1
|
Xu C, Ezzi SHA, Zou X, Dong Y, Alhaskawi A, Zhou H, Kota VG, Abdulla MHAH, Abdalbary SA, Lu H. The role of TNF in metabolic disorders and liver diseases. Cytokine 2025; 190:156933. [PMID: 40174483 DOI: 10.1016/j.cyto.2025.156933] [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: 01/20/2025] [Revised: 03/23/2025] [Accepted: 03/26/2025] [Indexed: 04/04/2025]
Abstract
Tumor necrosis factor (TNF) is identified as a pro-inflammatory cytokine critical to the pathology of liver disease. In the carbohydrate metabolism, TNF has been demonstrated to impede the insulin signaling pathway, thereby precipitating glucose intolerance and insulin resistance. In lipid metabolism, TNF upregulates genes implicated in fatty acid synthesis, resulting in increased lipid accumulation within the liver. In amino acid metabolism, TNF has shown to promote the gene expression for amino acid catabolism, leading to decreased protein synthesis. Additionally, TNF stimulates the production of other chemokines and inflammatory cytokines that can further exacerbate liver injury. Overall, TNF is crucial in developing liver diseases by disrupting various metabolic pathways in the liver, causing insulin resistance, lipid accumulation, and decreased protein synthesis. This review summarizes the present understanding of TNF's role in the regulation of carbohydrate, lipid and amino acid metabolism in liver disease together with its potential therapeutic implications.
Collapse
Affiliation(s)
- Chuze Xu
- School of Optometry and Ophthalmology, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | | | - Xiaodi Zou
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Yanzhao Dong
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Ahmad Alhaskawi
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Haiying Zhou
- Faculty of Medicine, The Chinese University of Hong Kong School of Biomedical Science, Hong Kong, China
| | | | | | - Sahar Ahmed Abdalbary
- Department of Orthopedic Physical Therapy, Faculty of Physical Therapy, Nahda University, Beni Suef, Egypt
| | - Hui Lu
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University, Hangzhou, China; Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Zhejiang University, Hangzhou, China.
| |
Collapse
|
2
|
Gu M, Hong Q, Cui J, Li W, Zhang J, Sun Y, Jiang J, Hu Y. Harnessing lactobacillus: a probiotic revolution in combating metabolic dysfunction-associated steatotic liver disease. Crit Rev Food Sci Nutr 2025:1-18. [PMID: 40370039 DOI: 10.1080/10408398.2025.2504162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) involves fat buildup in the liver and is connected to a disruption in gut microbiota and a reduction in beneficial microbiota. Lactobacillus is an important probiotic that can grow in anaerobic or low-oxygen environment and is widely used in food industry and health fields. In recent years, the improvement of MASLD by Lactobacillus has been extensively studied. However, the detailed mechanisms by which different species of Lactobacillus improve MASLD have not been summarized. In this review, we present the potential of Lactobacillus as a non-drug approach to mitigate MASLD. We will discuss the preclinical and clinical research backing this method and the ways these probiotics improve MASLD. This review mainly presents the different species of Lactobacillus that improve MASLD, and various mechanisms by which Lactobacillus strains alleviate MASLD, including improving intestinal permeability and inflammation, reducing oxidative stress, and restoring of gut metabolites. Future research into mechanisms and larger clinical trials will help confirm the effectiveness of using Lactobacillus and related genera in the treatment of MASLD.
