ABSTRACT

Hepatic encephalopathy (HE) is a neuropsychiatric disorder associated with liver dysfunction. This literature review explores the therapeutic potential of nutritional and microbiome-based interventions in managing HE, particularly minimal hepatic encephalopathy. Nutritional therapy, along with probiotics, prebiotics, and fecal microbiota transplants, has shown efficacy in reducing ammonia levels, improving cognitive function, and enhancing health-related quality of life. Additionally, the safety of 6'-sialyllactose, a human milk component, has been confirmed in clinical trials, supporting its potential use in HE treatment. Emerging evidence also suggests that bacteriophages within the gut microbiome are linked to cognitive impairment in cirrhotic patients, indicating that microbiome modulation could provide new therapeutic avenues. These findings highlight the promise of nutritional and microbiome-based strategies in managing HE and emphasize the need for further research to fully integrate these approaches into clinical practice.

Key words: hepatic encephalopathy, sialyllactose, human milk oligosaccharide, prebiotics, microbiota

INTRODUCTION

Hepatic encephalopathy (HE), also known as portosystemic encephalopathy, is a wide spectrum of neuropsychiatric abnormalities in patients who are suffering from liver failure or portosystemic shunt. It is characterized by a transient cognitive decline, personality changes, and behavioral abnormalities, and is associated with various neurological dysfunctions. Cirrhosis is the primary disease linked to hepatic encephalopathy, and its global incidence is on the rise. As of 2024, Cirrhosis is the 4th leading cause of death from non-communicable diseases worldwide, but it ranks 10th in developed countries.[13] Its prevalence in the United States is estimated at around 2.2 million.[3] Which is on the increase primarily due to the rising incidence of Nonalcoholic fatty liver disease (NAFLD), Hepatitis C virus (HCV), and Hepatocellular carcinoma (HCC).[4,5] The real problem lies in managing hepatic encephalopathy due to the complexity of its pathogenesis, most studies being conducted on animal models, and a wide range of presentations.[6] Unlike patients with overt HE, some patients develop a milder form of HE or minimal hepatic encephalopathy (MHE) that presents with mild signs of altered brain function.[7] The exact mechanisms behind MHE are complex and not yet fully understood. Research involving cirrhotic patients and animal models with MHE suggests that an imbalance in gut microbiota triggers systemic inflammation, elevated ammonia levels, and endotoxemia. These factors, in turn, contribute to neuroinflammation in the brain through the gut-liver-brain axis, and predispose to complications like spontaneous bacterial peritonitis, and gastrointestinal bleeding.[8] The role of gut microbiota dysbiosis is crucial in developing MHE.[9,10] Current clinical treatments for MHE, such as lactulose, rifaximin, probiotics, synbiotics, and fecal microbiota transplantation, primarily work by correcting gut microbiota imbalances.[1113] While managing HE is challenging. A promising yet unproven compound appears to be Human Milk Oligosaccharides(HMO), which is a long-chain carbohydrate, a non-nutritive bioactive prebiotic, that possesses antibacterial effects and can disrupt biofilms that bacteria use for protection, thereby influencing the gut microbiome and enhancing the effectiveness of antimicrobial agents.[14] Benefits of HMOs have been shown in different subgroups of patients, mainly pediatric, improvements in gastrointestinal function, and cognitive function, and selectively encouraging the growth of beneficial bacteria (probiotics).[15,16] What are the challenges that face HMOs to be used as a treatment for HE and can it be compared to a better-understood and widely studied oligosaccharide, lactulose?

PATHOGENESIS OF HEPATIC ENCEPHALOPATHY

The pathogenesis of HE is an interplay of various metabolic factors. The primary suspect is ammonia. Unfortunately, ammonia levels cannot be used to direct the management, diagnosis or correlating it to HE severity.[17,18] The main source of ammonia is the gut microbiome through the breakdown of proteins from dietary intake acting on urea through urease. In healthy hepatic cells, ammonia can be processed Primarily through transamination followed by deamination from amino groups of nitrogenous bases such as purines and pyrimidines.[19] However, in cirrhotic liver ammonia bypasses the liver directly to the bloodstream which is toxic to brain and muscle cells.[19] Thus, leading to transient attacks of brain edema, in the short term, and brain atrophy, in the long term.[20] The effects of ammonia on muscles manifest as sarcopenia, loss of muscle mass, and power, which is an indicator of poor morbidity and survival rates.[2124] Patients who suffer from sarcopenia present with low BMI, elevated myostatin levels, and reduced irisin levels. In a subgroup analysis between CHILD A and CHILD BC groups, serum irisin levels were the most significant predictor of cirrhosis-related mortality in the 4th year In the CHILD A group, whereas in the CHILD BC group, higher serum myostatin levels were more predictive of mortality, independent of sarcopenia status.[25]

SYSTEMIC INFLAMMATION AND NEUROTOXICITY

Hyperammonemia and frequent infections are the main causes of morbidity and mortality in HE cases.[26] However, cytokines and pro-inflammatory mediators play a major role in weakening the innate immune system that constitutes the blood-brain barrier. The result is a vicious cycle of infection or inflammation that puts stress on the immune system which responds to the stressor by releasing more pro-inflammatory mediators like tumor necrosis factor alpha (TNF)-α, interferon-gamma (IFN)-γ, interleukins like IL1, IL-6, IL-12. These cytokines influence leukocytes, specifically the neutrophils, to activate and contribute to further transcription of more inflammatory mediators. The result is the development of the systemic inflammatory response syndrome (SIRS) which is defined as (1) temperature > 38°C or < 36°C; (2) heart rate > 90 beats per minute; (3) respiratory rate > 20 breaths per minute or PaCO2 < 32 mmHg; and (4) white blood cell count > 12,000/m3 or < 4000/m3, or >10% immature (band forms). This syndrome developed irrespective of infection status but in response to the severity of the underlying cause.[27]

