1
|
Laghi L, Ortiz MÀ, Rossi G, Román E, Mengucci C, Cantó E, Biagini L, Sánchez E, Mulet M, García-Osuna Á, Urgell E, Kaur N, Poca M, Padrós J, Nadal MJ, Cuyàs B, Alvarado E, Vidal S, Juanes E, Ferrero-Gregori A, Escorsell À, Soriano G. Biomarkers of Frailty in Patients with Advanced Chronic Liver Disease Undergoing a Multifactorial Intervention Consisting of Home Exercise, Branched-Chain Amino Acids, and Probiotics. Biomolecules 2024; 14:1410. [PMID: 39595586 PMCID: PMC11592179 DOI: 10.3390/biom14111410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/23/2024] [Accepted: 10/30/2024] [Indexed: 11/28/2024] Open
Abstract
Frailty in cirrhosis or advanced chronic liver disease (ACLD) is a relevant prognostic factor. In the present study, we aimed to analyze potential biomarkers associated with frailty and its improvement in patients with ACLD. We analyzed the serum of outpatients with ACLD who participated in a previous study (Román, Hepatol Commun 2024) in which frailty was assessed using the liver frailty index (LFI), and patients who were frail or prefrail were randomized to a multifactorial intervention (home exercise, branched-chain amino acids, and probiotics) or control for 12 months. We determined a biomarker battery of inflammation, bacterial translocation, and liver damage in blood and urine and blood metabolomics by 1H-NMR. Thirty-seven patients were included. According to the LFI, 32 patients were frail or prefrail, and 5 were robust. At baseline, LFI correlated with LBP, sCD163, mtDNA, FGF-21, urinary NGAL, urinary claudin-3, and the metabolites mannose, ethanol, and isoleucine. During the study, patients in the intervention group showed an improvement in LFI and a decrease in CRP, LBP, sCD163, and ccK18 compared to the control group. Metabolomics showed a decrease in dimethyl sulfone and creatinine and an increase in malonate, ornithine, isoleucine, and valine in the intervention group. We conclude that frailty in patients with ACLD is associated with biomarkers of systemic inflammation, bacterial translocation, and liver damage, and alterations of amino acid and short-chain fatty acid metabolism.
Collapse
Affiliation(s)
- Luca Laghi
- Department of Agricultural and Food Sciences, University of Bologna, 47521 Cesena, Italy;
| | - Maria Àngels Ortiz
- Institut de Recerca Sant Pau (IR Sant Pau), 08041 Barcelona, Spain; (M.À.O.); (E.C.); (E.S.); (M.M.); (S.V.); (A.F.-G.)
| | - Giacomo Rossi
- School of Veterinary Medical Sciences, University of Camerino, 62032 Camerino, Italy; (G.R.); (L.B.)
| | - Eva Román
- CIBERehd (Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas), Instituto de Salud Carlos III, 28029 Madrid, Spain; (E.R.); (M.P.); (B.C.); (E.A.)
- University Nursing School EUI-Sant Pau, 08025 Barcelona, Spain
- Department of Gastroenterology, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (N.K.); (À.E.)
| | - Carlo Mengucci
- Department of Agricultural and Food Sciences, University of Bologna, 47521 Cesena, Italy;
| | - Elisabet Cantó
- Institut de Recerca Sant Pau (IR Sant Pau), 08041 Barcelona, Spain; (M.À.O.); (E.C.); (E.S.); (M.M.); (S.V.); (A.F.-G.)
| | - Lucia Biagini
- School of Veterinary Medical Sciences, University of Camerino, 62032 Camerino, Italy; (G.R.); (L.B.)
| | - Elisabet Sánchez
- Institut de Recerca Sant Pau (IR Sant Pau), 08041 Barcelona, Spain; (M.À.O.); (E.C.); (E.S.); (M.M.); (S.V.); (A.F.-G.)