Collapse
Affiliation(s)
- Minwen Gu
- College of Biological and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, China
| | - Qing Hong
- State Key Laboratory of Dairy Biotechnology, Shanghai Engineering Research Center of Dairy Biotechnology, Dairy Research Institute, Bright Dairy & Food Co., Ltd, Shanghai, China
| | - Jie Cui
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing, China
- State Key Laboratory of Materials-Oriented Chemical Engineering, Nanjing Tech University, Nanjing, China
| | - Wenhui Li
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing, China
- State Key Laboratory of Materials-Oriented Chemical Engineering, Nanjing Tech University, Nanjing, China
| | - Jian Zhang
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing, China
- State Key Laboratory of Materials-Oriented Chemical Engineering, Nanjing Tech University, Nanjing, China
| | - Ye Sun
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing, China
- State Key Laboratory of Materials-Oriented Chemical Engineering, Nanjing Tech University, Nanjing, China
| | - Jinchi Jiang
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing, China
- State Key Laboratory of Materials-Oriented Chemical Engineering, Nanjing Tech University, Nanjing, China
| | - Yonghong Hu
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing, China
- State Key Laboratory of Materials-Oriented Chemical Engineering, Nanjing Tech University, Nanjing, China
| |
Collapse
|
3
|
He F, Du W, Liu Y, Ling Y, Xu M, Liu J, Song P, Fang Z, Yue Z, Duan J, Wang L. Exosome-equipped TNF antisense oligodeoxynucleotide or 2-deoxy-D-glucose ameliorated nonalcoholic steatohepatitis by modulating superoxide dismutase 1 in mice. Redox Biol 2025; 80:103488. [PMID: 39778469 PMCID: PMC11763583 DOI: 10.1016/j.redox.2025.103488] [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: 10/05/2024] [Revised: 12/09/2024] [Accepted: 01/02/2025] [Indexed: 01/11/2025] Open
Abstract
Inflammatory mediators tumor necrosis factor (TNF) and interleukin 1 beta (IL1β), primarily derived from hepatic macrophages in the liver, play a crucial role in the progression of nonalcoholic steatohepatitis (NASH). Meanwhile, intravenously injected exosomes are mainly distributed in the liver and predominantly taken up by hepatic macrophage. Herein, we aimed to evaluate the feasibility of targeted inhibition of TNF and IL1β expression in hepatic macrophages via exosomes as a potential therapeutic strategy for NASH. In this study, we demonstrated that antisense oligodeoxynucleotide targeting TNF (ASO-TNF) or 2-deoxy-d-glucose (2DG) effectively suppressed the expression of TNF and/or IL1β in macrophages. Exosomes loaded with ASO-TNF or 2DG were able to suppress the expression of TNF and/or IL1β in macrophages in vitro or in vivo. Furthermore, infusion of Exo/ASO-TNF or Exo/2DG significantly attenuated experimental steatohepatitis in choline deficient amino acid-defined (CDAA) or methionine and choline deficient (MCD) diet-fed mice. RNA-seq results showed that treatment with Exo/ASO-TNF or Exo/2DG significantly inhibited pro-inflammatory signaling pathways. Mechanistically, we observed that administration of Exo/ASO-TNF or Exo/2DG could attenuate NASH progression by up-regulating the expression of superoxide dismutase 1 (Sod1). Combined, our findings demonstrated that infusion of exosomes loaded with ASO-TNF or 2DG alleviated experimental steatohepatitis in murine models. Thus, infusion of exosomes loaded with anti-inflammatory agents holds promise as a potential therapeutic strategy for NASH treatment.
Collapse
Affiliation(s)
- Fei He
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, 710032, Xi'an, China.
| | - Wei Du
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, 710032, Xi'an, China.
| | - Yingying Liu
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, 710032, Xi'an, China.
| | - Yuwei Ling
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, 710032, Xi'an, China.
| | - Ming Xu
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, 710032, Xi'an, China.
| | - Jingjing Liu
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, 710032, Xi'an, China.
| | - Ping Song
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, 710032, Xi'an, China.
| | - Zhiqiang Fang
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, 710032, Xi'an, China.
| | - Zhensheng Yue
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, 710032, Xi'an, China.
| | - Juanli Duan
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, 710032, Xi'an, China.
| | - Lin Wang
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, 710032, Xi'an, China.
