent (66, 90). Hepatocellular dysfunction and portosystemic shunting also result in biochemical and hormonal perturbations in cirrhosis that contribute to sarcopenia.

Increased skeletal muscle ammonia, reduction in testosterone and growth hormone, endotoxemia, as well as decreased dietary nutrient intake contribute to sarcopenia(89, 91–93). In addition, amino acid perturbations, specifically reduction in the branched chain amino acid, L-leucine, and consequent impaired global protein synthesis has also been reported to contribute to sarcopenia in cirrhosis(3, 94–97). To better understand the progressive depletion of muscle mass in cirrhotic patients, molecular mechanisms of muscle wasting have recently been investigated (Fig. 2). Molecular pathways that regulate skeletal muscle mass include myostatin, a TGFβ superfamily member that inhibits protein synthesis and potentially increases proteolysis (88). Data in animal models, humans and cellular systems have consistently shown that myostatin expression is increased in cirrhosis (95, 98, 99). In addition to impaired protein synthesis, proteolysis is also required for loss of muscle mass (63, 66). The ubiquitin proteasome pathway and autophagy are currently believed to be the dominant mechanisms of skeletal muscle proteolysis (63, 100). Human skeletal muscle from patients with cirrhosis and preclinical models of hyperammonaemia show increased autophagy with impaired or unaltered proteasome-mediated proteolysis (95, 100, 101). A more extensive view of molecular mechanisms of muscle wasting in patients with liver cirrhosis is reviewed in reference 19 (18).

Open in a separate windowFigure 2Mechanisms and potential targets for anabolic resistance and dysregulated proteostasis resulting in sarcopenia and/or failure to respond to standard supplementation.Strategies to improve muscle mass in cirrhosis

A number of potential therapeutic strategies to improve muscle mass in patients with cirrhosis have been evaluated. These include dietary manipulations, increased physical activity and exercise (3, 102–104), hormone replacement therapies, (105) ammonia-lowering strategies and targeting the underlying liver disease. (106–109)

Nutritional supplementation

It is advised that any nutritional interventions follow the general recommendations reported as “energy and protein requirements in cirrhotic patients” (previous paragraph). However, an adequate calorie and protein intake is difficult to achieve in malnourished sarcopenic patients with advanced liver disease. Oral nutritional supplement and branched chain amino acid (BCAA) supplements have been utilised in clinical trials to overcome this issue(110, 111) showing some benefits. In patients with malnutrition and cirrhosis, who are unable to achieve adequate dietary intake with the oral diet (even with oral supplements), short-term enteral or parenteral nutrition should be to overcome the phase of underfeeding. Nutritional guidelines proposed by international medical societies for enteral and parenteral nutrition in patients with chronic liver disease are reported (Table S1).

Enteral feeding has been utilised in malnourished cirrhotic patients admitted to hospital, but despite promising individual studies, systematic meta-analyses have not shown significant benefits in terms of survival(11, 12, 112). There are also conflicting data on the benefits of parenteral nutritional supplementation in patients with cirrhosis, but this is likely to have a beneficial role during prolonged periods of poor oral intake including encephalopathy, gastrointestinal bleeding and impaired gut motility or ileus (113). The use of enteral and parenteral nutrition in the perioperative setting is dealt with in a dedicated section.

There is limited but consistent data that supplemental nutrition improves quality of life if it results in an increase in lean body mass, even though direct studies on sarcopenia are currently unavailable (114).

Exercise and physical activity

In addition to nutritional supplementation, increased physical activity and exercise are also anabolic stimuli that can improve muscle mass and function. However, consistent long-term data in cirrhosis are lacking (87, 115). Endurance or aerobic exercise improves skeletal muscle functional capacity but not necessarily muscle mass (116). Resistance exercise promotes an increase in skeletal muscle mass(116). However, exercise also increases muscle ammonia generation and portal pressure (117, 118), both of which can have adverse effects in cirrhotic patients. Despite these potential adverse responses, beneficial effects have been reported (103, 104). Since both muscle loss and impaired contractile function are components of sarcopenia in cirrhosis, a combination of resistance and endurance exercise would probably be appropriate and beneficial, as confirmed by emerging data indicating the benefit of a moderate intensity exercise regimen in cirrhosis (104).

Nutrient supplementation following physical activity is beneficial in physiological states, but whether such an intervention is beneficial in cirrhosis is currently unknown (119, 120). Continued impaired functional capacity and reduced peak oxygen consumption are associated with decreased survival and poor post-transplant outcomes(121, 122). Hence measures to increase functional capacity are likely to improve long-term clinical outcomes in cirrhosis(102).

Other strategies

Hormone replacement therapy utilising growth hormone or testosterone has been proposed but has not been consistently effective(91, 92, 123, 124). Furthermore caution is needed when using testosterone because of the possibility of increasing the risk of hepatocellular carcinoma(105).

A number of reports in preclinical models have shown that hyperammonaemia results in impaired protein synthesis and increased autophagy, both of which result in loss of muscle mass(99, 100).

Long-term ammonia-lowering strategies may result in increased muscle mass and contractile strength but the data are derived from preclinical studies and require validation in human studies (109).

Nutritional approach and management of obesity in patients with liver cirrhosis

Two studies have shown that obesity is at least as frequent in compensated cirrhosis as it is in the general population, ranging from 20 to 35% (13, 125), regardless of the origin of liver disease. In NASH-related cirrhosis obesity is present in most cases. Moreover, a sedentary lifestyle is highly prevalent in patients with cirrhosis and might be seen as a cofactor, leading to an increase in BW in this population. In the HALT-C trial (125) the risk of histological progression or decompensation increased by 14% for each increase in BMI quartile, and the risk of progression increased by 35% in patients whose BW increased by >5% at one year.

In a randomised controlled trial comparing the use of timolol or placebo to prevent the onset of gastroesophageal varices, BMI was associated with clinical decompensation, independently of portal hypertension and albumin, in patients with no varices and an hepatic venous pressure gradient ≥6 mmHg (13).

Data from different studies suggest that a reduction in BW improves outcomes in obese patients with compensated cirrhosis (102, 125, 126). This was achieved by a programme of lifestyle intervention including nutritional therapy and supervised moderate intensity physical exercise. A weight decrease ≥5–10% is considered an adequate goal, associated with a reduced rate of disease progression in patients included in the HALT-C trial (125). Dietary intake is aimed to guarantee both moderate caloric restriction and adequate protein intake. Indeed, although good quality data are lacking, particular attention must be paid to the protein intake needed to maintain muscle mass, because of the potential risk of exacerbating sarcopenia during weight loss interventions.

No clear-cut data is available regarding the best type of physical exercise (aerobic vs. anaerobic; endurance vs. resistance/strength training) and its duration in this population. In patients with portal hypertension, avoidance of abdominal pressure seems reasonable even though there is some data suggesting that resistance exercise is probably safe (126). Exercise needs to be tailored to the patient’s ability, beginning with moderate intensity and maintained for the long-term.

RecommendationsNutritional counselling by a multidisciplinary team should be provided to cirrhotic patients with malnutrition, helping patients to achieve adequate caloric and protein intake. (Grade II-2, C2)Optimal daily energy intake should not be lower than the recommended 35 kcal/kg.BW/d (in non-obese individuals). (Grade II-2, B1)Optimal daily protein intake should not be lower than the recommended 1.2–1.5 g/kg.BW/d. (Grade II-2, B1)Include late evening oral nutritional supplementation and breakfast in dietary regime of malnourished decompensated cirrhotic patients. (Grade II-1, B1)BCAA supplements and leucine enriched amino acid supplements should be used in decompensated cirrhotic patients to achieve adequate nitrogen intake. (Grade II-1, C1)In patients with malnutrition and cirrhosis who are unable to achieve adequate dietary intake with the oral diet (even with oral supplements), a period of enteral nutrition is recommended. (Grade II-1, B1)Patients with cirrhosis, whenever possible, should be encouraged to avoid hypomobility and to progressively increase physical activity. (Grade II-1, C2)Implement a nutritional and lifestyle programme to achieve progressive weight loss (>5–10%) in obese cirrhotic patients (BMI >30 kg/m2 corrected for water retention). (Grade II-2, C1)A tailored, moderately hypocaloric (−500–800 kcal/d) diet, including adequate protein intake (>1.5 g proteins/kg.BW/d) can be adopted to achieve weight loss without compromising protein stores in obese cirrhotic patients. (Grade II-1, C2)New research should answer the following topicsDoes the improvement in muscle mass and/or muscle function improve clinical outcomes (lower the risk of first decompensation, ascites, infection and encephalopathy, reduce hospital readmissions, decrease length of hospital stay, reduce risk of falls, improve survival)?Do ammonia-lowering strategies in decompensated cirrhosis reverse muscle loss and improve clinical outcomes?Does a gradual increase in physical activity delay or reverse muscle loss and contractile dysfunction? What type and duration of exercise are beneficial in cirrhotic patients need to be determinedIs the addition of supplements (leucine, isoleucine or other nutrient supplements) needed to lower ammonia and increase mitochondrial intermediates during training?How can therapies targeting the muscle protein synthesis pathway or dysregulated muscle autophagy be implemented?How can anabolic resistance be overcome, or the underlying causes of anabolic resistance in cirrhotic patients be reversed?Micronutrients

In general, vitamin deficiencies in liver disease are related to hepatic dysfunction, diminished reserves and, with increasing disease severity, inadequate dietary intake and malabsorption.

Fat-soluble vitamin deficiencies are common. A retrospective study reported that the majority of liver disease patients being considered for transplantation presented with vitamin A and D deficiencies (127).

The prevalence of vitamin D deficiency in the general population ranges from 20 to 100% when referring to serum 25(OH)D concentrations Open in a separate windowFigure 3Diagnosis and management of bone disease in patients with chronic liver disease.Table 3.

Risk factors for the development of osteoporosis in chronic liver disease.

● alcohol abuse● smoking● body mass index lower than 19 Kg/m2● male hypogonadism● early menopause● secondary amenorrhea of more than 6 months● family history of osteoporotic fracture● treatment with corticosteroids (5 mg/d or more of prednisone for 3 months or longer)● advanced ageOpen in a separate window

A balanced diet is recommended because chronic liver disease patients often are malnourished. Supplements of calcium (1,000–1,500 mg/d) and 25-hydroxy-vitamin D (400–800 IU/d or 260 µg every 2 weeks) or the dose required to preserve normal levels should be provided.- There is however no definite data confirming the efficacy of these supplements in preventing bone loss in patients with liver disease (134). Physical activity is recommended, in particular with exercises designed to improve the mechanics of the spine. Factors contributing to bone loss need to be reduced to a minimum, including discontinuation of alcohol and tobacco use. Corticosteroids ought to be minimized whenever possible.

Different pharmacological therapies have been proposed for improving bone mass in patients with chronic liver disease, but most studies have included small numbers of patients, and therefore it is difficult to reach any definite conclusions. Furthermore, no clear anti-fracture effect has been demonstrated, and except for osteoporosis in PBC and after liver transplantation, no systematic studies have been reported.

There is no general agreement concerning the appropriate time to start treatment but patients with established osteoporosis, and therefore with fragility fractures, should be treated to reduce the risk of further fractures. Since patients with PBC with a lumbar or a proximal femur T-score lower than 40 kg· m−2) prior to liver transplantation are associated with increased mortality and morbidity (226–229). Severe obesity prior to liver transplantation is associated with a higher prevalence of comorbidities (diabetes, hypertension), cryptogenic cirrhosis and increased mortality from infectious complications, cardiovascular disease and cancer (228, 229). Some investigators found that severe obesity was associated with increased morbidity and mortality even when patients were classified according to “dry BMI” (229) while others have reported that the amount of ascites and not BMI contributes to the increase mortality risk (230).