- CIBERehd (Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas), Instituto de Salud Carlos III, 28029 Madrid, Spain; (E.R.); (M.P.); (B.C.); (E.A.)
| | - Maria Mulet
- Institut de Recerca Sant Pau (IR Sant Pau), 08041 Barcelona, Spain; (M.À.O.); (E.C.); (E.S.); (M.M.); (S.V.); (A.F.-G.)
| | - Álvaro García-Osuna
- Department of Biochemistry, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (Á.G.-O.); (E.U.)
| | - Eulàlia Urgell
- Department of Biochemistry, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (Á.G.-O.); (E.U.)
| | - Naujot Kaur
- Department of Gastroenterology, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (N.K.); (À.E.)
| | - Maria Poca
- CIBERehd (Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas), Instituto de Salud Carlos III, 28029 Madrid, Spain; (E.R.); (M.P.); (B.C.); (E.A.)
- Department of Gastroenterology, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (N.K.); (À.E.)
| | - Josep Padrós
- Department of Physical Medicine and Rehabilitation, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (J.P.); (M.J.N.)
| | - Maria Josep Nadal
- Department of Physical Medicine and Rehabilitation, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (J.P.); (M.J.N.)
| | - Berta Cuyàs
- CIBERehd (Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas), Instituto de Salud Carlos III, 28029 Madrid, Spain; (E.R.); (M.P.); (B.C.); (E.A.)
- Department of Gastroenterology, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (N.K.); (À.E.)
| | - Edilmar Alvarado
- CIBERehd (Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas), Instituto de Salud Carlos III, 28029 Madrid, Spain; (E.R.); (M.P.); (B.C.); (E.A.)
- Department of Gastroenterology, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (N.K.); (À.E.)
| | - Silvia Vidal
- Institut de Recerca Sant Pau (IR Sant Pau), 08041 Barcelona, Spain; (M.À.O.); (E.C.); (E.S.); (M.M.); (S.V.); (A.F.-G.)
- Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Elena Juanes
- Department of Pharmacy, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain;
| | - Andreu Ferrero-Gregori
- Institut de Recerca Sant Pau (IR Sant Pau), 08041 Barcelona, Spain; (M.À.O.); (E.C.); (E.S.); (M.M.); (S.V.); (A.F.-G.)
| | - Àngels Escorsell
- Department of Gastroenterology, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (N.K.); (À.E.)
- Faculty of Medicine, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - German Soriano
- CIBERehd (Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas), Instituto de Salud Carlos III, 28029 Madrid, Spain; (E.R.); (M.P.); (B.C.); (E.A.)
- Department of Gastroenterology, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (N.K.); (À.E.)
- Faculty of Medicine, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| |
Collapse
|
2
|
Zhang X, Xia Y, Cao L, Ji B, Ge Z, Zheng Q, Qi Z, Ding S. PC 18:1/18:1 mediates the anti-inflammatory effects of exercise and remodels tumor microenvironment of hepatocellular carcinoma. Life Sci 2024; 336:122335. [PMID: 38103729 DOI: 10.1016/j.lfs.2023.122335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 12/04/2023] [Accepted: 12/04/2023] [Indexed: 12/19/2023]
Abstract
AIM Phosphatidylcholine (PC) is essential for membrane structural integrity and lipid-dependent signaling pathways, and is an essential component required for cancer cell growth. Using hepatocellular carcinoma (HCC) as a tumor model, this study aims to further screen phospholipid biomarkers of the tumor microenvironment and explore the anti-tumor effects and mechanisms of aerobic exercise. MAIN METHODS The HCC of C57BL/6J mice was induced by the injection of the carcinogen diethylnitrosamine (DEN). Exercise was performed on an ungraded treadmill for weeks. The inflammation-related markers were detected by ELISA, PCR and immunohistochemistry, hepatic metabolic profile was analyzed by GC/MS, and lipid metabolism profile was further detected by lipid-targeted LC/MS. Cell culture was used to verify the anti-inflammatory effect of PC. KEY FINDINGS Exercise reduced hepatic inflammation, tumor incidence and volume. Metabolomics analysis showed that palmitic acid is a key metabolic marker for exercise to improve tumor microenvironment. Injection of exogenous palmitic acid following exercise impaired the anti-inflammatory and anti-tumor effects of exercise. Lipid metabolomics analysis further showed that metabolites for exercise were enriched in glycerol phospholipid metabolism, including 14 phosphatidylcholines (PCs), 18 phosphatidylethanolamines (PEs), and 6 triglycerides (TGs). These biomarkers contain different lengths of fatty acid chains and different numbers of unsaturated bonds, respectively. Cell culture verified that PC (18:1/18:1) mediated lipopolysaccharide (LPS)-induced inflammation in HepG2 cell. SIGNIFICANCE Our results suggest that exercise remodels glycerophospholipid metabolism and reduces hepatic palmitic acid loading and PC (18:1/18:1) level, thereby reconstructing a microenvironment that is hostile to HCC.