| |
Collapse
|
4
|
Campos-Bayardo TI, Román-Rojas D, García-Sánchez A, Cardona-Muñoz EG, Sánchez-Lozano DI, Totsuka-Sutto S, Gómez-Hermosillo LF, Casillas-Moreno J, Andrade-Sierra J, Pazarín-Villaseñor L, Campos-Pérez W, Martínez-López E, Miranda-Díaz AG. The Role of TLRs in Obesity and Its Related Metabolic Disorders. Int J Mol Sci 2025; 26:2229. [PMID: 40076851 PMCID: PMC11900219 DOI: 10.3390/ijms26052229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 02/24/2025] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
Obesity affects the adaptability of adipose tissue (AT), impairing its ability to regulate energy and metabolism. Obesity is associated with many metabolic disorders, including dyslipidemia, hypertension, sleep disorders, non-alcoholic liver disease, and some types of cancer. Toll-like receptors (TLRs) are important in obesity and related metabolic disorders. TLRs are pattern-recognizing receptors (PRRs) involved in the innate immune system and recognize pathogen-associated molecular patterns (PAMPs) and endogenous ligands. TLRs, especially TLR2 and TLR4, are activated by fatty acids, endotoxins, and other ligands. TLR2 and TLR4 activation triggers inflammatory responses. Chronic inflammation driven by TLR activation is a hallmark of obesity and metabolic diseases. The inflammatory response triggered by TLR activation alters insulin signaling, contributing to insulin resistance, a key feature of metabolic syndrome and type 2 diabetes. Modulation of TLR activity through lifestyle changes (diet and exercise), obesity surgery, and pharmacological agents is under study as a possible therapeutic approach to controlling obesity and its complications.
Collapse
Affiliation(s)
- Tannia Isabel Campos-Bayardo
- Department of Physiology, University Center of Health Sciences, University of Guadalajara, Guadalajara 44360, Jalisco, Mexico; (T.I.C.-B.); (D.R.-R.); (A.G.-S.); (E.G.C.-M.); (D.I.S.-L.); (S.T.-S.)
| | - Daniel Román-Rojas
- Department of Physiology, University Center of Health Sciences, University of Guadalajara, Guadalajara 44360, Jalisco, Mexico; (T.I.C.-B.); (D.R.-R.); (A.G.-S.); (E.G.C.-M.); (D.I.S.-L.); (S.T.-S.)
| | - Andrés García-Sánchez
- Department of Physiology, University Center of Health Sciences, University of Guadalajara, Guadalajara 44360, Jalisco, Mexico; (T.I.C.-B.); (D.R.-R.); (A.G.-S.); (E.G.C.-M.); (D.I.S.-L.); (S.T.-S.)
| | - Ernesto Germán Cardona-Muñoz
- Department of Physiology, University Center of Health Sciences, University of Guadalajara, Guadalajara 44360, Jalisco, Mexico; (T.I.C.-B.); (D.R.-R.); (A.G.-S.); (E.G.C.-M.); (D.I.S.-L.); (S.T.-S.)
| | - Daniela Itzel Sánchez-Lozano
- Department of Physiology, University Center of Health Sciences, University of Guadalajara, Guadalajara 44360, Jalisco, Mexico; (T.I.C.-B.); (D.R.-R.); (A.G.-S.); (E.G.C.-M.); (D.I.S.-L.); (S.T.-S.)
| | - Sylvia Totsuka-Sutto
- Department of Physiology, University Center of Health Sciences, University of Guadalajara, Guadalajara 44360, Jalisco, Mexico; (T.I.C.-B.); (D.R.-R.); (A.G.-S.); (E.G.C.-M.); (D.I.S.-L.); (S.T.-S.)
| | - Luis Francisco Gómez-Hermosillo
- Department of Laparoscopic Surgery, Hospital Civil de Guadalajara, “Juan I Menchaca”, Guadalajara 44360, Jalisco, Mexico; (L.F.G.-H.); (J.C.-M.)
| | - Jorge Casillas-Moreno
- Department of Laparoscopic Surgery, Hospital Civil de Guadalajara, “Juan I Menchaca”, Guadalajara 44360, Jalisco, Mexico; (L.F.G.-H.); (J.C.-M.)
| | - Jorge Andrade-Sierra
- Department of Nephrology, National Medical Center of the West, Mexican Social Security Institute, Guadalajara 44340, Jalisco, Mexico; (J.A.-S.); (L.P.-V.)
| | - Leonardo Pazarín-Villaseñor
- Department of Nephrology, National Medical Center of the West, Mexican Social Security Institute, Guadalajara 44340, Jalisco, Mexico; (J.A.-S.); (L.P.-V.)
| | - Wendy Campos-Pérez
- Department of Molecular Biology and Genomics, Institute of Nutrigenetics and Translational Nutrigenomics, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico; (W.C.-P.); (E.M.-L.)
| | - Erika Martínez-López
- Department of Molecular Biology and Genomics, Institute of Nutrigenetics and Translational Nutrigenomics, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico; (W.C.-P.); (E.M.-L.)