Numerous descriptive studies have shown higher morbidity and mortality in cirrhotic patients with protein malnutrition when such patients undergo liver transplantation (57, 231–233). Recently, sarcopenia and frailty have been shown to carry an increased risk of morbidity and mortality on the waiting list and after transplantation (58, 234–243). Patients on the wait are at risk due to an inadequate food or caloric intake (244) and those consuming a low protein diet (

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Clinical Practice Guideline Panel: Chair: Manuela Merli; Panel members: Shira Zelber-Sagi, Srinivasan Dasarathy, Sara Montagnese, Laurence Genton, Mathias Plauth, Albert Parés; EASL Governing Board representative: Annalisa Berzigotti

Supplementary Table 1. Nutritional guidelines for patients with chronic liver disease proposed by different medical societies in different settings.

Contributor Information

European Association for the Study of the Liver :

M Merli, A Berzigotti, S Zelber-Sagi, S Dasarathy, S Montagnese, L Genton, M Plauth, and A. ParésReferences1. Nutritional status in cirrhosis. Italian Multicentre Cooperative Project on Nutrition in Liver Cirrhosis. J Hepatol 1994;21:317–325. [PubMed] [Google Scholar]2. Caregaro L, Alberino F, Amodio P, Merkel C, Bolognesi M, Angeli P, Gatta A. Malnutrition in alcoholic and virus-related cirrhosis. The American journal of clinical nutrition 1996;63:602–609. [PubMed] [Google Scholar]3. Dasarathy S Consilience in sarcopenia of cirrhosis. J Cachexia Sarcopenia Muscle 2012;3:225–237. [PMC free article] [PubMed] [Google Scholar]4. Huisman EJ, Trip EJ, Siersema PD, van Hoek B, van Erpecum KJ. Protein energy malnutrition predicts complications in liver cirrhosis. Eur J Gastroenterol Hepatol 2011;23:982–989. [PubMed] [Google Scholar]5. Merli M, Lucidi C, Giannelli V, Giusto M, Riggio O, Falcone M, Ridola L, et al. Cirrhotic patients are at risk for health care-associated bacterial infections. Clin Gastroenterol Hepatol 2010;8:979–985. [PubMed] [Google Scholar]6. Merli M, Giusto M, Lucidi C, Giannelli V, Pentassuglio I, Di Gregorio V, Lattanzi B, et al. Muscle depletion increases the risk of overt and minimal hepatic encephalopathy: results of a prospective study. Metab Brain Dis 2013;28:281–284. [PubMed] [Google Scholar]7. Gunsar F, Raimondo ML, Jones S, Terreni N, Wong C, Patch D, Sabin C, et al. Nutritional status and prognosis in cirrhotic patients. Aliment Pharmacol Ther 2006;24:563–572. [PubMed] [Google Scholar]8. Montano-Loza AJ, Meza-Junco J, Prado CM, Lieffers JR, Baracos VE, Bain VG, Sawyer MB. Muscle wasting is associated with mortality in patients with cirrhosis. Clin Gastroenterol Hepatol 2012;10:166–173, 173 e161. [PubMed] [Google Scholar]9. Englesbe MJ, Patel SP, He K, Lynch RJ, Schaubel DE, Harbaugh C, Holcombe SA, et al. Sarcopenia and mortality after liver transplantation. J Am Coll Surg 2010;211:271–278. [PMC free article] [PubMed] [Google Scholar]10. Ney M, Vandermeer B, van Zanten SJ, Ma MM, Gramlich L, Tandon P. Meta-analysis: oral or enteral nutritional supplementation in cirrhosis. Aliment Pharmacol Ther 2013;37:672–679. [PubMed] [Google Scholar]11. Fialla AD, Israelsen M, Hamberg O, Krag A, Gluud LL. Nutritional therapy in cirrhosis or alcoholic hepatitis: a systematic review and meta-analysis. Liver Int 2015;35:2072–2078. [PubMed] [Google Scholar]12. Koretz RL, Avenell A, Lipman TO. Nutritional support for liver disease. Cochrane Database Syst Rev 2012:CD008344. [PMC free article] [PubMed]13. Berzigotti A, Garcia-Tsao G, Bosch J, Grace ND, Burroughs AK, Morillas R, Escorsell A, et al. Obesity is an independent risk factor for clinical decompensation in patients with cirrhosis. Hepatology 2011;54:555–561. [PMC free article] [PubMed] [Google Scholar]14. Montano-Loza AJ, Angulo P, Meza-Junco J, Prado CM, Sawyer MB, Beaumont C, Esfandiari N, et al. Sarcopenic obesity and myosteatosis are associated with higher mortality in patients with cirrhosis. J Cachexia Sarcopenia Muscle 2016;7:126–135. [PMC free article] [PubMed] [Google Scholar]15. Nishikawa H, Nishiguchi S. Sarcopenia and Sarcopenic Obesity Are Prognostic Factors for Overall Survival in Patients with Cirrhosis. Intern Med 2016;55:855–856. [PubMed] [Google Scholar]16. Andrews J, Guyatt G, Oxman AD, Alderson P, Dahm P, Falck-Ytter Y, Nasser M, et al. GRADE guidelines: 14. Going from evidence to recommendations: the significance and presentation of recommendations. J Clin Epidemiol 2013;66:719–725. [PubMed] [Google Scholar]17. Tandon P, Raman M, Mourtzakis M, Merli M. A practical approach to nutritional screening and assessment in cirrhosis. Hepatology 2017;65:1044–1057. [PubMed] [Google Scholar]18. Teitelbaum D, Guenter P, Howell WH, Kochevar ME, Roth J, Seidner DL. Definition of terms, style, and conventions used in A.S.P.E.N. guidelines and standards. Nutr Clin Pract 2005;20:281–285. [PubMed] [Google Scholar]19. Charney P Nutrition screening vs nutrition assessment: how do they differ? Nutr Clin Pract 2008;23:366–372. [PubMed] [Google Scholar]20. Cederholm T, Bosaeus I, Barazzoni R, Bauer J, Van Gossum A, Klek S, Muscaritoli M, et al. Diagnostic criteria for malnutrition - An ESPEN Consensus Statement. Clin Nutr 2015;34:335–340. [PubMed] [Google Scholar]21. Tandon P, Ney M, Irwin I, Ma MM, Gramlich L, Bain VG, Esfandiari N, et al. Severe muscle depletion in patients on the liver transplant wait list: its prevalence and independent prognostic value. Liver Transpl 2012;18:1209–1216. [PubMed] [Google Scholar]22. Borhofen SM, Gerner C, Lehmann J, Fimmers R, Gortzen J, Hey B, Geiser F, et al. The Royal Free Hospital-Nutritional Prioritizing Tool Is an Independent Predictor of Deterioration of Liver Function and Survival in Cirrhosis. Dig Dis Sci 2016;61:1735–1743. [PubMed] [Google Scholar]23. Booi AN, Menendez J, Norton HJ, Anderson WE, Ellis AC. Validation of a Screening Tool to Identify Undernutrition in Ambulatory Patients With Liver Cirrhosis. Nutr Clin Pract 2015;30:683–689. [PubMed] [Google Scholar]24. Cruz-Jentoft AJ, Baeyens JP, Bauer JM, Boirie Y, Cederholm T, Landi F, Martin FC, et al. Sarcopenia: European consensus on definition and diagnosis: Report of the European Working Group on Sarcopenia in Older People. Age Ageing 2010;39:412–423. [PMC free article] [PubMed] [Google Scholar]25. Peng S, Plank LD, McCall JL, Gillanders LK, McIlroy K, Gane EJ. Body composition, muscle function, and energy expenditure in patients with liver cirrhosis: a comprehensive study. Am J Clin Nutr 2007;85:1257–1266. [PubMed] [Google Scholar]26. Martin L, Birdsell L, Macdonald N, Reiman T, Clandinin MT, McCargar LJ, Murphy R, et al. Cancer cachexia in the age of obesity: skeletal muscle depletion is a powerful prognostic factor, independent of body mass index. J Clin Oncol 2013;31:1539–1547. [PubMed] [Google Scholar]27. Carey EJ, Lai JC, Wang CW, Dasarathy S, Lobach I, Montano-Loza AJ, Dunn MA, et al. A multicenter study to define sarcopenia in patients with end-stage liver disease. Liver Transpl 2017;23:625–633. [PMC free article] [PubMed] [Google Scholar]28. van Vugt JL, Levolger S, de Bruin RW, van Rosmalen J, Metselaar HJ, JN IJ. Systematic Review and Meta-Analysis of the Impact of Computed Tomography-Assessed Skeletal Muscle Mass on Outcome in Patients Awaiting or Undergoing Liver Transplantation. Am J Transplant 2016;16:2277–2292. [PubMed] [Google Scholar]29. Plauth M, Cabre E, Riggio O, Assis-Camilo M, Pirlich M, Kondrup J, Dgem, et al. ESPEN Guidelines on Enteral Nutrition: Liver disease. Clin Nutr 2006;25:285–294. [PubMed] [Google Scholar]30. Tandon P, Low G, Mourtzakis M, Zenith L, Myers RP, Abraldes JG, Shaheen AA, et al. A Model to Identify Sarcopenia in Patients With Cirrhosis. Clin Gastroenterol Hepatol 2016;14:1473–1480 e1473. [PubMed] [Google Scholar]31. Alberino F, Gatta A, Amodio P, Merkel C, Di Pascoli L, Boffo G, Caregaro L. Nutrition and survival in patients with liver cirrhosis. Nutrition 2001;17:445–450. [PubMed] [Google Scholar]32. Morgan MY, Madden AM, Soulsby CT, Morris RW. Derivation and validation of a new global method for assessing nutritional status in patients with cirrhosis. Hepatology 2006;44:823–835. [PubMed] [Google Scholar]33. Giusto M, Lattanzi B, Albanese C, Galtieri A, Farcomeni A, Giannelli V, Lucidi C, et al. Sarcopenia in liver cirrhosis: the role of computed tomography scan for the assessment of muscle mass compared with dual-energy X-ray absorptiometry and anthropometry. Eur J Gastroenterol Hepatol 2015;27:328–334. [PubMed] [Google Scholar]34. Wu LW, Lin YY, Kao TW, Lin CM, Liaw FY, Wang CC, Peng TC, et al. Mid-arm muscle circumference as a significant predictor of all-cause mortality in male individuals. PLoS One 2017;12:e0171707. [PMC free article] [PubMed] [Google Scholar]35. Morgan MY, Madden AM, Jennings G, Elia M, Fuller NJ. Two-component models are of limited value for the assessment of body composition in patients with cirrhosis. Am J Clin Nutr 2006;84:1151–1162. [PubMed] [Google Scholar]36. Alvares-da-Silva MR, Reverbel da Silveira T. Comparison between handgrip strength, subjective global assessment, and prognostic nutritional index in assessing malnutrition and predicting clinical outcome in cirrhotic outpatients. Nutrition 2005;21:113–117. [PubMed] [Google Scholar]37. Tandon P, Tangri N, Thomas L, Zenith L, Shaikh T, Carbonneau M, Ma M, et al. A Rapid Bedside Screen to Predict Unplanned Hospitalization and Death in Outpatients With Cirrhosis: A Prospective Evaluation of the Clinical Frailty Scale. Am J Gastroenterol 2016;111:1759–1767. [PubMed] [Google Scholar]38. Wang CW, Feng S, Covinsky KE, Hayssen H, Zhou LQ, Yeh BM, Lai JC. A Comparison of Muscle Function, Mass, and Quality in Liver Transplant Candidates: Results From the Functional Assessment in Liver Transplantation Study. Transplantation 2016;100:1692–1698. [PMC free article] [PubMed] [Google Scholar]39. Fried LP, Tangen CM, Walston J, Newman AB, Hirsch C, Gottdiener J, Seeman T, et al. Frailty in older adults: evidence for a phenotype. J Gerontol A Biol Sci Med Sci 2001;56:M146–156. [PubMed] [Google Scholar]40. Lai