Collapse
Affiliation(s)
- Xue Zhang
- School of Sports Medicine and Health, Chengdu Sport University, Chengdu 610041, China; School of Physical Education and Health, East China Normal University, Shanghai 200241, China; School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China
| | - Yixue Xia
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China
| | - Lu Cao
- School of Physical Education and Health, East China Normal University, Shanghai 200241, China
| | - Benlong Ji
- School of Physical Education and Health, East China Normal University, Shanghai 200241, China
| | - Zhe Ge
- School of Physical Education and Health, East China Normal University, Shanghai 200241, China
| | - Qingyun Zheng
- School of Physical Education and Sport, Henan University, Kaifeng 475001, China.
| | - Zhengtang Qi
- School of Physical Education and Health, East China Normal University, Shanghai 200241, China.
| | - Shuzhe Ding
- School of Physical Education and Health, East China Normal University, Shanghai 200241, China.
| |
Collapse
|
3
|
Polito R, Monaco ML, Mallardo M, Elce A, Daniele A, Nigro E. Treatment with sera from Water Polo athletes activates AMPKα and ACC proteins In HepG2 hepatoma cell line. SPORT SCIENCES FOR HEALTH 2021. [DOI: 10.1007/s11332-021-00742-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Abstract
Purpose
Physical activity and professional physical activity such as water polo (WP) sport, has numerous beneficial effects to fight metabolism-related disorders through several mechanisms, including the promotion of liver metabolic adaptations, and the modulation of cytokine production. The aim of this study was to investigate the effects of different types of physical activity on AMPKα and ACC, two proteins involved in liver metabolism; therefore, we treated the hepatoma cell line Hep G2 with sera from elite WP athletes and amateur (basket) players. As control, we used serum from both sedentary and obese subjects.
Methods
Help G2 cells were treated with 5% of human sera from the different subjects; after 24 h and 48 h, HepG2 cell viability was verified through MTT assay and activation status of AMPKα and ACC through western blotting. Cytokine’s serum levels were measured through ELISA assay.
Results
After 72 h, the treatment of HepG2 cells with sera from the different subjects produced no effect on cell viability. Furthermore, after 48 h of treatment, both AMPKα and ACC phosphorylation statistically increases in HepG2 cells treated with sera from WP athletes. Furthermore, IL-4, IL-6 and IL-10 levels resulted statistically increased in WP athlete’s sera than in sedentary subjects.
Conclusion
The specific activation of AMPKα and ACC by WP sera confirms that professional sport activity carried out by WP athletes can be considered as a physiological activator of these two proteins also in HepG2 liver cells. In addition, the increase of anti-inflammatory cytokines in WP sera confirms the ample evidence for multiple anti-inflammatory activities carried out by WP discipline.