| | - Alejandra Guillermina Miranda-Díaz
- Department of Physiology, University Center of Health Sciences, University of Guadalajara, Guadalajara 44360, Jalisco, Mexico; (T.I.C.-B.); (D.R.-R.); (A.G.-S.); (E.G.C.-M.); (D.I.S.-L.); (S.T.-S.)
| |
Collapse
|
5
|
Rondanelli M, Borromeo S, Cavioni A, Gasparri C, Gattone I, Genovese E, Lazzarotti A, Minonne L, Moroni A, Patelli Z, Razza C, Sivieri C, Valentini EM, Barrile GC. Therapeutic Strategies to Modulate Gut Microbial Health: Approaches for Chronic Metabolic Disorder Management. Metabolites 2025; 15:127. [PMID: 39997751 PMCID: PMC11857149 DOI: 10.3390/metabo15020127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 01/17/2025] [Accepted: 01/28/2025] [Indexed: 02/26/2025] Open
Abstract
Numerous recent studies have suggested that the composition of the intestinal microbiota can trigger metabolic disorders, such as diabetes, prediabetes, obesity, metabolic syndrome, sarcopenia, dyslipidemia, hyperhomocysteinemia, and non-alcoholic fatty liver disease. Since then, considerable effort has been made to understand the link between the composition of intestinal microbiota and metabolic disorders, as well as the role of probiotics in the modulation of the intestinal microbiota. The aim of this review was to summarize the reviews and individual articles on the state of the art regarding ideal therapy with probiotics and prebiotics in order to obtain the reversion of dysbiosis (alteration in microbiota) to eubiosis during metabolic diseases, such as diabetes, prediabetes, obesity, hyperhomocysteinemia, dyslipidemia, sarcopenia, and non-alcoholic fatty liver diseases. This review includes 245 eligible studies. In conclusion, a condition of dysbiosis, or in general, alteration of the intestinal microbiota, could be implicated in the development of metabolic disorders through different mechanisms, mainly linked to the release of pro-inflammatory factors. Several studies have already demonstrated the potential of using probiotics and prebiotics in the treatment of this condition, detecting significant improvements in the specific symptoms of metabolic diseases. These findings reinforce the hypothesis that a condition of dysbiosis can lead to a generalized inflammatory picture with negative consequences on different organs and systems. Moreover, this review confirms that the beneficial effects of probiotics on metabolic diseases are promising, but more research is needed to determine the optimal probiotic strains, doses, and administration forms for specific metabolic conditions.
Collapse
Affiliation(s)
- Mariangela Rondanelli
- Department of Public Health, Experimental and Forensic Medicine, University of Pavia, 27100 Pavia, Italy;
| | - Sara Borromeo
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Alessandro Cavioni
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Clara Gasparri
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Ilaria Gattone
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Elisa Genovese
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Alessandro Lazzarotti
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Leonardo Minonne
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Alessia Moroni
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Zaira Patelli
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Claudia Razza
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Claudia Sivieri
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Eugenio Marzio Valentini
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Gaetan Claude Barrile
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| |
Collapse
|
6
|
D’Ambrosio R, Cavallo S, Brunetti R, Pellicanò R, Vaccaro E, Borriello G, Paradiso R, Serpe FP, Lambiase S, Bruzzese F, Palma G, Rea D, Barbieri A, D’Amore M, Dimatteo M, degli Uberti B, Paciello O, Baldi L. The Use of Antimicrobials in Animal Husbandry as a Potential Factor for the Increased Incidence of Colorectal Cancer: Food Safety and Kinetics in a Murine Model. Animals (Basel) 2025; 15:315. [PMID: 39943084 PMCID: PMC11815752 DOI: 10.3390/ani15030315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 01/17/2025] [Accepted: 01/21/2025] [Indexed: 02/16/2025] Open
Abstract
The aim of this research was to investigate the effects of the prolonged use of the broad-spectrum antimicrobial widely used in animal husbandry. By means of a mouse model, a translational study was carried out on immunocompetent mice (with a complete immune system). This study highlighted the effect of antimicrobial residues taken in with food on the growth time of cancer and on alterations to the gut microbiota. This project considered the fight against antimicrobial resistance from a One Health perspectivethrough collaboration between human medicine and veterinary medicine. Regarding food safety, antimicrobial residues in products of animal origin are rarely detected; they therefore constitute a negligible factor in determining colorectal cancer.