Hyperammonemia can alter the synaptic transmission in the hippocampus by altering membrane receptors of GluA2 and GluA1.[28] A more recent in vivo study in 2023 found that another novel piece of the puzzle, miR-122-5p, and miR-183-5p, identified for the first time to be able to affect the blood-brain barrier (BBB) causing structural and functional changes.[25] The study also identified the targets of these miRNAs to be occludin and integrin β1 which are junctional proteins on the surface of endothelial cells that form the BBB. These results align with the growing evidence of another older study that proposed the relationship between the mi-R-183 family and the progression of liver fibrosis.[25]

GUT MICROBIOTA

The gut microbiome is a collection of many bacteria, viruses, and fungi that are considered a part of the innate immune system.[2931] That is directly evidenced by the immune homeostasis achieved through limiting pathogen overgrowth, modulating the immune response, and regulating the environment around them.[30,32] Any deficiency in these protective organisms results in disorders in the surrounding environment, altering metabolic pathways, and overgrowth of opportunistic pathogens, this phenomenon is termed dysbiosis. A well-known example of this is Clostridium difficile (CD) infection after recent antibiotic use.[33,34] Furthermore, patients with recurrent CD achieved clinical remission 2 to 4 weeks after Fecal Microbiota Transplantation(FMT) was 57% (95% CI 49% to 64%) with a low risk of heterogeneity (I2 = 37%). Moreover, analysis found that FMT significantly reduced Crohn’s disease activity index scores 4 to 8 weeks after the intervention(95% CI −1.12 to −0.20; I2 = 0).[35] This poses an interesting question: can the innate immune system be trained? Can this generalist form of immunity adapt to the influences around it?

DYSBIOSIS IN DIFFERENT LIVER PATHOLOGIES COMPARED TO HE

It is suggested that there is a link between the severity of nonalcoholic fatty liver disease (NAFLD) and dysbiosis. Fifty-seven patients with biopsy-confirmed NAFLD were studied and subcategorized based on taxonomic composition of gut microbiota using 16S ribosomal RNA gene sequencing of stool samples and found that Bacteroides as independently associated with NASH and Ruminococcus with significant fibrosis.[36] That is directly observed in two other studies that found a similar result which is the abundance of firmicutes, bacteroidetes, and proteobacteria species in the same patient group with a correlation to liver fibrosis.[37,38]

Bajaj et al. demonstrated the ability to estimate the degree of dysbiosis and correlate it to the severity of cirrhosis by introducing the concept of cirrhosis/dysbiosis ratio (CDR). The study found that the CDR was significantly lower in healthy individuals compared to those with cirrhosis.[39] Probiotics, prebiotics, and synbiotics are functional food components that modulate gut microflora, benefiting the host's health. Probiotics alter the intestinal flora by reducing the number of urease-producing bacteria, which decreases the production of toxic substances, especially ammonia. This has been shown to improve MHE and prevent overt hepatic encephalopathy (OHE) by lowering blood ammonia levels.[40] An interesting prebiotic is lactulose, one of the most well-studied oligosaccharides, it was found that combining lactulose with Rifaximin was more effective than lactulose alone for reducing the risk of overt HE recurrence and HE-related hospitalization in adults.[41] An unexpected finding was found in hospital patients with liver disease, who were treated with lactulose. Their microbiome supported Bifidobacteria species expansion which led to reduced incidence of infection and improved 90-day survival. That finding demonstrated that oligosaccharides can impact complications like infection rates and muscle wasting.[42,43]

Patients who are suffering from liver cirrhosis face two complications known for their high mortality rates and low life quality, spontaneous bacterial peritonitis and esophageal variceal bleeding. Both of these complications have something in common, dysbiosis and intestinal epithelial barrier dysfunction secondary to cirrhosis.[44,45] A hallmark of these disorders is reduced portal vein blood flow and intestinal vascular constriction directly leading to high intestinal permeability and leakage. This leads us to an important detail, patients with liver failure need constant monitoring and specific adjustments to their gut microbiome because of secondary biliary dysfunction resulting in favoring Escherichia coli and Enterococcus faecalis species.[46] These species are notorious for altering pH in their environment, thus facilitating ammonia absorption and transition from the colon lumen to the blood.[47]

PREBIOTICS, SYNBIOTICS AND PROBIOTICS—WHAT ARE THEY AND HOW DO THEY WORK?

Probiotics are living organisms that feed on bioactive non-digestible non-nutritious molecules, known as prebiotics. These microorganisms play a vital role in maintaining homeostasis. A commonly used prebiotic is oligosaccharides like inulin and oligofructose. In a well-balanced diet, a mixture of probiotics and prebiotics is referred to as synbiotics, which showed miraculous health benefits in improving insulin sensitivity, and weight reduction according to one randomized controlled trial.[48]

Other health benefits include but are not limited to the resolution of diarrhea or constipation, better bile salt conjugation, anti-inflammatory and anti-bacterial properties, combating allergies, and finally some positive effects on aging, fatigue, and diabetes.[49]

GUT-LIVER-BRAIN AXIS—FINAL PIECE OF THE PATHOGENESIS PUZZLE

It is well established that probiotics can improve cognitive function and decrease venous ammonia levels, even with combinations of bacterial species like Clostridium butyricum combined with Bifidobacterium infantis in minimal hepatic encephalopathy, which also showed a significant decrease in harmful Enterococcus and Enterobacteriaceae.[47] Probiotic treatment can be combined with nutritional therapy for maximum effect. Sharma et al., demonstrated that nutritional therapy is an effective intervention for patients with MHE. It not only significantly improves cognitive function and health-related quality of life (HRQOL) but also leads to beneficial changes in nutritional status, biochemical markers such as ammonia and myostatin levels, and reductions in endotoxins and inflammatory cytokines. Furthermore, therapy reduces the progression to overt hepatic encephalopathy.[44,50,51]