JC, Feng S, Terrault NA, Lizaola B, Hayssen H, Covinsky K. Frailty predicts waitlist mortality in liver transplant candidates. Am J Transplant 2014;14:1870–1879. [PMC free article] [PubMed] [Google Scholar]41. Hasse J, Strong S, Gorman MA, Liepa G. Subjective global assessment: alternative nutrition-assessment technique for liver-transplant candidates. Nutrition 1993;9:339–343. [PubMed] [Google Scholar]42. Bakshi N, Singh K. Nutrition assessment and its effect on various clinical variables among patients undergoing liver transplant. Hepatobiliary Surg Nutr 2016;5:358–371. [PMC free article] [PubMed] [Google Scholar]43. Ferreira LG, Anastacio LR, Lima AS, Correia MI. Assessment of nutritional status of patients waiting for liver transplantation. Clin Transplant 2011;25:248–254. [PubMed] [Google Scholar]44. Figueiredo FA, Dickson ER, Pasha TM, Porayko MK, Therneau TM, Malinchoc M, DiCecco SR, et al. Utility of standard nutritional parameters in detecting body cell mass depletion in patients with end-stage liver disease. Liver Transpl 2000;6:575–581. [PubMed] [Google Scholar]45. Fernandes SA, Bassani L, Nunes FF, Aydos ME, Alves AV, Marroni CA. Nutritional assessment in patients with cirrhosis. Arq Gastroenterol 2012;49:19–27. [PubMed] [Google Scholar]46. Figueiredo FA, Perez RM, Freitas MM, Kondo M. Comparison of three methods of nutritional assessment in liver cirrhosis: subjective global assessment, traditional nutritional parameters, and body composition analysis. J Gastroenterol 2006;41:476–482. [PubMed] [Google Scholar]47. Naveau S, Belda E, Borotto E, Genuist F, Chaput JC. Comparison of clinical judgment and anthropometric parameters for evaluating nutritional status in patients with alcoholic liver disease. J Hepatol 1995;23:234–235. [PubMed] [Google Scholar]48. Sasidharan M, Nistala S, Narendhran RT, Murugesh M, Bhatia SJ, Rathi PM. Nutritional status and prognosis in cirrhotic patients. Trop Gastroenterol 2012;33:257–264. [PubMed] [Google Scholar]49. Kalafateli M, Mantzoukis K, Choi Yau Y, Mohammad AO, Arora S, Rodrigues S, de Vos M, et al. Malnutrition and sarcopenia predict post-liver transplantation outcomes independently of the Model for End-stage Liver Disease score. J Cachexia Sarcopenia Muscle 2017;8:113–121. [PMC free article] [PubMed] [Google Scholar]50. Gabrielson DK, Scaffidi D, Leung E, Stoyanoff L, Robinson J, Nisenbaum R, Brezden-Masley C, et al. Use of an abridged scored Patient-Generated Subjective Global Assessment (abPG-SGA) as a nutritional screening tool for cancer patients in an outpatient setting. Nutr Cancer 2013;65:234–239. [PubMed] [Google Scholar]51. De Keyzer W, Huybrechts I, De Vriendt V, Vandevijvere S, Slimani N, Van Oyen H, De Henauw S. Repeated 24-hour recalls versus dietary records for estimating nutrient intakes in a national food consumption survey. Food Nutr Res 2011;55. [PMC free article] [PubMed]52. Ahluwalia N, Dwyer J, Terry A, Moshfegh A, Johnson C. Update on NHANES Dietary Data: Focus on Collection, Release, Analytical Considerations, and Uses to Inform Public Policy. Adv Nutr 2016;7:121–134. [PMC free article] [PubMed] [Google Scholar]53. Detsky AS, McLaughlin JR, Baker JP, Johnston N, Whittaker S, Mendelson RA, Jeejeebhoy KN. What is subjective global assessment of nutritional status? JPEN J Parenter Enteral Nutr 1987;11:8–13. [PubMed] [Google Scholar]54. Carias S, Castellanos AL, Vilchez V, Nair R, Dela Cruz AC, Watkins J, Barrett T, et al. Nonalcoholic steatohepatitis is strongly associated with sarcopenic obesity in patients with cirrhosis undergoing liver transplant evaluation. J Gastroenterol Hepatol 2016;31:628–633. [PMC free article] [PubMed] [Google Scholar]55. Choudhary NS, Saigal S, Saraf N, Mohanka R, Rastogi A, Goja S, Menon PB, et al. Sarcopenic obesity with metabolic syndrome: a newly recognized entity following living donor liver transplantation. Clin Transplant 2015;29:211–215. [PubMed] [Google Scholar]56. Dasarathy S Posttransplant sarcopenia: an underrecognized early consequence of liver transplantation. Dig Dis Sci 2013;58:3103–3111. [PMC free article] [PubMed] [Google Scholar]57. Merli M, Giusto M, Gentili F, Novelli G, Ferretti G, Riggio O, Corradini SG, et al. Nutritional status: its influence on the outcome of patients undergoing liver transplantation. Liver International 2010;30:208–214. [PubMed] [Google Scholar]58. Englesbe MJ, Patel SP, He K, Lynch RJ, Schaubel DE, Harbaugh C, Holcombe SA, et al. Sarcopenia and mortality after liver transplantation. Journal of the American College of Surgeons 2010;211:271–278. [PMC free article] [PubMed] [Google Scholar]59. Ney M, Vandermeer B, Van Zanten S, Ma M, Gramlich L, Tandon P. Meta-analysis: oral or enteral nutritional supplementation in cirrhosis. Alimentary pharmacology & therapeutics 2013;37:672–679. [PubMed] [Google Scholar]60. Merli M, Riggio O, Dally L. Does malnutrition affect survival in cirrhosis? PINC (Policentrica Italiana Nutrizione Cirrosi). Hepatology 1996;23:1041–1046. [PubMed] [Google Scholar]61. Glass C, Hipskind P, Tsien C, Malin SK, Kasumov T, Shah SN, Kirwan JP, et al. Sarcopenia and a physiologically low respiratory quotient in patients with cirrhosis: a prospective controlled study. J Appl Physiol (1985) 2013;114:559–565. [PMC free article] [PubMed] [Google Scholar]62. Glass C, Hipskind P, Cole D, Lopez R, Dasarathy S. Handheld calorimeter is a valid instrument to quantify resting energy expenditure in hospitalized cirrhotic patients: a prospective study. Nutr Clin Pract 2012;27:677–688. [PMC free article] [PubMed] [Google Scholar]63. Dasarathy S, Merli M. Sarcopenia from mechanism to diagnosis and treatment in liver disease. J Hepatol 2016;65:1232–1244. [PMC free article] [PubMed] [Google Scholar]64. Dasarathy S. Nutrition and Alcoholic Liver Disease: Effects of Alcoholism on Nutrition, Effects of Nutrition on Alcoholic Liver Disease, and Nutritional Therapies for Alcoholic Liver Disease. Clin Liver Dis 2016;20:535–550. [PMC free article] [PubMed] [Google Scholar]65. Dasarathy S. Cause and management of muscle wasting in chronic liver disease. Curr Opin Gastroenterol 2016;32:159–165. [PMC free article] [PubMed] [Google Scholar]66. Dasarathy J, McCullough AJ, Dasarathy S. Sarcopenia in Alcoholic Liver Disease: Clinical and Molecular Advances. Alcohol Clin Exp Res 2017;41:1419–1431. [PMC free article] [PubMed] [Google Scholar]67. Greco AV, Mingrone G, Benedetti G, Capristo E, Tataranni PA, Gasbarrini G. Daily energy and substrate metabolism in patients with cirrhosis. Hepatology 1998;27:346–350. [PubMed] [Google Scholar]68. Guglielmi FW, Panella C, Buda A, Budillon G, Caregaro L, Clerici C, Conte D, et al. Nutritional state and energy balance in cirrhotic patients with or without hypermetabolism. Multicentre prospective study by the ‘Nutritional Problems in Gastroenterology’ Section of the Italian Society of Gastroenterology (SIGE). Dig Liver Dis 2005;37:681–688. [PubMed] [Google Scholar]69. Riggio O, Angeloni S, Ciuffa L, Nicolini G, Attili AF, Albanese C, Merli M. Malnutrition is not related to alterations in energy balance in patients with stable liver cirrhosis. Clin Nutr 2003;22:553–559. [PubMed] [Google Scholar]70. Nielsen K, Kondrup J, Martinsen L, Dossing H, Larsson B, Stilling B, Jensen MG. Long-term oral refeeding of patients with cirrhosis of the liver. Br J Nutr 1995;74:557–567. [PubMed] [Google Scholar]71. Dolz C, Raurich JM, Ibanez J, Obrador A, Marse P, Gaya J. Ascites increases the resting energy expenditure in liver cirrhosis. Gastroenterology 1991;100:738–744. [PubMed] [Google Scholar]72. Madden AM, Morgan MY. Resting energy expenditure should be measured in patients with cirrhosis, not predicted. Hepatology 1999;30:655–664. [PubMed] [Google Scholar]73. Tajika M, Kato M, Mohri H, Miwa Y, Kato T, Ohnishi H, Moriwaki H. Prognostic value of energy metabolism in patients with viral liver cirrhosis. Nutrition 2002;18:229–234. [PubMed] [Google Scholar]74. Knudsen AW, Krag A, Nordgaard-Lassen I, Frandsen E, Tofteng F, Mortensen C, Becker U. Effect of paracentesis on metabolic activity in patients with advanced cirrhosis and ascites. Scand J Gastroenterol 2016;51:601–609. [PubMed] [Google Scholar]75. Muller MJ, Bottcher J, Selberg O, Weselmann S, Boker KH, Schwarze M, von zur Muhlen A, et al. Hypermetabolism in clinically stable patients with liver cirrhosis. Am J Clin Nutr 1999;69:1194–1201. [PubMed] [Google Scholar]76. Hipskind P, Glass C, Charlton D, Nowak D, Dasarathy S. Do handheld calorimeters have a role in assessment of nutrition needs in hospitalized patients? A systematic review of literature. Nutr Clin Pract 2011;26:426–433. [PMC free article] [PubMed] [Google Scholar]77. Iwasa M, Iwata K, Hara N, Hattori A, Ishidome M, Sekoguchi-Fujikawa N, Mifuji-Moroka R, et al. Nutrition therapy using a multidisciplinary team improves survival rates in patients with liver cirrhosis. Nutrition 2013;29:1418–1421. [PubMed] [Google Scholar]78. Tsien CD, McCullough AJ, Dasarathy S. Late evening snack: exploiting a period of anabolic opportunity in cirrhosis. J Gastroenterol Hepatol 2012;27:430–441. [PubMed] [Google Scholar]79. Vaisman N, Katzman H, Carmiel-Haggai M, Lusthaus M, Niv E. Breakfast improves cognitive function in cirrhotic patients with cognitive impairment. Am J Clin Nutr 2010;92:137–140. [PubMed] [Google Scholar]80. Plank LD, Gane EJ, Peng S, Muthu C, Mathur S, Gillanders L, McIlroy K, et al. Nocturnal nutritional supplementation improves total body protein status of patients with liver cirrhosis: a randomized 12-month trial. Hepatology 2008;48:557–566. [PubMed] [Google Scholar]81. Nielsen K, Kondrup J, Martinsen L, Stilling B, Wikman B. Nutritional assessment and adequacy of dietary intake in hospitalized patients with alcoholic liver cirrhosis. Br J Nutr 1993;69:665–679. [PubMed] [Google Scholar]82. Swart GR, van den Berg JW, van Vuure JK, Rietveld T, Wattimena DL, Frenkel M. Minimum protein requirements in liver cirrhosis determined by nitrogen balance measurements at three levels of protein intake. Clin Nutr 1989;8:329–336. [PubMed] [Google Scholar]83. Fenton JC, Knight EJ, Humpherson