Collapse
|
4
|
Tacke F, Weiskirchen R. Non-alcoholic fatty liver disease (NAFLD)/non-alcoholic steatohepatitis (NASH)-related liver fibrosis: mechanisms, treatment and prevention. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:729. [PMID: 33987427 PMCID: PMC8106094 DOI: 10.21037/atm-20-4354] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Liver fibrosis is the excessive expression and accumulation of extracellular matrix proteins in the liver. Fibrotic scarring occurs as the consequence of chronic injury and inflammation. While the successful treatment of hepatitis B and C reduced the burden of liver disease related to viral hepatitis, non-alcoholic fatty liver disease (NAFLD) or non-alcoholic steatohepatitis (NASH) are nowadays the leading causes of hepatic fibrosis worldwide. Although basic research activities have significantly advanced our understanding of the molecular disease pathogenesis, the present therapeutic options for fibrosis are still limited. In advanced disease stages, liver transplantation often remains the only curative treatment. This highlights the necessity of preventive strategies to avoid complications of fibrosis, particularly cirrhosis, portal hypertension and liver cancer. Lifestyle modifications (weight loss, exercise, healthy diet) are the basis for prevention and treatment of NAFLD-associated fibrosis. In the present review, we discuss recent advances in antifibrotic prevention and therapy. In particular, we review the current concepts for antifibrotic drug candidates in the treatment of NAFLD and NASH. While some compounds aim at reverting pathogenic liver metabolism, an alternative approach is to disconnect the injury (e.g., NAFLD) from inflammation and/or fibrosis. Investigational drugs typically target metabolic pathways, insulin resistance, hepatocyte death, inflammatory cell recruitment or activation, the gut-liver axis, matrix expression or matrix turnover. While several promising drug candidates failed in phase 2 or 3 clinical trials (including elafibranor, emricasan and selonsertib), promising results with the farnesoid X receptor agonist obeticholic acid, the pan-PPAR agonist lanifibranor and the chemokine receptor CCR2/CCR5 inhibitor cenicriviroc support the expectation of an effective pharmacological therapy for liver fibrosis in the near future. Tackling NAFLD-associated fibrosis from different directions by combinatorial drug treatment and effective lifestyle changes hold the greatest prospects.
Collapse
Affiliation(s)
- Frank Tacke
- Department of Hepatology and Gastroenterology, Charité University Medicine Berlin, Berlin, Germany
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, Aachen, Germany
| |
Collapse
|
5
|
Roeb E, Weiskirchen R. Fructose and Non-Alcoholic Steatohepatitis. Front Pharmacol 2021; 12:634344. [PMID: 33628193 PMCID: PMC7898239 DOI: 10.3389/fphar.2021.634344] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 01/07/2021] [Indexed: 12/12/2022] Open
Abstract
Background: The excessive consumption of free sugars is mainly responsible for the high prevalence of obesity and metabolic syndrome in industrialized countries. More and more studies indicate that fructose is involved in the pathophysiology and also in the degree of disease of non-alcoholic fatty liver disease (NAFLD). In epidemiologic studies, energy-adjusted higher fructose consumption correlates with NAFLD in overweight adults. In addition to glucose, fructose, as an equivalent component of conventional household sugar, appears to have negative metabolic effects in particular due to its exclusive hepatic metabolism. Liver-related mortality is strictly associated with the degree of fibrosis, whereas the most common cause of death in patients suffering from NAFLD and non-alcoholic steatohepatitis (NASH) are still cardiovascular diseases. In this review article, we have summarized the current state of knowledge regarding a relationship between fructose consumption, liver fibrosis and life expectancy in NASH. Method: Selective literature search in PubMed using the keywords 'non-alcoholic fatty liver', 'fructose', and 'fibrosis' was conducted. Results: The rate of overweight and obesity is significantly higher in both, adult and pediatric NASH patients. The consumption of free sugars is currently three times the maximum recommended amount of 10% of the energy intake. The current literature shows weight gain, negative effects on fat and carbohydrate metabolism and NASH with hypercaloric intake of fructose. Conclusions: Excessive fructose consumption is associated with negative health consequences. Whether this is due to an excess of energy or the particular metabolism of fructose remains open with the current study situation. The urgently needed reduction in sugar consumption could be achieved through a combination of binding nutritional policy measures including taxation of sugary soft drinks. Previous studies suggest that diet-related fructose intake exceeding the amount contained in vegetables and fruits lead to an increase of hepatic lipogenesis. Thus, further studies to clarify the protective contribution of low-fructose intake to positively influence NAFLD in industrial population are urgently required.