To support nutritional and probiotic therapy, this SR/MA evaluated the effectiveness of microbiome therapies, including probiotics, synbiotics, and FMT, in treating HE. The analysis included 21 randomized controlled trials with a total of 1746 participants. The findings demonstrated that microbiome therapies significantly improved MHE, reduced the development of overt HE, and decreased the frequency of serious adverse events. Additionally, these therapies lowered ammonia levels, neurocognitive test scores, and hospitalization rates compared to placebo or no treatment. The study concluded that microbiome therapies are more effective than placebo in improving MHE, preventing progression to overt HE, and reducing related health complications.[52]

In a neurological disorder with different pathophysiological mechanisms from HE, gut microbiome even showed promise in patient-specific Alzheimer’s disease management through symbiotics and fecal microbiota transplantation according to this review.[53] The gut microbiota plays a crucial role in various physiological processes, including short-chain fatty acid production, vitamin biosynthesis, bile salt transformation, and the conversion of complex dietary polysaccharides into more readily absorbable carbohydrates. Furthermore, certain bacterial species within the gut can produce neurotransmitters, such as GABA, noradrenaline, and dopamine, highlighting the potential impact of the gut microbiome on neurological and cognitive functions.[54]

In a randomized controlled trial, 70 participants were given a six-week regimen of a multi-strain probiotic (MSP) containing four strains: Limosilactobacillus fermentum LF16, Lacticaseibacillus rhamnosus LR06, Lactiplantibacillus plantarum LP01, and Bifidobacterium longum 04, with a total dosage of 4 × 10⁹ live cells per day. The results showed significant improvements in depression, anxiety, and mood scores along with increased plasma serotonin levels.[55] A table showing a summary of the main studies and its findings (Table 1).

Table 1: A table showing a summary of the main studies and its findings
Title Findings Sources
Pathogenesis of HE Ammonia is the primary factor but not directly correlate to HE severity [1719]
Ammonia bypasses the liver in cirrhosis, causing brain and muscle damage, leading to edema and sarcopenia. [1922]
Sarcopenia in HE patients is linked to higher morbidity, characterized by low BMI, high myostatin, and low irisin levels [25]
Systemic inflammation & neurotoxicity Hyperammonemia and infections cause morbidity in HE [26]
Cytokines such as TNF-α, IFN-γ, and IL-1, IL-6, IL-12 are involved in a vicious inflammatory cycle, leading to SIRS [27]
Novel miRNAs (miR-122-5p, miR-183-5p) affect the blood-brain barrier (BBB), contributing to HE neurotoxicity. [28,25]
Gut microbiota in HE Dysbiosis is linked to NAFLD, cirrhosis, and liver fibrosis, characterized by shifts in gut microbiome composition (e.g., Firmicutes, Bacteroides) [3641]
Probiotics, prebiotics, synbiotics Probiotics reduce urease-producing bacteria, lowering ammonia production and improving outcomes in HE (MHE & OHE) [4043]
Prebiotics like lactulose combined with rifaximin were more effective than lactulose alone in preventing HE recurrence [41]
Gut-liver-brain axis Probiotic therapies (e.g., Clostridium butyricum, Bifidobacterium infantis) improve cognitive function and reduce ammonia levels. [4749]
Nutritional therapy and probiotics together improve cognitive function, HRQOL, and reduce inflammatory markers in MHE [50,51]
Microbiome therapy in HE A systematic review/meta-analysis of 21 RCTs showed microbiome therapies (e.g., probiotics, FMT) significantly improve MHE, reduce OHE, and decrease adverse events. [52]
Gut microbiome & neurological disorders Gut microbiome therapies show promise in managing Alzheimer's disease and other neurological disorders through symbiotics and FMT. [53,54]
Probiotics in mental health A 6-week trial of a multi-strain probiotic improved depression, anxiety, and mood scores, along with increased serotonin levels [55]

COMPOSITION AND CONCENTRATION OF HUMAN MILK OLIGOSACCHARIDES

The composition of human milk is primarily water (approximately 87%), with smaller amounts of proteins (around 1%), fats (3%–5%), and carbohydrates, which make up about 7% of its composition (Figure 1). The major carbohydrate in human milk is lactose, but an essential subset of carbohydrates, known as human milk oligosaccharides (HMOs), contributes to its complexity and bioactivity.[5660]

Figure 1

Figure 1. A pie chart illustrating the composition of human milk.

A NOVEL PREBIOTIC FOR HE, HUMAN MILK OLIGOSACCHARIDE

Human milk oligosaccharides are a diverse group of complex carbohydrates found in abundance in human breast milk, second only to lactose in terms of concentration. These unique structures are not digestible by human enzymes, but instead serve as a prebiotic, selectively fostering the growth of beneficial bacteria in the infant gut microbiome.[60] One of the primary bacterial groups that can metabolize human milk oligosaccharides is the Bifidobacterium species. These anaerobic bacteria are considered a hallmark of a healthy infant gut, as they facilitate the development of the immune system and protect against pathogenic microorganisms.[61]

The main source of HMO is from the mother’s colostrum. As a result, the composition and diversity of human milk oligosaccharides can vary significantly between individuals, and even within a single mother's milk over time.[64] This heterogeneity may contribute to the establishment of a unique gut microbiome in each infant, as different oligosaccharide structures can selectively enrich distinct microbial communities. Furthermore, other factors such as gestational age, host genetics, and maternal diet may also play a role in shaping the infant gut microbiome, even in the presence of human milk oligosaccharides.[62]

An earlier study involving 125 cirrhotic patients received either Bifidobacterium with fructooligosaccharides (FOS) or lactulose for 60 days. After 30 days patients who were treated with Bifidobacterium plus FOS had significantly better cognitive performance, with lower scores on the Trail Making Test B and higher scores on the Symbol Digit Modalities Test and Block Design Test compared to those treated with lactulose. After 60 days, the Bifidobacterium plus FOS group continued to show superior outcomes, with significantly reduced fasting ammonia levels and improved results on cognitive tests. The study concludes that Bifidobacterium + FOS is an effective alternative to lactulose in cirrhotic patients, offering benefits in lowering blood ammonia levels and enhancing cognitive function.[63]