PL. Milk-and-cheese diet in portal-systemic encephalopathy. Lancet 1966;1:164–166. [PubMed] [Google Scholar]84. Bianchi GP, Marchesini G, Fabbri A, Rondelli A, Bugianesi E, Zoli M, Pisi E. Vegetable versus animal protein diet in cirrhotic patients with chronic encephalopathy. A randomized cross-over comparison. J Intern Med 1993;233:385–392. [PubMed] [Google Scholar]85. Gheorghe L, Iacob R, Vadan R, Iacob S, Gheorghe C. Improvement of hepatic encephalopathy using a modified high-calorie high-protein diet. Rom J Gastroenterol 2005;14:231–238. [PubMed] [Google Scholar]86. Cordoba J, Lopez-Hellin J, Planas M, Sabin P, Sanpedro F, Castro F, Esteban R, et al. Normal protein diet for episodic hepatic encephalopathy: results of a randomized study. J Hepatol 2004;41:38–43. [PubMed] [Google Scholar]87. Rennie MJ, Tipton KD. Protein and amino acid metabolism during and after exercise and the effects of nutrition. Annu Rev Nutr 2000;20:457–483. [PubMed] [Google Scholar]88. Periyalwar P, Dasarathy S. Malnutrition in cirrhosis: contribution and consequences of sarcopenia on metabolic and clinical responses. Clin Liver Dis 2012;16:95–131. [PMC free article] [PubMed] [Google Scholar]89. Tessari P Protein metabolism in liver cirrhosis: from albumin to muscle myofibrils. Curr Opin Clin Nutr Metab Care 2003;6:79–85. [PubMed] [Google Scholar]90. DiCecco SR, Wieners EJ, Wiesner RH, Southorn PA, Plevak DJ, Krom RA. Assessment of nutritional status of patients with end-stage liver disease undergoing liver transplantation. Mayo Clin Proc 1989;64:95–102. [PubMed] [Google Scholar]91. Sinclair M, Grossmann M, Hoermann R, Angus PW, Gow PJ. Testosterone therapy increases muscle mass in men with cirrhosis and low testosterone: A randomised controlled trial. J Hepatol 2016;65:906–913. [PubMed] [Google Scholar]92. Assy N, Hochberg Z, Amit T, Shen-Orr Z, Enat R, Baruch Y. Growth hormone-stimulated insulin-like growth factor (IGF) I and IGF-binding protein-3 in liver cirrhosis. J Hepatol 1997;27:796–802. [PubMed] [Google Scholar]93. Chen HW, Dunn MA. Muscle at Risk: The Multiple Impacts of Ammonia on Sarcopenia and Frailty in Cirrhosis. Clin Transl Gastroenterol 2016;7:e170. [PMC free article] [PubMed] [Google Scholar]94. Dam G, Ott P, Aagaard NK, Vilstrup H. Branched-chain amino acids and muscle ammonia detoxification in cirrhosis. Metab Brain Dis 2013;28:217–220. [PubMed] [Google Scholar]95. Tsien C, Davuluri G, Singh D, Allawy A, Ten Have GA, Thapaliya S, Schulze JM, et al. Metabolic and molecular responses to leucine-enriched branched chain amino acid supplementation in the skeletal muscle of alcoholic cirrhosis. Hepatology 2015;61:2018–2029. [PMC free article] [PubMed] [Google Scholar]96. Nishikawa H, Enomoto H, Ishii A, Iwata Y, Miyamoto Y, Ishii N, Yuri Y, et al. Elevated serum myostatin level is associated with worse survival in patients with liver cirrhosis. J Cachexia Sarcopenia Muscle 2017. [PMC free article] [PubMed]97. Holecek M Branched-chain amino acid supplementation in treatment of liver cirrhosis: Updated views on how to attenuate their harmful effects on cataplerosis and ammonia formation. Nutrition 2017;41:80–85. [PubMed] [Google Scholar]98. Dasarathy S, McCullough AJ, Muc S, Schneyer A, Bennett CD, Dodig M, Kalhan SC. Sarcopenia associated with portosystemic shunting is reversed by follistatin. J Hepatol 2011;54:915–921. [PMC free article] [PubMed] [Google Scholar]99. Qiu J, Thapaliya S, Runkana A, Yang Y, Tsien C, Mohan ML, Narayanan A, et al. Hyperammonemia in cirrhosis induces transcriptional regulation of myostatin by an NF-kappaB-mediated mechanism. Proc Natl Acad Sci U S A 2013;110:18162–18167. [PMC free article] [PubMed] [Google Scholar]100. Qiu J, Tsien C, Thapalaya S, Narayanan A, Weihl CC, Ching JK, Eghtesad B, et al. Hyperammonemia-mediated autophagy in skeletal muscle contributes to sarcopenia of cirrhosis. Am J Physiol Endocrinol Metab 2012;303:E983–993. [PMC free article] [PubMed] [Google Scholar]101. Thapaliya S, Runkana A, McMullen MR, Nagy LE, McDonald C, Naga Prasad SV, Dasarathy S. Alcohol-induced autophagy contributes to loss in skeletal muscle mass. Autophagy 2014;10:677–690. [PMC free article] [PubMed] [Google Scholar]102. Zenith L, Meena N, Ramadi A, Yavari M, Harvey A, Carbonneau M, Ma M, et al. Eight weeks of exercise training increases aerobic capacity and muscle mass and reduces fatigue in patients with cirrhosis. Clin Gastroenterol Hepatol 2014;12:1920–1926 e1922. [PubMed] [Google Scholar]103. Berzigotti A, Saran U, Dufour JF. Physical activity and liver diseases. Hepatology 2016;63:1026–1040. [PubMed] [Google Scholar]104. Berzigotti A, Albillos A, Villanueva C, Genesca J, Ardevol A, Augustin S, Calleja JL, et al. Effects of an intensive lifestyle intervention program on portal hypertension in patients with cirrhosis and obesity: The SportDiet study. Hepatology 2017;65:1293–1305. [PubMed] [Google Scholar]105. Nagasue N, Yukaya H, Chang YC, Ogawa Y, Kohno H, Ito A. Active uptake of testosterone by androgen receptors of hepatocellular carcinoma in humans. Cancer 1986;57:2162–2167. [PubMed] [Google Scholar]106. Gorostiaga EM, Navarro-Amezqueta I, Calbet JA, Sanchez-Medina L, Cusso R, Guerrero M, Granados C, et al. Blood ammonia and lactate as markers of muscle metabolites during leg press exercise. J Strength Cond Res 2014;28:2775–2785. [PubMed] [Google Scholar]107. Takeda K, Takemasa T. Expression of ammonia transporters Rhbg and Rhcg in mouse skeletal muscle and the effect of 6-week training on these proteins. Physiol Rep 2015;3. [PMC free article] [PubMed]108. McDaniel J, Davuluri G, Hill EA, Moyer M, Runkana A, Prayson R, van Lunteren E, et al. Hyperammonemia results in reduced muscle function independent of muscle mass. Am J Physiol Gastrointest Liver Physiol 2016;310:G163–170. [PMC free article] [PubMed] [Google Scholar]109. Kumar A, Davuluri G, Silva RNE, Engelen M, Ten Have GAM, Prayson R, Deutz NEP, et al. Ammonia lowering reverses sarcopenia of cirrhosis by restoring skeletal muscle proteostasis. Hepatology 2017;65:2045–2058. [PMC free article] [PubMed] [Google Scholar]110. Nakaya Y, Harada N, Kakui S, Okada K, Takahashi A, Inoi J, Ito S. Severe catabolic state after prolonged fasting in cirrhotic patients: effect of oral branched-chain amino-acid-enriched nutrient mixture. Journal of gastroenterology 2002;37:531–536. [PubMed] [Google Scholar]111. Yoshida T, Muto Y, Moriwaki H, Yamato M. Effect of long-term oral supplementation with branched-chain amino acid granules on the prognosis of liver cirrhosis. Gastroenterol Jpn 1989;24:692–698. [PubMed] [Google Scholar]112. Antar R, Wong P, Ghali P. A meta-analysis of nutritional supplementation for management of hospitalized alcoholic hepatitis. Can J Gastroenterol 2012;26:463–467. [PMC free article] [PubMed] [Google Scholar]113. Plauth M, Cabre E, Campillo B, Kondrup J, Marchesini G, Schutz T, Shenkin A, et al. ESPEN Guidelines on Parenteral Nutrition: hepatology. Clin Nutr 2009;28:436–444. [PubMed] [Google Scholar]114. Maharshi S, Sharma BC, Sachdeva S, Srivastava S, Sharma P. Efficacy of nutritional therapy for patients with cirrhosis and minimal hepatic encephalopathy in a randomized trial. Clinical Gastroenterology and Hepatology 2016;14:454–460. e453. [PubMed] [Google Scholar]115. Liao CD, Tsauo JY, Wu YT, Cheng CP, Chen HC, Huang YC, Chen HC, et al. Effects of protein supplementation combined with resistance exercise on body composition and physical function in older adults: a systematic review and meta-analysis. Am J Clin Nutr 2017;106:1078–1091. [PubMed] [Google Scholar]116. Baar K Training for endurance and strength: lessons from cell signaling. Med Sci Sports Exerc 2006;38:1939–1944. [PubMed] [Google Scholar]117. Dietrich R, Bachmann C, Lauterburg BH. Exercise-induced hyperammonemia in patients with compensated chronic liver disease. Scand J Gastroenterol 1990;25:329–334. [PubMed] [Google Scholar]118. Garcia-Pagan JC, Santos C, Barbera JA, Luca A, Roca J, Rodriguez-Roisin R, Bosch J, et al. Physical exercise increases portal pressure in patients with cirrhosis and portal hypertension. Gastroenterology 1996;111:1300–1306. [PubMed] [Google Scholar]119. Schoenfeld BJ, Aragon AA, Krieger JW. The effect of protein timing on muscle strength and hypertrophy: a meta-analysis. J Int Soc Sports Nutr 2013;10:53. [PMC free article] [PubMed] [Google Scholar]120. Beale DJ. Evidence inconclusive - comment on article by Schoenfeld et al. J Int Soc Sports Nutr 2016;13:37. [PMC free article] [PubMed] [Google Scholar]121. Jones JC, Coombes JS, Macdonald GA. Exercise capacity and muscle strength in patients with cirrhosis. Liver Transpl 2012;18:146–151. [PubMed] [Google Scholar]122. Dharancy S, Lemyze M, Boleslawski E, Neviere R, Declerck N, Canva V, Wallaert B, et al. Impact of impaired aerobic capacity on liver transplant candidates. Transplantation 2008;86:1077–1083. [PubMed] [Google Scholar]123. Matsumoto R, Fukuoka H, Iguchi G, Nishizawa H, Bando H, Suda K, Takahashi M, et al. Long-term effects of growth hormone replacement therapy on liver function in adult patients with growth hormone deficiency. Growth Horm IGF Res 2014;24:174–179. [PubMed] [Google Scholar]124. Sinclair M, Gow PJ, Grossmann M, Angus PW. Review article: sarcopenia in cirrhosis--aetiology, implications and potential therapeutic interventions. Aliment Pharmacol Ther 2016;43:765–777. [PubMed] [Google Scholar]125. Everhart JE, Lok AS, Kim HY, Morgan TR, Lindsay KL, Chung RT, Bonkovsky HL, et al. Weight-related effects on disease progression in the hepatitis C antiviral long-term treatment against cirrhosis trial. Gastroenterology 2009;137:549–557. [PMC free article] [PubMed] [Google Scholar]126. Macias-Rodriguez RU, Ilarraza-Lomeli H, Ruiz-Margain A, Ponce-de-Leon-Rosales S, Vargas-Vorackova F, Garcia-Flores O, Torre A, et al. Changes in Hepatic Venous Pressure Gradient Induced by Physical Exercise in Cirrhosis: Results of a Pilot Randomized Open Clinical Trial. Clin Transl Gastroenterol 2016;7:e180. [PMC free article] [PubMed] [Google Scholar]127. Venu M, Martin E, Saeian K, Gawrieh S. High prevalence of vitamin A deficiency