Collapse
Affiliation(s)
- Rosa D’Ambrosio
- Istituto Zooprofilattico Sperimentale del Mezzogiorno (IZSM), 80055 Portici, Italy; (R.D.); (S.C.); (R.P.); (G.B.); (R.P.); (F.P.S.); (S.L.); (M.D.); (B.d.U.); (L.B.)
| | - Stefania Cavallo
- Istituto Zooprofilattico Sperimentale del Mezzogiorno (IZSM), 80055 Portici, Italy; (R.D.); (S.C.); (R.P.); (G.B.); (R.P.); (F.P.S.); (S.L.); (M.D.); (B.d.U.); (L.B.)
| | - Roberta Brunetti
- Istituto Zooprofilattico Sperimentale del Mezzogiorno (IZSM), 80055 Portici, Italy; (R.D.); (S.C.); (R.P.); (G.B.); (R.P.); (F.P.S.); (S.L.); (M.D.); (B.d.U.); (L.B.)
| | - Roberta Pellicanò
- Istituto Zooprofilattico Sperimentale del Mezzogiorno (IZSM), 80055 Portici, Italy; (R.D.); (S.C.); (R.P.); (G.B.); (R.P.); (F.P.S.); (S.L.); (M.D.); (B.d.U.); (L.B.)
| | - Emanuela Vaccaro
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, 80137 Napoli, Italy; (E.V.); (O.P.)
| | - Giorgia Borriello
- Istituto Zooprofilattico Sperimentale del Mezzogiorno (IZSM), 80055 Portici, Italy; (R.D.); (S.C.); (R.P.); (G.B.); (R.P.); (F.P.S.); (S.L.); (M.D.); (B.d.U.); (L.B.)
| | - Rubina Paradiso
- Istituto Zooprofilattico Sperimentale del Mezzogiorno (IZSM), 80055 Portici, Italy; (R.D.); (S.C.); (R.P.); (G.B.); (R.P.); (F.P.S.); (S.L.); (M.D.); (B.d.U.); (L.B.)
| | - Francesco Paolo Serpe
- Istituto Zooprofilattico Sperimentale del Mezzogiorno (IZSM), 80055 Portici, Italy; (R.D.); (S.C.); (R.P.); (G.B.); (R.P.); (F.P.S.); (S.L.); (M.D.); (B.d.U.); (L.B.)
| | - Sara Lambiase
- Istituto Zooprofilattico Sperimentale del Mezzogiorno (IZSM), 80055 Portici, Italy; (R.D.); (S.C.); (R.P.); (G.B.); (R.P.); (F.P.S.); (S.L.); (M.D.); (B.d.U.); (L.B.)
| | - Francesca Bruzzese
- Experimental Animal Unit, Istituto Nazionale Tumori-IRCCS-Fondazione “G. Pascale”, 80131 Naples, Italy; (F.B.); (G.P.)
| | - Giuseppe Palma
- Experimental Animal Unit, Istituto Nazionale Tumori-IRCCS-Fondazione “G. Pascale”, 80131 Naples, Italy; (F.B.); (G.P.)
| | - Domenica Rea
- Laboratory Medicine Unit, Istituto Nazionale Tumori- IRCCS- Fondazione “G. Pascale”, 80131 Naples, Italy;
| | - Antonio Barbieri
- ASL Salerno UOC Laboratorio d’Analisi, Vallo della Lucania, 84078 Salerno, Italy
| | - Marianna D’Amore
- Istituto Zooprofilattico Sperimentale del Mezzogiorno (IZSM), 80055 Portici, Italy; (R.D.); (S.C.); (R.P.); (G.B.); (R.P.); (F.P.S.); (S.L.); (M.D.); (B.d.U.); (L.B.)
| | - Maria Dimatteo
- Istituto Zooprofilattico Sperimentale del Mezzogiorno (IZSM), 80055 Portici, Italy; (R.D.); (S.C.); (R.P.); (G.B.); (R.P.); (F.P.S.); (S.L.); (M.D.); (B.d.U.); (L.B.)