HMO IN OTHER CONDITIONS

Beyond HE, HMOs have shown potential in other conditions like irritable bowel syndrome (IBS). This is evident with 2'-O-fucosyllactose (2'FL) and lacto-N-neotetraose (LNnT) supplementation on the gut microbiota and metabolite profiles in patients with IBS. The intervention led to moderate changes in fecal microbiota composition, while mucosal microbiota composition remained unchanged. Both fecal and mucosal levels of Bifidobacterium species, including Bifidobacterium adolescentis and Bifidobacterium longum, increased following 2'FL/LNnT intake. The supplementation also influenced fecal and plasma metabolite profiles, with these changes being linked to the increased abundance of bifidobacteria, although it did not affect urine metabolite profiles or host mucosal gene expression. This concludes that 2'FL/LNnT supplementation effectively modulates the gut microbiota and metabolite environment in IBS patients, potentially contributing to health improvements.[64]

In another study involving adults with prediabetes and insulin resistance (PreDM-IR) examined the effects of red raspberry (RRB) supplementation, with or without a fructo-oligosaccharide (FOS) prebiotic, on cardiometabolic risk biomarkers and gut microbiota composition. Participants consumed either RRB alone or RRB + FOS daily for 4 weeks, with a 4-week washout between periods. The results showed that RRB supplementation reduced hepatic insulin resistance by 30.1% and lowered plasma total and LDL cholesterol levels. Adding FOS further improved β-cell function, though it did not significantly affect cholesterol levels. Gut microbiota analysis revealed that RRB increased Eubacterium eligens and decreased Ruminococcus gnavus, while RRB + FOS increased Bifidobacterium spp. and decreased Blautia wexlerae. Notably, R. gnavus was positively correlated with hepatic insulin resistance, while E. eligens and Bifidobacterium catenulatum were negatively correlated with cholesterol levels. These findings suggest that changes in specific gut bacteria linked to RRB and FOS supplementation may contribute to metabolic improvements in PreDM-IR, highlighting the potential for targeted dietary interventions to modulate gut microbiota and improve cardiometabolic health.[65]

SAFETY AND ADVERSE EFFECTS

The safety of HMOs, specifically 2'FL and LNnT, was evaluated at various doses up to 20 grams daily for 2 weeks. HMO supplementation was found to be safe and well-tolerated, as indicated by the gastrointestinal symptoms rating scale. Notably, no participants discontinued the study prematurely due to adverse effects. Additionally, 16S rRNA sequencing revealed that HMO supplementation significantly altered the gut microbiota, notably increasing the abundance of beneficial bacteria such as Actinobacteria and Bifidobacterium, while reducing the relative abundance of Firmicutes and Proteobacteria. These findings suggest that HMO supplementation is an effective strategy to positively influence gut microbiota by promoting the growth of beneficial bifidobacteria.[66]

Another study evaluated participants who consumed either 1 or 3 bottles daily of the test product, which included yogurt strains and probiotic strains Lactobacillus paracasei and Lactobacillus rhamnosus. Safety was evaluated through various health parameters, including adverse events, metabolic profiles, and digestive symptoms. The results showed no significant safety differences between the test and control groups across all parameters. Gut microbiota analysis revealed that the probiotic strains were detectable only during the consumption period, with higher levels in those consuming 3 bottles per day. While the overall gut microbiota structure remained unchanged after 4 weeks, a few bacterial genera exhibited differential responses to the product. Metagenomic analysis indicated that the probiotics contributed functionally to the gut microbiome.[67]

Additionally, a study investigating the safety of 6'-sialyllactose (6'-SL), a component abundant in human milk, compared it with a placebo (maltodextrin). Participants received 3 grams of 6'-SL powder or placebo twice daily for 12 weeks. The study found no serious adverse effects, such as life-threatening complications or significant changes in clinical chemistry tests (e.g., aspartate aminotransferase, alanine aminotransferase, and creatinine) between the 6'-SL and placebo groups. While some gastrointestinal issues like diarrhea and bloating were reported, their frequency was similar between both groups. These findings suggest that 6'-SL is safe for human use, supporting its potential for therapeutic applications.[68]

ETHICAL CONSIDERATIONS, SAFETY, AND COST-BENEFIT ANALYSIS

Human milk is considered the gold standard for oligosaccharide structure and diversity, providing numerous benefits. However, due to practical and ethical constraints, these compounds cannot be easily obtained in large quantities for research purposes. Consequently, colostrum or milk from dairy animals can serve as an alternative source of oligosaccharides, which can be utilized for the development of functional foods and clinical applications.[61]

INNOVATIVE RESEARCH DIRECTIONS AND PERSONALIZED MEDICINE

While lactulose treatment does not significantly change fecal microbiome composition, it may improve dysbiosis and alter microbial metabolic functions.[69] The effects of lactulose on human microbiota composition are both patient- and dose-dependent. Interestingly, a combination of Bifidobacterium and fructo-oligosaccharides (FOS) has shown promise in reducing blood ammonia levels and improving psychometric tests in HE patients, potentially offering an alternative to lactulose treatment.[63,70]

CLINICAL IMPLICATIONS AND KNOWLEDGE GAPS

This review shines a light on the importance of a personalized approach specifically through nutritional and microbiome-based therapy to guide clinical and pharmacological management.

Further research is needed to elucidate the specific mechanisms by which human milk oligosaccharides can influence liver function and the gut microbiome in the context of hepatic encephalopathy. Nevertheless, the existing evidence highlights the potential of this novel therapeutic strategy to improve clinical outcomes in patients with advanced liver disease and associated complications.