and vitamin D deficiency in patients evaluated for liver transplantation. Liver Transpl 2013;19:627–633. [PMC free article] [PubMed] [Google Scholar]128. Holick MF, Binkley NC, Bischoff-Ferrari HA, Gordon CM, Hanley DA, Heaney RP, Murad MH, et al. Evaluation, treatment, and prevention of vitamin D deficiency: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab 2011;96:1911–1930. [PubMed] [Google Scholar]129. Trautwein C, Possienke M, Schlitt HJ, Boker KH, Horn R, Raab R, Manns MP, et al. Bone density and metabolism in patients with viral hepatitis and cholestatic liver diseases before and after liver transplantation. Am J Gastroenterol 2000;95:2343–2351. [PubMed] [Google Scholar]130. Stokes CS, Volmer DA, Grunhage F, Lammert F. Vitamin D in chronic liver disease. Liver Int 2013;33:338–352. [PubMed] [Google Scholar]131. Barchetta I, Angelico F, Del Ben M, Baroni MG, Pozzilli P, Morini S, Cavallo MG. Strong association between non alcoholic fatty liver disease (NAFLD) and low 25(OH) vitamin D levels in an adult population with normal serum liver enzymes. BMC Med 2011;9:85. [PMC free article] [PubMed] [Google Scholar]132. Petta S, Camma C, Scazzone C, Tripodo C, Di Marco V, Bono A, Cabibi D, et al. Low vitamin D serum level is related to severe fibrosis and low responsiveness to interferon-based therapy in genotype 1 chronic hepatitis C. Hepatology 2010;51:1158–1167. [PubMed] [Google Scholar]133. European Association for the Study of the L. EASL Clinical Practice Guidelines: management of cholestatic liver diseases. J Hepatol 2009;51:237–267. [PubMed] [Google Scholar]134. Dasarathy J, Varghese R, Feldman A, Khiyami A, McCullough AJ, Dasarathy S. Patients with Nonalcoholic Fatty Liver Disease Have a Low Response Rate to Vitamin D Supplementation. J Nutr 2017. [PMC free article] [PubMed]135. Kril JJ, Butterworth RF. Diencephalic and cerebellar pathology in alcoholic and nonalcoholic patients with end-stage liver disease. Hepatology 1997;26:837–841. [PubMed] [Google Scholar]136. Bemeur C, Butterworth RF. Nutrition in the management of cirrhosis and its neurological complications. J Clin Exp Hepatol 2014;4:141–150. [PMC free article] [PubMed] [Google Scholar]137. Cosgray RE, Hanna V, Davidhizar RE, Smith J. The water-intoxicated patient. Arch Psychiatr Nurs 1990;4:308–312. [PubMed] [Google Scholar]138. Kleinschmidt-DeMasters BK, Norenberg MD. Rapid correction of hyponatremia causes demyelination: relation to central pontine myelinolysis. Science 1981;211:1068–1070. [PubMed] [Google Scholar]139. European Association for the Study of the L. EASL clinical practice guidelines on the management of ascites, spontaneous bacterial peritonitis, and hepatorenal syndrome in cirrhosis. J Hepatol 2010;53:397–417. [PubMed] [Google Scholar]140. Gu XB, Yang XJ, Zhu HY, Xu BY. Effect of a diet with unrestricted sodium on ascites in patients with hepatic cirrhosis. Gut Liver 2012;6:355–361. [PMC free article] [PubMed] [Google Scholar]141. Morando F, Rosi S, Gola E, Nardi M, Piano S, Fasolato S, Stanco M, et al. Adherence to a moderate sodium restriction diet in outpatients with cirrhosis and ascites: a real-life cross-sectional study. Liver Int 2015;35:1508–1515. [PubMed] [Google Scholar]142. Huskisson E, Maggini S, Ruf M. The influence of micronutrients on cognitive function and performance. J Int Med Res 2007;35:1–19. [PubMed] [Google Scholar]143. Bresci G, Parisi G, Banti S. Management of hepatic encephalopathy with oral zinc supplementation: a long-term treatment. Eur J Med 1993;2:414–416. [PubMed] [Google Scholar]144. Takuma Y, Nouso K, Makino Y, Hayashi M, Takahashi H. Clinical trial: oral zinc in hepatic encephalopathy. Aliment Pharmacol Ther 2010;32:1080–1090. [PubMed] [Google Scholar]145. Katayama K, Saito M, Kawaguchi T, Endo R, Sawara K, Nishiguchi S, Kato A, et al. Effect of zinc on liver cirrhosis with hyperammonemia: a preliminary randomized, placebo-controlled double-blind trial. Nutrition 2014;30:1409–1414. [PubMed] [Google Scholar]146. Himoto T, Yoneyama H, Kurokohchi K, Inukai M, Masugata H, Goda F, Haba R, et al. Selenium deficiency is associated with insulin resistance in patients with hepatitis C virus-related chronic liver disease. Nutr Res 2011;31:829–835. [PubMed] [Google Scholar]147. Inoue E, Hori S, Narumi Y, Fujita M, Kuriyama K, Kadota T, Kuroda C. Portal-systemic encephalopathy: presence of basal ganglia lesions with high signal intensity on MR images. Radiology 1991;179:551–555. [PubMed] [Google Scholar]148. Thompson J, Schafer D, Haun J, Schafer G. Adequate diet prevents hepatic coma in dogs with Eck fistulas. Surgery, gynecology & obstetrics 1986;162:126–130. [PubMed] [Google Scholar]149. Kalaitzakis E, Olsson R, Henfridsson P, Hugosson I, Bengtsson M, Jalan R, Björnsson E. Malnutrition and diabetes mellitus are related to hepatic encephalopathy in patients with liver cirrhosis. Liver International 2007;27:1194–1201. [PubMed] [Google Scholar]150. Merli M, Giusto M, Lucidi C, Giannelli V, Pentassuglio I, Di Gregorio V, Lattanzi B, et al. Muscle depletion increases the risk of overt and minimal hepatic encephalopathy: results of a prospective study. Metabolic brain disease 2013;28:281–284. [PubMed] [Google Scholar]151. Nardelli S, Lattanzi B, Torrisi S, Greco F, Farcomeni A, Gioia S, Merli M, et al. Sarcopenia is risk factor for development of hepatic encephalopathy after transjugular intrahepatic portosystemic shunt placement. Clinical Gastroenterology and Hepatology 2017;15:934–936. [PubMed] [Google Scholar]152. Olde Damink SW, Jalan R, Deutz NE, Redhead DN, Dejong CH, Hynd P, Jalan RA, et al. The kidney plays a major role in the hyperammonemia seen after simulated or actual GI bleeding in patients with cirrhosis. Hepatology 2003;37:1277–1285. [PubMed] [Google Scholar]153. Chatauret N, Desjardins P, Zwingmann C, Rose C, Rao KR, Butterworth RF. Direct molecular and spectroscopic evidence for increased ammonia removal capacity of skeletal muscle in acute liver failure. Journal of hepatology 2006;44:1083–1088. [PubMed] [Google Scholar]154. McDaniel J, Davuluri G, Hill EA, Moyer M, Runkana A, Prayson R, van Lunteren E, et al. Hyperammonemia results in reduced muscle function independent of muscle mass. American Journal of Physiology-Gastrointestinal and Liver Physiology 2016;310:G163–G170. [PMC free article] [PubMed] [Google Scholar]155. Kumar A, Davuluri G, Engelen MP, Ten Have GA, Prayson R, Deutz NE, Dasarathy S. Ammonia lowering reverses sarcopenia of cirrhosis by restoring skeletal muscle proteostasis. Hepatology 2017;65:2045–2058. [PMC free article] [PubMed] [Google Scholar]156. Amodio P, Bemeur C, Butterworth R, Cordoba J, Kato A, Montagnese S, Uribe M, et al. The nutritional management of hepatic encephalopathy in patients with cirrhosis: International Society for Hepatic Encephalopathy and Nitrogen Metabolism Consensus. Hepatology 2013;58:325–336. [PubMed] [Google Scholar]157. Schwartz R, Phillips GB, Seegmiller JE, Gabuzda GJ Jr, Davidson CS. Dietary protein in the genesis of hepatic coma. New England Journal of Medicine 1954;251:685–689. [PubMed] [Google Scholar]158. Summerskill W, Wolfe SJ, Davidson CS. The management of hepatic coma in relation to protein withdrawal and certain specific measures. The American journal of medicine 1957;23:59–76. [PubMed] [Google Scholar]159. Soulsby CT, Morgan MY. Dietary management of hepatic encephalopathy in cirrhotic patients: survey of current practice in United Kingdom. Bmj 1999;318:1391–1391. [PMC free article] [PubMed] [Google Scholar]160. Campollo O, Sprengers D, Dam G, Vilstrup H, McIntyre N. Protein tolerance to standard and high protein meals in patients with liver cirrhosis. World J Hepatol 2017;9:667–676. [PMC free article] [PubMed] [Google Scholar]161. Condon RE. Effect of dietary protein on symptoms and survival in dogs with an Eck fistula. The American Journal of Surgery 1971;121:107–114. [PubMed] [Google Scholar]162. Bessman AN, Mirick GS. Blood ammonia levels following the ingestion of casein and whole blood. Journal of Clinical Investigation 1958;37:990. [PMC free article] [PubMed] [Google Scholar]163. Fenton J, Knight E, Humpherson P. Milk-and-cheese diet in portal-systemic encephalopathy. The Lancet 1966;287:164–166. [PubMed] [Google Scholar]164. Greenberger NJ, Carley J, Schenker S, Bettinger I, Stamnes C, Beyer P. Effect of vegetable and animal protein diets in chronic hepatic encephalopathy. Digestive Diseases and Sciences 1977;22:845–855. [PubMed] [Google Scholar]165. Amodio P, Caregaro L, Patteno E, Marcon M, Del Piccolo F, Gatta A. Vegetarian diets in hepatic encephalopathy: facts or fantasies? Dig Liver Dis 2001;33:492–500. [PubMed] [Google Scholar]166. Gheorghe L, Iacob R, Vadan R, Iacob S, Gheorghe C. Improvement of hepatic encephalopathy using a modified high-calorie high-protein diet. Rom J Gastroenterol 2005;14:231–238. [PubMed] [Google Scholar]167. Uribe M, Dibildox M, Malpica S, Guillermo E, Villallobos A, Nieto L, Vargas F, et al. Beneficial effect of vegetable protein diet supplemented with psyllium plantago in patients with hepatic encephalopathy and diabetes mellitus. Gastroenterology 1985;88:901–907. [PubMed] [Google Scholar]168. Kawaguchi T, Izumi N, Charlton MR, Sata M. Branched-chain amino acids as pharmacological nutrients in chronic liver disease. Hepatology 2011;54:1063–1070. [PubMed] [Google Scholar]169. Holecek M Three targets of branched-chain amino acid supplementation in the treatment of liver disease. Nutrition 2010;26:482–490. [PubMed] [Google Scholar]170. Dam G, Ott P, Aagaard NK, Vilstrup H. Branched-chain amino acids and muscle ammonia detoxification in cirrhosis. Metabolic brain disease 2013;28:217–220. [PubMed] [Google Scholar]171. Marchesini G, Bianchi G, Merli M, Amodio P, Panella C, Loguercio C, Fanelli FR, et al. Nutritional supplementation with branched-chain amino acids in advanced cirrhosis: a double-blind, randomized trial. Gastroenterology 2003;124:1792–1801. [PubMed] [Google Scholar]172. Davuluri G, Krokowski D, Guan BJ, Kumar A, Thapaliya S, Singh D, Hatzoglou M, et al. Metabolic adaptation of skeletal muscle to hyperammonemia