| | - Barbara degli Uberti
- Istituto Zooprofilattico Sperimentale del Mezzogiorno (IZSM), 80055 Portici, Italy; (R.D.); (S.C.); (R.P.); (G.B.); (R.P.); (F.P.S.); (S.L.); (M.D.); (B.d.U.); (L.B.)
| | - Orlando Paciello
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, 80137 Napoli, Italy; (E.V.); (O.P.)
| | - Loredana Baldi
- Istituto Zooprofilattico Sperimentale del Mezzogiorno (IZSM), 80055 Portici, Italy; (R.D.); (S.C.); (R.P.); (G.B.); (R.P.); (F.P.S.); (S.L.); (M.D.); (B.d.U.); (L.B.)
| |
Collapse
|
7
|
Wang LJ, Sun JG, Chen SC, Sun YL, Zheng Y, Feng JC. The role of intestinal flora in metabolic dysfunction-associated steatotic liver disease and treatment strategies. Front Med (Lausanne) 2025; 11:1490929. [PMID: 39839647 PMCID: PMC11746088 DOI: 10.3389/fmed.2024.1490929] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 11/28/2024] [Indexed: 01/23/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a common multi-factorial liver disease, and its incidence is gradually increasing worldwide. Many reports have revealed that intestinal flora plays a crucial role for the occurrence and development of MASLD, through mechanisms such as flora translocation, endogenous ethanol production, dysregulation of choline metabolism and bile acid, and endotoxemia. Here, we review the relationship between intestinal flora and MASLD, as well as interventions for MASLD, such as prebiotics, probiotics, synbiotics, and intestinal flora transplantation. Intervention strategies targeting the intestinal flora along with its metabolites may be new targets for preventing and treating MASLD.
Collapse
Affiliation(s)
- Li Jun Wang
- Department of Traditional Chinese Medicine, Binzhou Medical University, Yantai, China
| | - Jian Guang Sun
- The First Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Shu Cheng Chen
- School of Nursing, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Yu Li Sun
- Department of Hepatology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yang Zheng
- Department of Acupuncture and Moxibustion, Zibo Hospital, Zibo, China
| | - Jian Chao Feng
- Department of Acupuncture and Moxibustion, Zibo Hospital, Zibo, China
| |
Collapse
|
8
|
Mushraf S, Chawla K, Fayaz SMA, Mathew AJ, Reddy GPK, Kappettu Gadahad MR, Shenoy PA, Devi V, Adiga S, Nayak V. Exploring the effects of probiotics on olanzapine-induced metabolic syndrome through the gut microbiota. Gut Pathog 2024; 16:77. [PMID: 39709451 DOI: 10.1186/s13099-024-00664-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 11/07/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND Maintaining gut microbial homeostasis is crucial for human health, as imbalances in the gut microbiota (GM) can lead to various diseases, including metabolic syndrome (MS), exacerbated by the use of antipsychotic medications such as olanzapine (OLZ). Understanding the role of the GM in OLZ-induced MS could lead to new therapeutic strategies. This study used metagenomic analysis to explore the impact of OLZ on the GM composition and examined how probiotics can mitigate its adverse effects in a rat model. Changes in weight, blood pressure, and lipid levels, which are key parameters defining MS, were assessed. Additionally, this study investigated serotonin, dopamine, and histopathological changes to explore their possible link with the microbiota-gut-brain axis (MGBA). RESULTS OLZ had an antagonistic effect on serotonin and dopamine receptors, and it was consistently found to alter the composition of the GM, with an increase in the relative abundance (RA) of the Firmicutes/Bacteroidetes phyla ratio and TM7 genera, indicating that the anticommonsal action of OLZ affects appetite and energy expenditure, contributing to obesity, dyslipidemia and increased blood pressure, which are core components of MS. Hepatic steatosis and intestinal damage in OLZ-treated rat tissues further indicate its role in MS. Conversely, the administration of probiotics, either alone or in combination with OLZ, was found to mitigate these OLZ-induced symptoms of MS by altering the GM composition. These alterations included increases in the abundances of the taxa Bacteroidetes, Actinobacteria, Prevotella, Blautia, Bacteroides, Bacteroidales, and Ruminococcaceae and a decrease in Firmicute abundance. These changes helped maintain gut barrier integrity and modulated neurotransmitter levels, suggesting that probiotics can counteract the adverse metabolic effects of OLZ by restoring the GM balance. Moreover, this study highlights the modulation of the MGBA by OLZ as a potential mechanism through which probiotics modulate serotonin and dopamine levels, influencing metabolic health. CONCLUSION These findings emphasise the significant impact of OLZ on the GM and its contribution to MS. These findings suggest that interventions targeting the GM, such as probiotics, could mitigate the metabolic side effects of OLZ. Future research should focus on developing integrative treatment approaches that consider the health of the gut microbiome in managing antipsychotic-induced adverse effects.