CONCLUSION

This literature review highlights the significant potential of HMOs and other prebiotics, such as FOS, in modulating gut microbiota and improving clinical outcomes in various health conditions, including HE, IBS, and cardiometabolic disorders. Studies indicate that HMOs, particularly 2'FL and LNnT, not only promote the growth of beneficial gut bacteria like Bifidobacterium species but also exert positive effects on cognitive function, metabolic health, and immune responses. Additionally, safety evaluations demonstrate that these prebiotics are well-tolerated and can be administered without significant adverse effects, further supporting their therapeutic potential.

In hepatic encephalopathy, the inclusion of HMOs and Bifidobacterium-based interventions has shown promise in improving cognitive function and reducing blood ammonia levels, offering an alternative to traditional treatments like lactulose. Moreover, in other conditions such as IBS and prediabetes, prebiotics have been shown to modulate gut microbiota composition, reduce insulin resistance, and improve lipid profiles, underscoring their role in personalized medicine.

Microbiome-based treatments, including probiotics, synbiotics, and fecal microbiota transplant (FMT), have shown substantial benefits in the treatment of hepatic encephalopathy. These therapies are effective in reversing MHE, reducing the incidence of OHE, and lowering ammonia levels. They also decrease hospitalization rates and the frequency of serious adverse events compared to placebo treatments. The positive outcomes suggest that modulating the gut microbiome can be an effective strategy for improving cognitive function and overall health in patients with HE.

Clinical trials have demonstrated the safety of 6'-SL, a component found abundantly in human milk. While some gastrointestinal issues, such as diarrhea and bloating, were reported, they were not significantly different from those experienced with a placebo. Importantly, no severe adverse effects or significant clinical chemistry changes were noted, supporting the use of 6'-SL for therapeutic purposes. These findings reinforce the safety profile of 6'-SL and its potential for broader applications in clinical settings.

Emerging research highlights a notable link between gut microbiome components, particularly bacteriophages, and cognitive function in patients with cirrhosis. Changes in bacteriophage profiles associated with specific bacteria correlate with cognitive impairment and variations in MHE status. These observations suggest that bacteriophages could influence cognitive outcomes through their effects on neuroactive metabolites produced by gut bacteria. This connection opens new avenues for exploring microbiome modulation as a potential therapeutic approach for improving cognitive function in patients with hepatic encephalopathy.

DECLARATIONS

Author contributions

Kareem Wael Raafat and Ismail Elkhattib were co-first authors.

Use of large language models, AI and machine learning tools

None declared.

Informed consent

Not applicable.

Ethical approval

Not applicable.

Conflicts of interest

There is no conflict of interest among the authors.

Data sharing statement

No additional data is available.