drives the beneficial effects of l-leucine in cirrhosis. J Hepatol 2016;65:929–937. [PMC free article] [PubMed] [Google Scholar]173. Gluud LL, Dam G, Les I, Cordoba J, Marchesini G, Borre M, Aagaard NK, et al. Branched-chain amino acids for people with hepatic encephalopathy. Cochrane Database Syst Rev 2015:CD001939. [PubMed]174. Gluud LL, Dam G, Les I, Marchesini G, Borre M, Aagaard NK, Vilstrup H. Branched-chain amino acids for people with hepatic encephalopathy. Cochrane Database Syst Rev 2017;5:CD001939. [PMC free article] [PubMed] [Google Scholar]175. Guanabens N, Pares A. Liver and bone. Arch Biochem Biophys 2010;503:84–94. [PubMed] [Google Scholar]176. Compston JE. Hepatic osteodystrophy: vitamin D metabolism in patients with liver disease. Gut 1986;27:1073–1090. [PMC free article] [PubMed] [Google Scholar]177. Assessment of fracture risk and its application to screening for postmenopausal osteoporosis. Report of a WHO Study Group. World Health Organ Tech Rep Ser 1994;843:1–129. [PubMed] [Google Scholar]178. Menon KV, Angulo P, Weston S, Dickson ER, Lindor KD. Bone disease in primary biliary cirrhosis: independent indicators and rate of progression. J Hepatol 2001;35:316–323. [PubMed] [Google Scholar]179. Pares A, Guanabens N, Alvarez L, De Osaba MJ, Oriola J, Pons F, Caballeria L, et al. Collagen type Ialpha1 and vitamin D receptor gene polymorphisms and bone mass in primary biliary cirrhosis. Hepatology 2001;33:554–560. [PubMed] [Google Scholar]180. Guanabens N, Pares A, Ros I, Caballeria L, Pons F, Vidal S, Monegal A, et al. Severity of cholestasis and advanced histological stage but not menopausal status are the major risk factors for osteoporosis in primary biliary cirrhosis. J Hepatol 2005;42:573–577. [PubMed] [Google Scholar]181. Guichelaar MM, Kendall R, Malinchoc M, Hay JE. Bone mineral density before and after OLT: long-term follow-up and predictive factors. Liver Transpl 2006;12:1390–1402. [PubMed] [Google Scholar]182. Guanabens N, Cerda D, Monegal A, Pons F, Caballeria L, Peris P, Pares A. Low bone mass and severity of cholestasis affect fracture risk in patients with primary biliary cirrhosis. Gastroenterology 2010;138:2348–2356. [PubMed] [Google Scholar]183. Angulo P, Grandison GA, Fong DG, Keach JC, Lindor KD, Bjornsson E, Koch A. Bone disease in patients with primary sclerosing cholangitis. Gastroenterology 2011;140:180–188. [PMC free article] [PubMed] [Google Scholar]184. Diamond T, Stiel D, Lunzer M, Wilkinson M, Roche J, Posen S. Osteoporosis and skeletal fractures in chronic liver disease. Gut 1990;31:82–87. [PMC free article] [PubMed] [Google Scholar]185. Chen CC, Wang SS, Jeng FS, Lee SD. Metabolic bone disease of liver cirrhosis: is it parallel to the clinical severity of cirrhosis? J Gastroenterol Hepatol 1996;11:417–421. [PubMed] [Google Scholar]186. Monegal A, Navasa M, Guanabens N, Peris P, Pons F, Martinez de Osaba MJ, Rimola A, et al. Osteoporosis and bone mineral metabolism disorders in cirrhotic patients referred for orthotopic liver transplantation. Calcif Tissue Int 1997;60:148–154. [PubMed] [Google Scholar]187. Ninkovic M, Skingle SJ, Bearcroft PW, Bishop N, Alexander GJ, Compston JE. Incidence of vertebral fractures in the first three months after orthotopic liver transplantation. Eur J Gastroenterol Hepatol 2000;12:931–935. [PubMed] [Google Scholar]188. Carey EJ, Balan V, Kremers WK, Hay JE. Osteopenia and osteoporosis in patients with end-stage liver disease caused by hepatitis C and alcoholic liver disease: not just a cholestatic problem. Liver Transpl 2003;9:1166–1173. [PubMed] [Google Scholar]189. Gonzalez-Calvin JL, Mundi JL, Casado-Caballero FJ, Abadia AC, Martin-Ibanez JJ. Bone mineral density and serum levels of soluble tumor necrosis factors, estradiol, and osteoprotegerin in postmenopausal women with cirrhosis after viral hepatitis. J Clin Endocrinol Metab 2009;94:4844–4850. [PubMed] [Google Scholar]190. Monegal A, Navasa M, Peris P, Colmenero J, Cuervo A, Muxi A, Gifre L, et al. Bone disease in patients awaiting liver transplantation. Has the situation improved in the last two decades? Calcif Tissue Int 2013;93:571–576. [PubMed] [Google Scholar]191. Sinigaglia L, Fargion S, Fracanzani AL, Binelli L, Battafarano N, Varenna M, Piperno A, et al. Bone and joint involvement in genetic hemochromatosis: role of cirrhosis and iron overload. J Rheumatol 1997;24:1809–1813. [PubMed] [Google Scholar]192. Guggenbuhl P, Deugnier Y, Boisdet JF, Rolland Y, Perdriger A, Pawlotsky Y, Chales G. Bone mineral density in men with genetic hemochromatosis and HFE gene mutation. Osteoporos Int 2005;16:1809–1814. [PubMed] [Google Scholar]193. Valenti L, Varenna M, Fracanzani AL, Rossi V, Fargion S, Sinigaglia L. Association between iron overload and osteoporosis in patients with hereditary hemochromatosis. Osteoporos Int 2009;20:549–555. [PubMed] [Google Scholar]194. Guanabens N, Pares A, Navasa M, Martinez de Osaba MJ, Hernandez ME, Munoz J, Rodes J. Cyclosporin A increases the biochemical markers of bone remodeling in primary biliary cirrhosis. J Hepatol 1994;21:24–28. [PubMed] [Google Scholar]195. Springer JE, Cole DE, Rubin LA, Cauch-Dudek K, Harewood L, Evrovski J, Peltekova VD, et al. Vitamin D-receptor genotypes as independent genetic predictors of decreased bone mineral density in primary biliary cirrhosis. Gastroenterology 2000;118:145–151. [PubMed] [Google Scholar]196. Newton J, Francis R, Prince M, James O, Bassendine M, Rawlings D, Jones D. Osteoporosis in primary biliary cirrhosis revisited. Gut 2001;49:282–287. [PMC free article] [PubMed] [Google Scholar]197. Solerio E, Isaia G, Innarella R, Di Stefano M, Farina M, Borghesio E, Framarin L, et al. Osteoporosis: still a typical complication of primary biliary cirrhosis? Dig Liver Dis 2003;35:339–346. [PubMed] [Google Scholar]198. Bonkovsky HL, Hawkins M, Steinberg K, Hersh T, Galambos JT, Henderson JM, Millikan WJ, et al. Prevalence and prediction of osteopenia in chronic liver disease. Hepatology 1990;12:273–280. [PubMed] [Google Scholar]199. Ninkovic M, Love SA, Tom B, Alexander GJ, Compston JE. High prevalence of osteoporosis in patients with chronic liver disease prior to liver transplantation. Calcif Tissue Int 2001;69:321–326. [PubMed] [Google Scholar]200. Sokhi RP, Anantharaju A, Kondaveeti R, Creech SD, Islam KK, Van Thiel DH. Bone mineral density among cirrhotic patients awaiting liver transplantation. Liver Transpl 2004;10:648–653. [PubMed] [Google Scholar]201. Olsson R, Johansson C, Lindstedt G, Mellstrom D. Risk factors for bone loss in chronic active hepatitis and primary biliary cirrhosis. Scand J Gastroenterol 1994;29:753–756. [PubMed] [Google Scholar]202. Monegal A, Navasa M, Guanabens N, Peris P, Pons F, Martinez de Osaba MJ, Ordi J, et al. Bone disease after liver transplantation: a long-term prospective study of bone mass changes, hormonal status and histomorphometric characteristics. Osteoporos Int 2001;12:484–492. [PubMed] [Google Scholar]203. Leidig-Bruckner G, Hosch S, Dodidou P, Ritschel D, Conradt C, Klose C, Otto G, et al. Frequency and predictors of osteoporotic fractures after cardiac or liver transplantation: a follow-up study. Lancet 2001;357:342–347. [PubMed] [Google Scholar]204. Navasa M, Monegal A, Guanabens N, Peris P, Rimola A, Munoz-Gomez J, Visa J, et al. Bone fractures in liver transplant patients. Br J Rheumatol 1994;33:52–55. [PubMed] [Google Scholar]205. Compston JE. Osteoporosis after liver transplantation. Liver Transpl 2003;9:321–330. [PubMed] [Google Scholar]206. Pares A, Guanabens N. Treatment of bone disorders in liver disease. J Hepatol 2006;45:445–453. [PubMed] [Google Scholar]207. Guanabens N, Pares A, Alvarez L, Martinez de Osaba MJ, Monegal A, Peris P, Ballesta AM, et al. Collagen-related markers of bone turnover reflect the severity of liver fibrosis in patients with primary biliary cirrhosis. J Bone Miner Res 1998;13:731–738. [PubMed] [Google Scholar]208. Guanabens N, Monegal A, Muxi A, Martinez-Ferrer A, Reyes R, Caballeria J, Del Rio L, et al. Patients with cirrhosis and ascites have false values of bone density: implications for the diagnosis of osteoporosis. Osteoporos Int 2012;23:1481–1487. [PubMed] [Google Scholar]209. Guanabens N, Pares A, Monegal A, Peris P, Pons F, Alvarez L, de Osaba MJ, et al. Etidronate versus fluoride for treatment of osteopenia in primary biliary cirrhosis: preliminary results after 2 years. Gastroenterology 1997;113:219–224. [PubMed] [Google Scholar]210. Guanabens N, Pares A, Ros I, Alvarez L, Pons F, Caballeria L, Monegal A, et al. Alendronate is more effective than etidronate for increasing bone mass in osteopenic patients with primary biliary cirrhosis. Am J Gastroenterol 2003;98:2268–2274. [PubMed] [Google Scholar]211. Lindor KD, Jorgensen RA, Tiegs RD, Khosla S, Dickson ER. Etidronate for osteoporosis in primary biliary cirrhosis: a randomized trial. J Hepatol 2000;33:878–882. [PubMed] [Google Scholar]212. Wolfhagen FH, van Buuren HR, den Ouden JW, Hop WC, van Leeuwen JP, Schalm SW, Pols HA. Cyclical etidronate in the prevention of bone loss in corticosteroid-treated primary biliary cirrhosis. A prospective, controlled pilot study. J Hepatol 1997;26:325–330. [PubMed] [Google Scholar]213. Guanabens N, Monegal A, Cerda D, Muxi A, Gifre L, Peris P, Pares A. Randomized trial comparing monthly ibandronate and weekly alendronate for osteoporosis in patients with primary biliary cirrhosis. Hepatology 2013;58:2070–2078. [PubMed] [Google Scholar]214. Zein CO, Jorgensen RA, Clarke B, Wenger DE, Keach JC, Angulo P, Lindor KD. Alendronate improves bone mineral density in primary biliary cirrhosis: a randomized placebo-controlled trial. Hepatology 2005;42:762–771. [PubMed] [Google Scholar]215. Ninkovic M, Love S, Tom BD, Bearcroft PW, Alexander GJ, Compston JE. Lack of effect of intravenous pamidronate on fracture incidence and bone mineral density after orthotopic liver transplantation. J Hepatol 2002;37:93–100. [PubMed] [Google Scholar]216. Monegal A, Guanabens N, Suarez MJ, Suarez F, Clemente G, Garcia-Gonzalez M, De la Mata M, et al. Pamidronate in the prevention of bone loss after