Collapse
Affiliation(s)
- Elke Roeb
- Department of Gastroenterology, Justus Liebig University Giessen and University Hospital Giessen, Giessen, Germany
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, Aachen, Germany
| |
Collapse
|
6
|
Khoo WY, Chrisfield BJ, Sae-Tan S, Lambert JD. Mitigation of nonalcoholic fatty liver disease in high-fat-fed mice by the combination of decaffeinated green tea extract and voluntary exercise. J Nutr Biochem 2020; 76:108262. [PMID: 31759197 PMCID: PMC6995759 DOI: 10.1016/j.jnutbio.2019.108262] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 09/26/2019] [Accepted: 10/22/2019] [Indexed: 12/12/2022]
Abstract
We have shown that combination treatment with decaffeinated green tea extract (GTE) and voluntary exercise (Ex) reduces obesity and insulin resistance in high-fat (HF)-fed mice to a greater extent than either treatment alone. Here, we investigated the effects of GTE-, Ex- or the combination on the development of obesity-related NAFLD. Male C57BL/6 J mice were treated for 16 weeks with HF diet (60% energy from fat), HF supplemented with 7.7 g GTE/kg, HF plus access to a voluntary running wheel, or the combination. We found that treatment of mice with the combination mitigated the development of HF-induced NAFLD to a greater extent than either treatment alone. Combination-treated mice had lower plasma alanine aminotransferase (92% lower) and hepatic lipid accumulation (80% lower) than HF-fed controls: the effect of the single treatments was less significant. Mitigation of NAFLD was associated with higher fecal lipid and nitrogen levels. Combination treated, but not singly treated mice, had higher hepatic expression of genes related to mitochondrial biogenesis (sirtuin 1 [59%]; peroxisome proliferator-activated receptor γ coactivator 1α [42%]; nuclear respiratory factor 1 [38%]; and transcription factor B1, mitochondrial [89%]) compared to the HF-fed controls. GTE-, Ex-, and the combination-treatment groups also had higher hepatic expression of genes related to cholesterol synthesis and uptake, but the combination was not better than the single treatments. Our results suggest the combination of GTE and Ex can effectively mitigate NAFLD. Future studies should determine if the combination is additive or synergistic compared to the single treatments.
Collapse
Affiliation(s)
- Weslie Y Khoo
- Department of Food Science, The Pennsylvania State University, University Park, USA
| | | | - Sudathip Sae-Tan
- Department of Food Science, The Pennsylvania State University, University Park, USA
| | - Joshua D Lambert
- Department of Food Science, The Pennsylvania State University, University Park, USA; Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, USA.
| |
Collapse
|
7
|
Current Status in Testing for Nonalcoholic Fatty Liver Disease (NAFLD) and Nonalcoholic Steatohepatitis (NASH). Cells 2019; 8:cells8080845. [PMID: 31394730 PMCID: PMC6721710 DOI: 10.3390/cells8080845] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 08/05/2019] [Accepted: 08/06/2019] [Indexed: 12/19/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disease in Western countries with almost 25% affected adults worldwide. The growing public health burden is getting evident when considering that NAFLD-related liver transplantations are predicted to almost double within the next 20 years. Typically, hepatic alterations start with simple steatosis, which easily progresses to more advanced stages such as nonalcoholic steatohepatitis (NASH), fibrosis and cirrhosis. This course of disease finally leads to end-stage liver disease such as hepatocellular carcinoma, which is associated with increased morbidity and mortality. Although clinical trials show promising results, there is actually no pharmacological agent approved to treat NASH. Another important problem associated with NASH is that presently the liver biopsy is still the gold standard in diagnosis and for disease staging and grading. Because of its invasiveness, this technique is not well accepted by patients and the method is prone to sampling error. Therefore, an urgent need exists to find reliable, accurate and noninvasive biomarkers discriminating between different disease stages or to develop innovative imaging techniques to quantify steatosis.