REFERENCES

  1. Louissaint J, Deutsch-Link S, Tapper EB. Changing epidemiology of cirrhosis and hepatic encephalopathy. Clin Gastroenterol Hepatol 2022;20:S1-S8    DOI: 10.1016/j.cgh.2022.04.036    PMID: 35940729
  2. Dávalos Moscol M, Bustios Sanchez C. The burden of hepatic encephalopathy in Latin America. Annals of Hepatology 2011;10:S31-S35    DOI: 10.1016/s1665-2681(19)31603-5
  3. Abdelwahed AH, Aboeldahb M, Wu GY. Effects of transjugular intrahepatic portosystemic shunt on renal and pulmonary function in hepatic decompensation with and without hepatorenal and hepatopulmonary syndromes: A review. J Clin Transl Hepatol 2024;12:780-791    DOI: 10.14218/JCTH.2024.00188    PMID: 39280072
  4. Romero-Gómez M, Boza F, García-Valdecasas MS, García E, Aguilar-Reina J. Subclinical hepatic encephalopathy predicts the development of overt hepatic encephalopathy. Am J Gastroenterol 2001;96:2718-2723    DOI: 10.1111/j.1572-0241.2001.04130.x    PMID: 11569701
  5. Boyer TD, Haskal ZJ, American Association for the Study of Liver Diseases. The role of transjugular intrahepatic portosystemic shunt in the management of portal hypertension. Hepatology 2005;41:386-400    DOI: 10.1002/hep.20559    PMID: 15660434
  6. Lima LCD, Miranda AS, Ferreira RN, Rachid MA, Simões E Silva AC. Hepatic encephalopathy: Lessons from preclinical studies. World J Hepatol 2019;11:173-185    DOI: 10.4254/wjh.v11.i2.173    PMID: 30820267
  7. Zhan T, Stremmel W. The diagnosis and treatment of minimal hepatic encephalopathy. Dtsch Arztebl Int 2012;109:180-187    DOI: 10.3238/arztebl.2012.0180    PMID: 22470407
  8. Dazıroğlu MEÇ, Yıldıran H. Intestinal dysbiosis and probiotic use: its place in hepatic encephalopathy in cirrhosis. Ann Gastroenterol 2023;36:141-148    DOI: 10.20524/aog.2023.0776    PMID: 36864944
  9. Won SM, Oh KK, Gupta H, Ganesan R, Sharma SP, Jeong JJ, et al. The Link between Gut Microbiota and Hepatic Encephalopathy. Int J Mol Sci 2022;23    DOI: 10.3390/ijms23168999    PMID: 36012266
  10. Luo M, Xin RJ, Hu FR, Yao L, Hu SJ, Bai FH. Role of gut microbiota in the pathogenesis and therapeutics of minimal hepatic encephalopathy via the gut-liver-brain axis. World J Gastroenterol 2023;29:144-156    DOI: 10.3748/wjg.v29.i1.144    PMID: 36683714
  11. Bajaj JS, Schubert CM, Heuman DM, Wade JB, Gibson DP, Topaz A, et al. Persistence of cognitive impairment after resolution of overt hepatic encephalopathy. Gastroenterology 2010;138:2332-2340    DOI: 10.1053/j.gastro.2010.02.015    PMID: 20178797
  12. Ortiz M, Córdoba J, Jacas C, Flavià M, Esteban R, Guardia J. Neuropsychological abnormalities in cirrhosis include learning impairment. J Hepatol 2006;44:104-110    DOI: 10.1016/j.jhep.2005.06.013    PMID: 16169117
  13. Schomerus H, Hamster W, Blunck H, Reinhard U, Mayer K, Dölle W. Latent portasystemic encephalopathy. I. Nature of cerebral functional defects and their effect on fitness to drive. Dig Dis Sci 1981;26:622-630    DOI: 10.1007/BF01367675    PMID: 7249898
  14. Ackerman DL, Craft KM, Doster RS, Weitkamp JH, Aronoff DM, Gaddy JA, Townsend SD. Antimicrobial and antibiofilm activity of human milk oligosaccharides against Streptococcus agalactiae, Staphylococcus aureus, and Acinetobacter baumannii. ACS Infect Dis 2018;4:315-324    DOI: 10.1021/acsinfecdis.7b00183    PMID: 29198102
  15. Gomez-Gallego C, Garcia-Mantrana I, Salminen S, Collado MC. The human milk microbiome and factors influencing its composition and activity. Semin Fetal Neonatal Med 2016;21:400-405    DOI: 10.1016/j.siny.2016.05.003    PMID: 27286644
  16. Ballard O, Morrow AL. Human milk composition: nutrients and bioactive factors. Pediatr Clin North Am 2013;60:49-74    DOI: 10.1016/j.pcl.2012.10.002    PMID: 23178060
  17. Hannah N, Vasic D, Kansal A, Al-Ani A, Hebbard G, Sood S. Serum ammonia does not guide management and is overutilised in patients with cirrhosis in hospital settings. Intern Med J 2023;53:2057-2064    DOI: 10.1111/imj.16053    PMID: 36891668
  18. Bajaj JS, Pyrsopoulos NT, Rahimi RS, Heimanson Z, Allen C, Rockey DC. Serum ammonia levels do not correlate with overt hepatic encephalopathy severity in hospitalized patients with cirrhosis. Clin Gastroenterol Hepatol 2024;22:1950-1952.e1    DOI: 10.1016/j.cgh.2024.02.015    PMID: 38423347
  19. Mohiuddin SS, Khattar D. Biochemistry, Ammonia, StatPearls, Feb. 2023, Accessed: Aug. 09, 2024. Online. Available from: https://www.ncbi.nlm.nih.gov/books/NBK541039/
  20. Ferenci P. Hepatic encephalopathy. Gastroenterol Rep (Oxf) 2017;5:138-147    DOI: 10.1093/gastro/gox013    PMID: 28533911
  21. Matsui T, Nagai H, Watanabe G, Mouri K, Yoshimine N, Amanuma M, et al. Measurement of skeletal muscle volume is useful for predicting prognosis in patients with liver cirrhosis. Eur J Gastroenterol Hepatol 2022;34:1151-1157    DOI: 10.1097/MEG.0000000000002435    PMID: 36170684
  22. Sarcopenia is the independent predictor of mortality in critically ill patients with cirrhosis. J Clin Transl Res 2022    DOI: 10.18053/jctres.08.202203.006
  23. Boga S, Yildirim AE, Ucbilek E, Koksal AR, Sisman ST, Durak I, et al. The effect of sarcopenia and serum myokines on prognosis and survival in cirrhotic patients: a multicenter cross-sectional study. Eur J Gastroenterol Hepatol 2022;34:1261-1268    DOI: 10.1097/MEG.0000000000002461    PMID: 36281901
  24. Guo G, Li C, Hui Y, Mao L, Sun M, Li Y, et al. Sarcopenia and frailty combined increases the risk of mortality in patients with decompensated cirrhosis. Ther Adv Chronic Dis 2022;13:20406223221109651    DOI: 10.1177/20406223221109651    PMID: 35875834