liver transplantation: a randomized controlled trial. Transpl Int 2009;22:198–206. [PubMed] [Google Scholar]217. Millonig G, Graziadei IW, Eichler D, Pfeiffer KP, Finkenstedt G, Muehllechner P, Koenigsrainer A, et al. Alendronate in combination with calcium and vitamin D prevents bone loss after orthotopic liver transplantation: a prospective single-center study. Liver Transpl 2005;11:960–966. [PubMed] [Google Scholar]218. Crawford BA, Kam C, Pavlovic J, Byth K, Handelsman DJ, Angus PW, McCaughan GW. Zoledronic acid prevents bone loss after liver transplantation: a randomized, double-blind, placebo-controlled trial. Ann Intern Med 2006;144:239–248. [PubMed] [Google Scholar]219. Atamaz F, Hepguler S, Akyildiz M, Karasu Z, Kilic M. Effects of alendronate on bone mineral density and bone metabolic markers in patients with liver transplantation. Osteoporos Int 2006;17:942–949. [PubMed] [Google Scholar]220. Olsson R, Mattsson LA, Obrant K, Mellstrom D. Estrogen-progestogen therapy for low bone mineral density in primary biliary cirrhosis. Liver 1999;19:188–192. [PubMed] [Google Scholar]221. Pereira SP, O’Donohue J, Moniz C, Phillips MG, Abraha H, Buxton-Thomas M, Williams R. Transdermal hormone replacement therapy improves vertebral bone density in primary biliary cirrhosis: results of a 1-year controlled trial. Aliment Pharmacol Ther 2004;19:563–570. [PubMed] [Google Scholar]222. Ormarsdottir S, Mallmin H, Naessen T, Petren-Mallmin M, Broome U, Hultcrantz R, Loof L. An open, randomized, controlled study of transdermal hormone replacement therapy on the rate of bone loss in primary biliary cirrhosis. J Intern Med 2004;256:63–69. [PubMed] [Google Scholar]223. Isoniemi H, Appelberg J, Nilsson CG, Makela P, Risteli J, Hockerstedt K. Transdermal oestrogen therapy protects postmenopausal liver transplant women from osteoporosis. A 2-year follow-up study. J Hepatol 2001;34:299–305. [PubMed] [Google Scholar]224. Diamond T, Stiel D, Posen S. Effects of testosterone and venesection on spinal and peripheral bone mineral in six hypogonadal men with hemochromatosis. J Bone Miner Res 1991;6:39–43. [PubMed] [Google Scholar]225. Dresner-Pollak R, Gabet Y, Steimatzky A, Hamdani G, Bab I, Ackerman Z, Weinreb M. Human parathyroid hormone 1–34 prevents bone loss in experimental biliary cirrhosis in rats. Gastroenterology 2008;134:259–267. [PubMed] [Google Scholar]226. Keeffe EB, Gettys C, Esquivel CO. Liver transplantation in patients with severe obesity. Transplantation 1994;57:309–311. [PubMed] [Google Scholar]227. Sawyer RG, Pelletier SJ, Pruett TL. Increased early morbidity and mortality with acceptable long-term function in severely obese patients undergoing liver transplantation. Clinical transplantation 1999;13:126–130. [PubMed] [Google Scholar]228. Nair S, Verma S, Thuluvath PJ. Obesity and its effect on survival in patients undergoing orthotopic liver transplantation in the United States. Hepatology 2002;35:105–109. [PubMed] [Google Scholar]229. Dick AA, Spitzer AL, Seifert CF, Deckert A, Carithers RL, Reyes JD, Perkins JD. Liver transplantation at the extremes of the body mass index. Liver Transplantation 2009;15:968–977. [PubMed] [Google Scholar]230. Leonard J, Heimbach J, Malinchoc M, Watt K, Charlton M. The Impact of Obesity on Long-term Outcomes in Liver Transplant Recipients—Results of the NIDDK Liver Transplant Database. American journal of transplantation 2008;8:667–672. [PubMed] [Google Scholar]231. Pikul J, Sharpe MD, Lowndes R, Ghent CN. Degree of preoperative malnutrition is predictive of postoperative morbidity and mortality in liver transplant recipients. Transplantation 1994;57:469–472. [PubMed] [Google Scholar]232. Harrison J, McKiernan J, Neuberger JM. A prospective study on the effect of recipient nutritional status on outcome in liver transplantation. Transplant International 1997;10:369–374. [PubMed] [Google Scholar]233. Selberg O, Bottcher J, Tusch G, Pichlmayr R, Henkel E, Muller M-J. Identification of high-and low-risk patients before liver transplantation: a prospective cohort study of nutritional and metabolic parameters in 150 patients. Hepatology 1997;25:652–657. [PubMed] [Google Scholar]234. Tandon P, Ney M, Irwin I, Ma MM, Gramlich L, Bain VG, Esfandiari N, et al. Severe muscle depletion in patients on the liver transplant wait list: its prevalence and independent prognostic value. Liver Transplantation 2012;18:1209–1216. [PubMed] [Google Scholar]235. Montano–Loza AJ, Meza–Junco J, Prado CM, Lieffers JR, Baracos VE, Bain VG, Sawyer MB. Muscle wasting is associated with mortality in patients with cirrhosis. Clinical Gastroenterology and Hepatology 2012;10:166–173. e161. [PubMed] [Google Scholar]236. DiMartini A, Cruz RJ, Dew MA, Myaskovsky L, Goodpaster B, Fox K, Kim KH, et al. Muscle mass predicts outcomes following liver transplantation. Liver Transplantation 2013;19:1172–1180. [PMC free article] [PubMed] [Google Scholar]237. Durand F, Buyse S, Francoz C, Laouenan C, Bruno O, Belghiti J, Moreau R, et al. Prognostic value of muscle atrophy in cirrhosis using psoas muscle thickness on computed tomography. J Hepatol 2014;60:1151–1157. [PubMed] [Google Scholar]238. Yadav A, Chang YH, Carpenter S, Silva AC, Rakela J, Aqel BA, Byrne TJ, et al. Relationship between sarcopenia, six-minute walk distance and health-related quality of life in liver transplant candidates. Clinical transplantation 2015;29:134–141. [PubMed] [Google Scholar]239. Wang CW, Feng S, Covinsky KE, Hayssen H, Zhou L-Q, Yeh BM, Lai JC. A comparison of muscle function, mass, and quality in liver transplant candidates: results from the functional assessment in liver transplantation study. Transplantation 2016;100:1692–1698. [PMC free article] [PubMed] [Google Scholar]240. Dunn MA, Josbeno DA, Schmotzer AR, Tevar AD, DiMartini AF, Landsittel DP, Delitto A. The gap between clinically assessed physical performance and objective physical activity in liver transplant candidates. Liver Transplantation 2016;22:1324–1332. [PubMed] [Google Scholar]241. Lai JC, Volk ML, Strasburg D, Alexander N. Performance-Based Measures Associate With Frailty in Patients With End-Stage Liver Disease. Transplantation 2016;100:2656–2660. [PMC free article] [PubMed] [Google Scholar]242. Kalafateli M, Mantzoukis K, Choi Yau Y, Mohammad AO, Arora S, Rodrigues S, Vos M, et al. Malnutrition and sarcopenia predict post-liver transplantation outcomes independently of the Model for End-stage Liver Disease score. Journal of cachexia, sarcopenia and muscle 2017;8:113–121. [PMC free article] [PubMed] [Google Scholar]243. Sinclair M, Poltavskiy E, Dodge JL, Lai JC. Frailty is independently associated with increased hospitalisation days in patients on the liver transplant waitlist. World J Gastroenterol 2017;23:899–905. [PMC free article] [PubMed] [Google Scholar]244. Ferreira LG, Ferreira Martins AI, Cunha CE, Anastacio LR, Lima AS, Correia MI. Negative energy balance secondary to inadequate dietary intake of patients on the waiting list for liver transplantation. Nutrition 2013;29:1252–1258. [PubMed] [Google Scholar]245. Ney M, Abraldes JG, Ma M, Belland D, Harvey A, Robbins S, Den Heyer V, et al. Insufficient protein intake is associated with increased mortality in 630 patients with cirrhosis awaiting liver transplantation. Nutrition in Clinical Practice 2015;30:530–536. [PubMed] [Google Scholar]246. Le Cornu KA, McKiernan FJ, Kapadia SA, Neuberger JM. A prospective randomized study of preoperative nutritional supplementation in patients awaiting elective orthotopic liver transplantation. Transplantation 2000;69:1364–1369. [PubMed] [Google Scholar]247. Plank LD, McCall JL, Gane EJ, Rafique M, Gillanders LK, McIlroy K, Munn SR. Pre- and postoperative immunonutrition in patients undergoing liver transplantation: a pilot study of safety and efficacy. Clin Nutr 2005;24:288–296. [PubMed] [Google Scholar]248. Plank LD, Mathur S, Gane EJ, Peng SL, Gillanders LK, McIlroy K, Chavez CP, et al. Perioperative immunonutrition in patients undergoing liver transplantation: A randomized double-blind trial. Hepatology 2015;61:639–647. [PubMed] [Google Scholar]249. Lei Q, Wang X, Zheng H, Bi J, Tan S, Li N. Peri-operative immunonutrition in patients undergoing liver transplantation: a meta-analysis of randomized controlled trials. Asia Pacific journal of clinical nutrition 2015;24:583–590. [PubMed] [Google Scholar]250. Kaido T, Mori A, Ogura Y, Hata K, Yoshizawa A, Iida T, Yagi S, et al. Impact of enteral nutrition using a new immuno-modulating diet after liver transplantation. Hepatogastroenterology 2010;57:1522–1525. [PubMed] [Google Scholar]251. Hanai T, Shiraki M, Nishimura K, Ohnishi S, Imai K, Suetsugu A, Takai K, et al. Sarcopenia impairs prognosis of patients with liver cirrhosis. Nutrition 2015;31:193–199. [PubMed] [Google Scholar]252. Garrison RN, Cryer HM, Howard DA, Polk H Jr. Clarification of risk factors for abdominal operations in patients with hepatic cirrhosis. Annals of surgery 1984;199:648. [PMC free article] [PubMed] [Google Scholar]253. Merli M, Nicolini G, Angeloni S, Riggio O. Malnutrition is a risk factor in cirrhotic patients undergoing surgery. Nutrition 2002;18:978–986. [PubMed] [Google Scholar]254. Swart G, Van den Berg J, Van Vuure J, Rietveld T, Wattimena D, Frenkel M. Minimum protein requirements in liver cirrhosis determined by nitrogen balance measurements at three levels of protein intake. Clinical Nutrition 1989;8:329–336. [PubMed] [Google Scholar]255. Zillikens M, Van den Berg J, Wattimena J, Rietveld T, Swart G. Nocturnal oral glucose supplementation: the effects on protein metabolism in cirrhotic patients and in healthy controls. Journal of hepatology 1993;17:377–383. [PubMed] [Google Scholar]256. Coolsen MM, Wong-Lun-Hing EM, Dam RM, Wilt AA, Slim K, Lassen K, Dejong CH. A systematic review of outcomes in patients undergoing liver surgery in an enhanced recovery after surgery pathways. HPB 2013;15:245–251. [PMC free article] [PubMed] [Google Scholar]257. Hughes MJ, McNally S, Wigmore SJ. Enhanced recovery following liver surgery: a systematic review and meta-analysis. HPB 2014;16:699–706. [PMC