Collapse
Affiliation(s)
- Syed Mushraf
- Division of Pharmacology, Department of Basic Medical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Kiran Chawla
- Department of Microbiology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Shaik Mohammed Abdul Fayaz
- Department of Biotechnology, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| | - Aranjani Jesil Mathew
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Gayam Prasanna Kumar Reddy
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Mohandas Rao Kappettu Gadahad
- Division of Anatomy, Department of Basic Medical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Padmaja A Shenoy
- Department of Microbiology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Vasudha Devi
- Department of Pharmacology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Shalini Adiga
- Department of Pharmacology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Veena Nayak
- Department of Pharmacology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| |
Collapse
|
9
|
Guraka A, Sreedharan S, Arasaradnam R, Tripathi G, Kermanizadeh A. The Role of the Gut Microbiome in the Development and Progression of Type 2 Diabetes and Liver Disease. Nutr Rev 2024:nuae172. [PMID: 39673297 DOI: 10.1093/nutrit/nuae172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2024] Open
Abstract
Type 2 diabetes mellitus (T2DM) and progressive liver disease are 2 of the most significant global health concerns, and they have alarming and ever-increasing prevalence. A growing body of literature has demonstrated a potential multilateral link between gut microbiome dysbiosis and the development and progression of the above-mentioned conditions. Modulation of gut microbial composition from the norm is due to changes in diet allied with external factors such as age, genetics, and environmental changes. In this comprehensive review, we recapitulate the research to date investigating the links between gut microbiome dysbiosis and T2DM or liver disease, with special attention to the importance of diet. Additionally, we review the most commonly used tools and methodologies of investigating changes in the gut microbiome, highlighting the advantages and limitations of each strategy, before introducing a novel in vitro approach to the problem. Finally, the review offers recommendations for future research in this field that will allow better understanding of how the gut microbiota affects disease progression and of the prospects for intestinal microbiota-based therapeutic options.
Collapse
Affiliation(s)
- Asha Guraka
- University of Derby, College of Science and Engineering, Derby, DE22 1GB, United Kingdom
| | - Sreejesh Sreedharan
- University of Derby, College of Science and Engineering, Derby, DE22 1GB, United Kingdom
| | - Ramesh Arasaradnam
- University of Warwick, Warick Medical School, Warwick, CV4 7AL, United Kingdom
| | - Gyan Tripathi
- Nottingham Trent University, School of Science and Technology, Nottingham, NG18 5BH, United Kingdom
| | - Ali Kermanizadeh
- University of Derby, College of Science and Engineering, Derby, DE22 1GB, United Kingdom
| |
Collapse
|
10
|
Scarpellini E, Scarcella M, Tack JF, Scarlata GGM, Zanetti M, Abenavoli L. Gut Microbiota and Metabolic Dysfunction-Associated Steatotic Liver Disease. Antioxidants (Basel) 2024; 13:1386. [PMID: 39594528 PMCID: PMC11591341 DOI: 10.3390/antiox13111386] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/24/2024] [Accepted: 10/30/2024] [Indexed: 11/28/2024] Open
Abstract
Background: The gut microbiota constitutes a complex microorganism community that harbors bacteria, viruses, fungi, protozoa, and archaea. The human gut bacterial microbiota has been extensively proven to participate in human metabolism, immunity, and nutrient absorption. Its imbalance, namely "dysbiosis", has been linked to disordered metabolism. Metabolic dysfunction-associated steatotic liver disease (MASLD) is one of the features of deranged human metabolism and is the leading cause of liver cirrhosis and hepatocellular carcinoma. Thus, there is a pathophysiological link between gut dysbiosis and MASLD. Aims and Methods: We aimed to review the literature data on the composition of the human bacterial gut microbiota and its dysbiosis in MASLD and describe the concept of the "gut-liver axis". Moreover, we reviewed the approaches for gut microbiota modulation in MASLD treatment. Results: There is consolidated evidence of particular gut dysbiosis associated with MASLD and its stages. The model explaining the relationship between gut microbiota and the liver has a bidirectional organization, explaining the physiopathology of MASLD. Oxidative stress is one of the keystones in the pathophysiology of MASLD and fibrosis generation. There is promising and consolidated evidence for the efficacy of pre- and probiotics in reversing gut dysbiosis in MASLD patients, with therapeutic effects. Few yet encouraging data on fecal microbiota transplantation (FMT) in MASLD are available in the literature. Conclusions: The gut dysbiosis characteristic of MASLD is a key target in its reversal and treatment via diet, pre/probiotics, and FMT treatment. Oxidative stress modulation remains a promising target for MASLD treatment, prevention, and reversal.