  25. Orzeł-Gajowik K, Milewski K, Zielińska M. miRNA-ome plasma analysis unveils changes in blood-brain barrier integrity associated with acute liver failure in rats. Fluids Barriers CNS 2023;20:92    DOI: 10.1186/s12987-023-00484-7    PMID: 38066639
  26. Riggio O, Celsa C, Calvaruso V, Merli M, Caraceni P, Montagnese S, et al. Hepatic encephalopathy increases the risk for mortality and hospital readmission in decompensated cirrhotic patients: a prospective multicenter study. Front Med (Lausanne) 2023;10:1184860    DOI: 10.3389/fmed.2023.1184860    PMID: 37305121
  27. Tranah TH, Vijay GK, Ryan JM, Shawcross DL. Systemic inflammation and ammonia in hepatic encephalopathy. Metab Brain Dis 2013;28:1-5    DOI: 10.1007/s11011-012-9370-2    PMID: 23224356
  28. Pun CK, Huang HC, Chang CC, Hsu SJ, Huang YH, Hou MC, et al. Hepatic encephalopathy: From novel pathogenesis mechanism to emerging treatments. J Chin Med Assoc 2024;87:245-251    DOI: 10.1097/JCMA.0000000000001041    PMID: 38109364
  29. Seekatz AM, Young VB. Clostridium difficile and the microbiota. J Clin Invest 2014;124:4182-4189    DOI: 10.1172/JCI72336    PMID: 25036699
  30. Lynch SV, Pedersen O. The human intestinal microbiome in health and disease. N Engl J Med 2016;375:2369-2379    DOI: 10.1056/NEJMra1600266    PMID: 27974040
  31. Dalal SR, Chang EB. The microbial basis of inflammatory bowel diseases. J Clin Invest 2014;124:4190-4196    DOI: 10.1172/JCI72330    PMID: 25083986
  32. Surana NK, Kasper DL. Deciphering the tête-à-tête between the microbiota and the immune system. J Clin Invest 2014;124:4197-4203    DOI: 10.1172/JCI72332    PMID: 25036709
  33. Chang JT. Pathophysiology of inflammatory bowel diseases. N Engl J Med 2020;383:2652-2664    DOI: 10.1056/NEJMra2002697    PMID: 33382932
  34. Tang WH, Hazen SL. The contributory role of gut microbiota in cardiovascular disease. J Clin Invest 2014;124:4204-4211    DOI: 10.1172/JCI72331    PMID: 25271725
  35. Zhou S, Cui Y, Zhang Y, Zhao T, Cong J. Fecal microbiota transplantation for induction of remission in Crohn's disease: a systematic review and meta-analysis. Int J Colorectal Dis 2023;38:62    DOI: 10.1007/s00384-023-04354-4    PMID: 36882658
  36. Boursier J, Mueller O, Barret M, Machado M, Fizanne L, Araujo-Perez F, et al. The severity of nonalcoholic fatty liver disease is associated with gut dysbiosis and shift in the metabolic function of the gut microbiota. Hepatology 2016;63:764-775    DOI: 10.1002/hep.28356    PMID: 26600078
  37. Caussy C, Hsu C, Lo MT, Liu A, Bettencourt R, Ajmera VH, et al. Link between gut-microbiome derived metabolite and shared gene-effects with hepatic steatosis and fibrosis in NAFLD. Hepatology 2018;68:918-932    DOI: 10.1002/hep.29892    PMID: 29572891
  38. Loomba R, Seguritan V, Li W, Long T, Klitgord N, Bhatt A, et al. Gut microbiome-based metagenomic signature for non-invasive detection of advanced fibrosis in human nonalcoholic fatty liver disease. Cell Metab 2017;25:1054-1062.e5    DOI: 10.1016/j.cmet.2017.04.001    PMID: 28467925
  39. Bajaj JS, Hylemon PB, Ridlon JM, Heuman DM, Daita K, White MB, et al. Colonic mucosal microbiome differs from stool microbiome in cirrhosis and hepatic encephalopathy and is linked to cognition and inflammation. Am J Physiol Gastrointest Liver Physiol 2012;303:G675-G685    DOI: 10.1152/ajpgi.00152.2012    PMID: 22821944
  40. Amodio P, Bemeur C, Butterworth R, Cordoba J, Kato A, Montagnese Set 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    DOI: 10.1002/hep.26370    PMID: 23471642
  41. Sanyal AJ, Kowdley KV, Reau NS, Pyrsopoulos NT, Allen C, Heimanson Z, et al. Rifaximin plus lactulose versus lactulose alone for reducing the risk of HE recurrence. Hepatol Commun 2024;8    DOI: 10.1097/HC9.0000000000000436    PMID: 38727685
  42. Odenwald MA, Lin H, Lehmann C, Dylla NP, Cole CG, Mostad JD, et al. Bifidobacteria metabolize lactulose to optimize gut metabolites and prevent systemic infection in patients with liver disease. Nat Microbiol 2023;8:2033-2049    DOI: 10.1038/s41564-023-01493-w    PMID: 37845315
  43. Wijarnpreecha K, Werlang M, Panjawatanan P, Kroner PT, Cheungpasitporn W, Lukens FJ, et al. Association between sarcopenia and hepatic encephalopathy: A systematic review and meta-analysis. Ann Hepatol 2020;19:245-250    DOI: 10.1016/j.aohep.2019.06.007    PMID: 31422030
  44. Liboredo JC, Vilela EG, Ferrari Mde L, Lima AS, Correia MI. Nutrition status and intestinal permeability in patients eligible for liver transplantation. JPEN J Parenter Enteral Nutr 2015;39:163-170    DOI: 10.1177/0148607113513465    PMID: 24255087
  45. Henao-Mejia J, Elinav E, Thaiss CA, Licona-Limon P, Flavell RA. Role of the intestinal microbiome in liver disease. J Autoimmun 2013;46:66-73    DOI: 10.1016/j.jaut.2013.07.001    PMID: 24075647
  46. Sung JY, Shaffer EA, Costerton JW. Antibacterial activity of bile salts against common biliary pathogens. Effects of hydrophobicity of the molecule and in the presence of phospholipids. Dig Dis Sci 1993;38:2104-2112    DOI: 10.1007/BF01297092    PMID: 8223087
  47. Xia X, Chen J, Xia J, Wang B, Liu H, Yang L, et al. Role of probiotics in the treatment of minimal hepatic encephalopathy in patients with HBV-induced liver cirrhosis. J Int Med Res 2018;46:3596-3604    DOI: 10.1177/0300060518776064    PMID: 29806520
  48. Tajabadi-Ebrahimi M, Sharifi N, Farrokhian A, Raygan F, Karamali F, Razzaghi R, et al. A randomized controlled clinical trial investigating the effect of synbiotic administration on markers of insulin metabolism and lipid profiles in overweight type 2 diabetic patients with coronary heart disease. Exp Clin Endocrinol Diabetes 2017;125:21-27    DOI: 10.1055/s-0042-105441    PMID: 27219886