free article] [PubMed] [Google Scholar]258. Weimann A, Braga M, Carli F, Higashiguchi T, Hübner M, Klek S, Laviano A, et al. ESPEN guideline: Clinical nutrition in surgery. Clinical Nutrition 2017;36:623–650. [PubMed] [Google Scholar]259. Reilly J, Mehta R, Teperman L, Cemaj S, Tzakis A, Yanaga K, Ritter P, et al. Nutritional support after liver transplantation: a randomized prospective study. Journal of Parenteral and Enteral Nutrition 1990;14:386–391. [PubMed] [Google Scholar]260. Hasse JM, Blue LS, Liepa GU, Goldstein RM, Jennings LW, Mor E, Husberg BS, et al. Early enteral nutrition support in patients undergoing liver transplantation. Journal of Parenteral and Enteral Nutrition 1995;19:437–443. [PubMed] [Google Scholar]261. Wicks C, Routley D, Williams R, Somasundaram S, Bjarnason I, Potter D, Tan K, et al. Comparison of enteral feeding and total parenteral nutrition after liver transplantation. The Lancet 1994;344:837–840. [PubMed] [Google Scholar]262. Rayes N, Seehofer D, Hansen S, Boucsein K, Muller AR, Serke S, Bengmark S, et al. Early enteral supply of lactobacillus and fiber versus selective bowel decontamination: a controlled trial in liver transplant recipients. Transplantation 2002;74:123–127. [PubMed] [Google Scholar]263. Pescovitz MD, Mehta PL, Leapman SB, Milgrom ML, Jindal RM, Filo RS. Tube jejunostomy in liver transplant recipients. Surgery 1995;117:642–647. [PubMed] [Google Scholar]264. Mehta PL, Alaka KJ, Filo RS, Leapman SB, Milgrom ML, Pescovitz MD. Nutrition support following liver transplantation: comparison of jejunal versus parenteral routes. Clin Transplant 1995;9:364–369. [PubMed] [Google Scholar]265. Hu Q-G, Zheng Q-C. The influence of enteral nutrition in postoperative patients with poor liver function. World journal of gastroenterology: WJG 2003;9:843. [PMC free article] [PubMed] [Google Scholar]266. Plank LD, Metzger DJ, McCall JL, Barclay KL, Gane EJ, Streat SJ, Munn SR, et al. Sequential changes in the metabolic response to orthotopic liver transplantation during the first year after surgery. Annals of surgery 2001;234:245. [PMC free article] [PubMed] [Google Scholar]267. Weijs PJ, Cynober L, DeLegge M, Kreymann G, Wernerman J, Wolfe RR. Proteins and amino acids are fundamental to optimal nutrition support in critically ill patients. Crit Care 2014;18:591. [PMC free article] [PubMed] [Google Scholar]268. McClave SA, DiBaise JK, Mullin GE, Martindale RG. ACG Clinical Guideline: Nutrition Therapy in the Adult Hospitalized Patient. Am J Gastroenterol 2016;111:315–334; quiz 335. [PubMed] [Google Scholar]269. McClave SA, Taylor BE, Martindale RG, Warren MM, Johnson DR, Braunschweig C, McCarthy MS, et al. Guidelines for the Provision and Assessment of Nutrition Support Therapy in the Adult Critically Ill Patient: Society of Critical Care Medicine (SCCM) and American Society for Parenteral and Enteral Nutrition (A.S.P.E.N.). JPEN J Parenter Enteral Nutr 2016;40:159–211. [PubMed] [Google Scholar]270. Kyoung K-H, Lee S-G, Nam CW, Nah YW. Beneficial effect of low caloric intake in the early period after orthotopic liver transplantation: a new concept using graft weight. Hepato-gastroenterology 2014;61:1668–1672. [PubMed] [Google Scholar]271. Marik PE. Is early starvation beneficial for the critically ill patient? Curr Opin Clin Nutr Metab Care 2016;19:155–160. [PubMed] [Google Scholar]272. Zhu X, Wu Y, Qiu Y, Jiang C, Ding Y. Effects of omega-3 fish oil lipid emulsion combined with parenteral nutrition on patients undergoing liver transplantation. JPEN J Parenter Enteral Nutr 2013;37:68–74. [PubMed] [Google Scholar]273. Zhu XH, Wu YF, Qiu YD, Jiang CP, Ding YT. Liver-protecting effects of omega-3 fish oil lipid emulsion in liver transplantation. World J Gastroenterol 2012;18:6141–6147. [PMC free article] [PubMed] [Google Scholar]274. Kuse ER, Kotzerke J, Müller S, Nashan B, Lück R, Jaeger K. Hepatic reticuloendothelial function during parenteral nutrition including an MCT/LCT or LCT emulsion after liver transplantation–a double-blind study. Transplant international 2002;15:272–277. [PubMed] [Google Scholar]275. McClave SA, Kushner R, Van Way CW 3rd, Cave M, DeLegge M, Dibaise J, Dickerson R, et al. Nutrition therapy of the severely obese, critically ill patient: summation of conclusions and recommendations. JPEN J Parenter Enteral Nutr 2011;35:88S–96S. [PubMed] [Google Scholar]276. Lundbom N, Laurila O, Laurila S. Central pontine myelinolysis after correction of chronic hyponatraemia. Lancet 1993;342:247–248. [PubMed] [Google Scholar]277. Mcdiarmid SV, JOHN O COLONNA I, Shaked A, Ament ME, Busuttil RW. A comparison of renal function in cyclosporine-and FK-506-treated patients after primary orthotopic liver transplantation. Transplantation 1993;56:847–853. [PubMed] [Google Scholar]278. Murray M, Grogan TA, Lever J, Warty VS, Fung J, Venkataramanan R. Comparison of tacrolimus absorption in transplant patients receiving continuous versus interrupted enteral nutritional feeding. Annals of Pharmacotherapy 1998;32:633–636. [PubMed] [Google Scholar]279. Richards J, Gunson B, Johnson J, Neuberger J. Weight gain and obesity after liver transplantation. Transplant international 2005;18:461–466. [PubMed] [Google Scholar]280. Laryea M, Watt KD, Molinari M, Walsh MJ, McAlister VC, Marotta PJ, Nashan B, et al. Metabolic syndrome in liver transplant recipients: prevalence and association with major vascular events. Liver transplantation 2007;13:1109–1114. [PubMed] [Google Scholar]281. Bianchi G, Marchesini G, Marzocchi R, Pinna AD, Zoli M. Metabolic syndrome in liver transplantation: relation to etiology and immunosuppression. Liver Transpl 2008;14:1648–1654. [PubMed] [Google Scholar]282. Schutz T, Hudjetz H, Roske AE, Katzorke C, Kreymann G, Budde K, Fritsche L, et al. Weight gain in long-term survivors of kidney or liver transplantation--another paradigm of sarcopenic obesity? Nutrition 2012;28:378–383. [PubMed] [Google Scholar]283. Krasnoff JB, Vintro AQ, Ascher NL, Bass NM, Dodd MJ, Painter PL. Objective measures of health-related quality of life over 24 months post-liver transplantation. Clin Transplant 2005;19:1–9. [PubMed] [Google Scholar]284. Krasnoff JB, Vintro AQ, Ascher NL, Bass NM, Paul SM, Dodd MJ, Painter PL. A randomized trial of exercise and dietary counseling after liver transplantation. Am J Transplant 2006;6:1896–1905. [PubMed] [Google Scholar]285. Roman E, Torrades MT, Nadal MJ, Cardenas G, Nieto JC, Vidal S, Bascunana H, et al. Randomized pilot study: effects of an exercise programme and leucine supplementation in patients with cirrhosis. Dig Dis Sci 2014;59:1966–1975. [PubMed] [Google Scholar]286. Harimoto N, Shirabe K, Yamashita YI, Ikegami T, Yoshizumi T, Soejima Y, Ikeda T, et al. Sarcopenia as a predictor of prognosis in patients following hepatectomy for hepatocellular carcinoma. Br J Surg 2013;100:1523–1530. [PubMed] [Google Scholar]287. Voron T, Tselikas L, Pietrasz D, Pigneur F, Laurent A, Compagnon P, Salloum C, et al. Sarcopenia Impacts on Short- and Long-term Results of Hepatectomy for Hepatocellular Carcinoma. Ann Surg 2015;261:1173–1183. [PubMed] [Google Scholar]288. Kanematsu T, Koyanagi N, Matsumata T, Kitano S, Takenaka K, Sugimachi K. Lack of preventive effect of branched-chain amino acid solution on postoperative hepatic encephalopathy in patients with cirrhosis: a randomized, prospective trial. Surgery 1988;104:482–488. [PubMed] [Google Scholar]289. Tang ZF, Ling YB, Lin N, Hao Z, Xu RY. Glutamine and recombinant human growth hormone protect intestinal barrier function following portal hypertension surgery. World J Gastroenterol 2007;13:2223–2228. [PMC free article] [PubMed] [Google Scholar]290. Addolorato G, Capristo E, Greco AV, Caputo F, Stefanini GF, Gasbarrini G. Three months of abstinence from alcohol normalizes energy expenditure and substrate oxidation in alcoholics: a longitudinal study. Am J Gastroenterol 1998;93:2476–2481. [PubMed] [Google Scholar]291. Levine JA, Harris MM, Morgan MY. Energy expenditure in chronic alcohol abuse. Eur J Clin Invest 2000;30:779–786. [PubMed] [Google Scholar]292. Nasrallah SM, Galambos JT. Aminoacid therapy of alcoholic hepatitis. Lancet 1980;2:1276–1277. [PubMed] [Google Scholar]293. Morgan TR, Moritz TE, Mendenhall CL, Haas R. Protein consumption and hepatic encephalopathy in alcoholic hepatitis. VA Cooperative Study Group #275. J Am Coll Nutr 1995;14:152–158. [PubMed] [Google Scholar]294. Cabre E, Rodriguez-Iglesias P, Caballeria J, Quer JC, Sanchez-Lombrana JL, Pares A, Papo M, et al. Short- and long-term outcome of severe alcohol-induced hepatitis treated with steroids or enteral nutrition: a multicenter randomized trial. Hepatology 2000;32:36–42. [PubMed] [Google Scholar]295. Moreno C, Deltenre P, Senterre C, Louvet A, Gustot T, Bastens B, Hittelet A, et al. Intensive Enteral Nutrition Is Ineffective for Patients With Severe Alcoholic Hepatitis Treated With Corticosteroids. Gastroenterology 2016;150:903–910 e908. [PubMed] [Google Scholar]296. Zacharias T, Ferreira N, Carin AJ. Preoperative immunonutrition in liver resection-a propensity score matched case-control analysis. Eur J Clin Nutr 2014;68:964–969. [PubMed] [Google Scholar]297. Badalamenti S, Salerno F, Salmeron JM, Lorenzano E, Rimola A, Gines P, Jimenez W, et al. Lack of renal effects of fish oil administration in patients with advanced cirrhosis and impaired glomerular filtration. Hepatology 1997;25:313–316. [PubMed] [Google Scholar]298. de Ledinghen V, Beau P, Mannant PR, Borderie C, Ripault MP, Silvain C, Beauchant M. Early feeding or enteral nutrition in patients with cirrhosis after bleeding from esophageal varices? A randomized controlled study. Dig Dis Sci 1997;42:536–541. [PubMed] [Google Scholar]299. Hebuterne X, Vanbiervliet G. Feeding the patients with upper gastrointestinal bleeding. Curr Opin Clin Nutr Metab Care 2011;14:197–201. [PubMed] [Google Scholar]300. McClave SA, Chang WK. When to feed the patient with gastrointestinal bleeding. Nutr Clin Pract 2005;20:544–550. [PubMed] [Google Scholar]


【本文地址】

公司简介

联系我们

今日新闻

    推荐新闻

    专题文章
      CopyRight 2018-2019 实验室设备网 版权所有