Collapse
|
8
|
Weiskirchen R, Weiskirchen S, Tacke F. Recent advances in understanding liver fibrosis: bridging basic science and individualized treatment concepts. F1000Res 2018; 7:F1000 Faculty Rev-921. [PMID: 30002817 PMCID: PMC6024236 DOI: 10.12688/f1000research.14841.1] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/22/2018] [Indexed: 02/06/2023] Open
Abstract
Hepatic fibrosis is characterized by the formation and deposition of excess fibrous connective tissue, leading to progressive architectural tissue remodeling. Irrespective of the underlying noxious trigger, tissue damage induces an inflammatory response involving the local vascular system and the immune system and a systemic mobilization of endocrine and neurological mediators, ultimately leading to the activation of matrix-producing cell populations. Genetic disorders, chronic viral infection, alcohol abuse, autoimmune attacks, metabolic disorders, cholestasis, alterations in bile acid composition or concentration, venous obstruction, and parasite infections are well-established factors that predispose one to hepatic fibrosis. In addition, excess fat and other lipotoxic mediators provoking endoplasmic reticulum stress, alteration of mitochondrial function, oxidative stress, and modifications in the microbiota are associated with non-alcoholic fatty liver disease and, subsequently, the initiation and progression of hepatic fibrosis. Multidisciplinary panels of experts have developed practice guidelines, including recommendations of preferred therapeutic approaches to a specific cause of hepatic disease, stage of fibrosis, or occurring co-morbidities associated with ongoing loss of hepatic function. Here, we summarize the factors leading to liver fibrosis and the current concepts in anti-fibrotic therapies.
Collapse
Affiliation(s)
- Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, Pauwelsstraße 30, Germany
| | - Sabine Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, Pauwelsstraße 30, Germany
| | - Frank Tacke
- Department of Medicine III, RWTH University Hospital Aachen, D-52074 Aachen, Pauwelsstraße 30, Germany
| |
Collapse
|
9
|
Lambertz J, Weiskirchen S, Landert S, Weiskirchen R. Fructose: A Dietary Sugar in Crosstalk with Microbiota Contributing to the Development and Progression of Non-Alcoholic Liver Disease. Front Immunol 2017; 8:1159. [PMID: 28970836 PMCID: PMC5609573 DOI: 10.3389/fimmu.2017.01159] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 09/01/2017] [Indexed: 12/12/2022] Open
Abstract
Fructose is one of the key dietary catalysts in the development of non-alcoholic fatty liver disease (NAFLD). NAFLD comprises a complex disease spectrum, including steatosis (fatty liver), non-alcoholic steatohepatitis, hepatocyte injury, inflammation, and fibrosis. It is also the hepatic manifestation of the metabolic syndrome, which covers abdominal obesity, insulin resistance, dyslipidemia, glucose intolerance, or type 2 diabetes mellitus. Commensal bacteria modulate the host immune system, protect against exogenous pathogens, and are gatekeepers in intestinal barrier function and maturation. Dysbalanced intestinal microbiota composition influences a variety of NAFLD-associated clinical conditions. Conversely, nutritional supplementation with probiotics and preobiotics impacting composition of gut microbiota can improve the outcome of NAFLD. In crosstalk with the host immune system, the gut microbiota is able to modulate inflammation, insulin resistance, and intestinal permeability. Moreover, the composition of microbiota of an individual is a kind of fingerprint highly influenced by diet. In addition, not only the microbiota itself but also its metabolites influence the metabolism and host immune system. The gut microbiota can produce vitamins and a variety of nutrients including short-chain fatty acids. Holding a healthy balance of the microbiota is therefore highly important. In the present review, we discuss the impact of long-term intake of fructose on the composition of the intestinal microbiota and its biological consequences in regard to liver homeostasis and disease. In particular, we will refer about fructose-induced alterations of the tight junction proteins affecting the gut permeability, leading to the translocation of bacteria and bacterial endotoxins into the blood circulation.