  49. Pandey KR, Naik SR, Vakil BV. Probiotics, prebiotics and synbiotics- a review. J Food Sci Technol 2015;52:7577-7587    DOI: 10.1007/s13197-015-1921-1    PMID: 26604335
  50. Lu H, Zhang H, Wu Z, Li L. Microbiota-gut-liver-brain axis and hepatic encephalopathy. Microbiome Res Rep 2024;3:17    DOI: 10.20517/mrr.2023.44    PMID: 38841407
  51. Sharma BC, Maharshi S, Sachdeva S, Mahajan B, Sharma A, Bara S, et al. Nutritional therapy for persistent cognitive impairment after resolution of overt hepatic encephalopathy in patients with cirrhosis: A double-blind randomized controlled trial. J Gastroenterol Hepatol 2023;38:1917-1925    DOI: 10.1111/jgh.16266    PMID: 37354045
  52. Gao J, Nie R, Chang H, Yang W, Ren Q. A meta-analysis of microbiome therapies for hepatic encephalopathy. Eur J Gastroenterol Hepatol 2023;35:927-937    DOI: 10.1097/MEG.0000000000002596    PMID: 37505972
  53. Zhang T, Gao G, Kwok LY, Sun Z. Gut microbiome-targeted therapies for Alzheimer's disease. Gut Microbes 2023;15:2271613    DOI: 10.1080/19490976.2023.2271613    PMID: 37934614
  54. Biagini F, Calvigioni M, Lapomarda A, Vecchione A, Magliaro C, De Maria C, et al. A novel 3D in vitro model of the human gut microbiota. Sci Rep 2020;10:21499    DOI: 10.1038/s41598-020-78591-w    PMID: 33299026
  55. Walden KE, Moon JM, Hagele AM, Allen LE, Gaige CJ, Krieger JM, et al. Corrigendum: A randomized controlled trial to examine the impact of a multi-strain probiotic on self-reported indicators of depression, anxiety, mood, and associated biomarkers. Front Nutr 2023;10:1324536    DOI: 10.3389/fnut.2023.1324536    PMID: 38075220
  56. Rousseaux A, Brosseau C, Le Gall S, Piloquet H, Barbarot S, Bodinier M. Human Milk Oligosaccharides: Their Effects on the Host and Their Potential as Therapeutic Agents. Front Immunol 2021;12:680911    DOI: 10.3389/fimmu.2021.680911    PMID: 34108974
  57. Boquien CY. Human milk: An ideal food for nutrition of preterm newborn. Front Pediatr 2018;6:295    DOI: 10.3389/fped.2018.00295    PMID: 30386758
  58. Garwolińska D, Namieśnik J, Kot-Wasik A, Hewelt-Belka W. Chemistry of human breast milk-a comprehensive review of the composition and role of milk metabolites in child development. J Agric Food Chem 2018;66:11881-11896    DOI: 10.1021/acs.jafc.8b04031    PMID: 30247884
  59. Breast Milk Composition: Key Nutrients & Changes | Medela. Accessed: Oct. 20, 2024. Online. Available from: https://www.medela.com/en/breastfeeding-pumping/articles/power-of-breast-milk/breast-milk-composition-what-s-in-your-breast-milk
  60. Cho S, Samuel TM, Li T, Howell BR, Baluyot K, Hazlett HC, et al. Interactions between Bifidobacterium and Bacteroides and human milk oligosaccharides and their associations with infant cognition. Front Nutr 2023;10:1216327    DOI: 10.3389/fnut.2023.1216327    PMID: 37457984
  61. Taufik E, Arief II, Budiman C, Wibisono Y, Noviyanto A. Characterization of bioactive sialyl oligosaccharides separated from colostrum of indonesia dairy goat. Food Sci Anim Resour 2022;42:426-440    DOI: 10.5851/kosfa.2022.e13    PMID: 35611077
  62. Ortega MA, Fraile-Martínez O, Naya I, García-Honduvilla N, Álvarez-Mon M, Buján J, et al. Type 2 diabetes mellitus associated with obesity (diabesity). The central role of gut microbiota and its translational applications. Nutrients 2020;12    DOI: 10.3390/nu12092749    PMID: 32917030
  63. Malaguarnera M, Gargante MP, Malaguarnera G, Salmeri M, Mastrojeni S, Rampello L, et al. Bifidobacterium combined with fructo-oligosaccharide versus lactulose in the treatment of patients with hepatic encephalopathy. Eur J Gastroenterol Hepatol 2010;22:199-206    DOI: 10.1097/MEG.0b013e328330a8d3    PMID: 19730107
  64. Iribarren C, Magnusson MK, Vigsnæs LK, Aziz I, Amundsen ID, Šuligoj T, et al. The effects of human milk oligosaccharides on gut microbiota, metabolite profiles and host mucosal response in patients with irritable bowel syndrome. Nutrients 2021;13    DOI: 10.3390/nu13113836    PMID: 34836092
  65. Zhang X, Zhao A, Sandhu AK, Edirisinghe I, Burton-Freeman BM. Red raspberry and fructo-oligosaccharide supplementation, metabolic biomarkers, and the gut microbiota in adults with prediabetes: A randomized crossover clinical trial. J Nutr 2022;152:1438-1449    DOI: 10.1093/jn/nxac037    PMID: 35421233
  66. Elison E, Vigsnaes LK, Rindom Krogsgaard L, Rasmussen J, Sørensen N, McConnell B, et al. Oral supplementation of healthy adults with 2'-O-fucosyllactose and lacto-N-neotetraose is well tolerated and shifts the intestinal microbiota. Br J Nutr 2016;116:1356-1368    DOI: 10.1017/S0007114516003354    PMID: 27719686
  67. Alvarez AS, Tap J, Chambaud I, Cools-Portier S, Quinquis L, et al. Safety and functional enrichment of gut microbiome in healthy subjects consuming a multi-strain fermented milk product: a randomized controlled trial. Sci Rep 2020;10:15974    DOI: 10.1038/s41598-020-72161-w    PMID: 32994487
  68. Kim JH, Yong SY, Kim SH, Baek A, Go TH, Kang DR. Randomized, triple-blind, placebo-controlled study to evaluate the safety of 6'-Sialyllactose in healthy adults. Regul Toxicol Pharmacol 2022;129:105110    DOI: 10.1016/j.yrtph.2021.105110    PMID: 34958861
  69. Rai R, Saraswat VA, Dhiman RK. Gut microbiota: its role in hepatic encephalopathy. J Clin Exp Hepatol 2015;5:S29-S36    DOI: 10.1016/j.jceh.2014.12.003    PMID: 26041954
  70. Ruszkowski J, Witkowski JM. Lactulose: Patient- and dose-dependent prebiotic properties in humans. Anaerobe 2019;59:100-106    DOI: 10.1016/j.anaerobe.2019.06.002    PMID: 31176002