Collapse
Affiliation(s)
- Jessica Lambertz
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, Aachen, Germany
| | - Sabine Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, Aachen, Germany
| | - Silvano Landert
- Culture Collection of Switzerland AG (CCOS), Wädenswil, Switzerland
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, Aachen, Germany
| |
Collapse
|
10
|
Huber Y, Gehrke N, Biedenbach J, Helmig S, Simon P, Straub BK, Bergheim I, Huber T, Schuppan D, Galle PR, Wörns MA, Schuchmann M, Schattenberg JM. Voluntary distance running prevents TNF-mediated liver injury in mice through alterations of the intrahepatic immune milieu. Cell Death Dis 2017. [PMID: 28640248 PMCID: PMC5520921 DOI: 10.1038/cddis.2017.266] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Physical activity confers a broad spectrum of health benefits. Beyond the obvious role in metabolically driven diseases, the role of physical activity in acute liver injury is poorly explored. To study the role of physical activity in acute liver injury, a novel model of voluntary distance running in mice was developed and mice were subjected to acute liver injury induced by N-galactosamine (GalN) and lipopolysaccharide (LPS). Analyses included histological stains, immunoblotting, qRT-PCR and FACS analysis. Voluntary distance running increased to an average of 10.3 km/day after a learning curve. Running lead to a decrease in the absolute numbers of intrahepatic CD4+ T and B lymphocytes and macrophages after 7 weeks. In parallel, hepatic mRNA expression of inflammatory cytokines including IL-6 and IL-1beta, TGF-beta and monocyte chemoattractant protein-1 (MCP-1/CCL2) were suppressed, while TNF-α was not affected by exercise. Likewise, expression of the macrophage-specific antigen F4/80 was downregulated 1.6-fold from exercise. Notably, acute liver injury from GaIN/LPS was significantly blunted following 7 weeks of voluntary exercise as determined by liver histology, a 84.6% reduction of alanine aminotransferase (P<0.01) and a 54.6% reduction of aspartate aminotransferase (P<0.05) compared with sedentary mice. Additionally, proinflammatory cytokines, activation of caspase 3 and JNK were significantly lower, while antiapoptotic protein A20 increased. Voluntary distance running alters the intrahepatic immune phenotype producing an environment that is less susceptible to acute liver injury.
Collapse
Affiliation(s)
- Yvonne Huber
- I. Department of Medicine, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Nadine Gehrke
- I. Department of Medicine, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Jana Biedenbach
- I. Department of Medicine, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Susanne Helmig
- Department of Sports Medicine, Rehabilitation and Prevention, Johannes Gutenberg University, Mainz, Germany
| | - Perikles Simon
- Department of Sports Medicine, Rehabilitation and Prevention, Johannes Gutenberg University, Mainz, Germany
| | - Beate K Straub
- Institute of Pathology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Ina Bergheim
- Department of Nutritional Sciences, Molecular Nutritional Science, University of Vienna, Vienna A-1090, Austria
| | - Tobias Huber
- Department of General, Visceral and Transplant Surgery, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Detlef Schuppan
- Institute of Translational Immunology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Peter R Galle
- I. Department of Medicine, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Marcus A Wörns
- I. Department of Medicine, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | | | - Jörn M Schattenberg
- I. Department of Medicine, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
11
|
The Therapeutic Potential of Anti-Inflammatory Exerkines in the Treatment of Atherosclerosis. Int J Mol Sci 2017; 18:ijms18061260. [PMID: 28608819 PMCID: PMC5486082 DOI: 10.3390/ijms18061260] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 05/22/2017] [Accepted: 06/09/2017] [Indexed: 12/15/2022] Open
Abstract
Although many cardiovascular (CVD) medications, such as antithrombotics, statins, and antihypertensives, have been identified to treat atherosclerosis, at most, many of these therapeutic agents only delay its progression. A growing body of evidence suggests physical exercise could be implemented as a non-pharmacologic treatment due to its pro-metabolic, multisystemic, and anti-inflammatory benefits. Specifically, it has been discovered that certain anti-inflammatory peptides, metabolites, and RNA species (collectively termed “exerkines”) are released in response to exercise that could facilitate these benefits and could serve as potential therapeutic targets for atherosclerosis. However, much of the relationship between exercise and these exerkines remains unanswered, and there are several challenges in the discovery and validation of these exerkines. This review primarily highlights major anti-inflammatory exerkines that could serve as potential therapeutic targets for atherosclerosis. To provide some context and comparison for the therapeutic potential of exerkines, the anti-inflammatory, multisystemic benefits of exercise, the basic mechanisms of atherosclerosis, and the limited efficacies of current anti-inflammatory therapeutics for atherosclerosis are briefly summarized. Finally, key challenges and future directions for exploiting these exerkines in the treatment of atherosclerosis are discussed.
Collapse
|