Collapse
Affiliation(s)
- Emidio Scarpellini
- Translational Research in Gastroeintestinal Disorders, Gasthuisberg University Hospital, KULeuven, Herestraat 49, 3000 Lueven, Belgium;
| | - Marialaura Scarcella
- Anesthesia, Intensive Care and Nutritional Science-Azienda Ospedaliera “Santa Maria”, Via Tristano di Joannuccio, 05100 Terni, Italy;
| | - Jan F. Tack
- Translational Research in Gastroeintestinal Disorders, Gasthuisberg University Hospital, KULeuven, Herestraat 49, 3000 Lueven, Belgium;
| | | | - Michela Zanetti
- Geriatrics Department, Nutrition and Malnutrition Unit, Azienda Sanitario-Universitaria Giuliano Isontina, Ospedale Maggiore, piazza dell’Ospitale 1, 34100 Triste, Italy;
| | - Ludovico Abenavoli
- Department of Health Sciences, University “Magna Graecia”, 88100 Catanzaro, Italy; (G.G.M.S.); (L.A.)
| |
Collapse
|
11
|
Lei L, Li J, Liu Z, Zhang D, Liu Z, Wang Q, Gao Y, Mo B, Li J. Identification of diagnostic markers pyrodeath-related genes in non-alcoholic fatty liver disease based on machine learning and experiment validation. Sci Rep 2024; 14:25541. [PMID: 39462099 PMCID: PMC11513955 DOI: 10.1038/s41598-024-77409-3] [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: 08/04/2024] [Accepted: 10/22/2024] [Indexed: 10/28/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) poses a global health challenge. While pyroptosis is implicated in various diseases, its specific involvement in NAFLD remains unclear. Thus, our study aims to elucidate the role and mechanisms of pyroptosis in NAFLD. Utilizing data from the Gene Expression Omnibus (GEO) database, we analyzed the expression levels of pyroptosis-related genes (PRGs) in NAFLD and normal tissues using the R data package. We investigated protein interactions, correlations, and functional enrichment of these genes. Key genes were identified employing multiple machine learning techniques. Immunoinfiltration analyses were conducted to discern differences in immune cell populations between NAFLD patients and controls. Key gene expression was validated using a cell model. Analysis of GEO datasets, comprising 206 NAFLD samples and 10 controls, revealed two key PRGs (TIRAP, and GSDMD). Combining these genes yielded an area under the curve (AUC) of 0.996 for diagnosing NAFLD. In an external dataset, the AUC for the two key genes was 0.825. Nomogram, decision curve, and calibration curve analyses further validated their diagnostic efficacy. These genes were implicated in multiple pathways associated with NAFLD progression. Immunoinfiltration analysis showed significantly lower numbers of various immune cell types in NAFLD patient samples compared to controls. Single sample gene set enrichment analysis (ssGSEA) was employed to assess the immune microenvironment. Finally, the expression of the two key genes was validated in cell NAFLD model using qRT-PCR. We developed a prognostic model for NAFLD based on two PRGs, demonstrating robust predictive efficacy. Our findings enhance the understanding of pyroptosis in NAFLD and suggest potential avenues for therapeutic exploration.
Collapse
Affiliation(s)
- Liping Lei
- Department of Geriatric Medicine, The Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi, China
- Division of Hepatobiliary Surgery, The Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi, China
| | - Jixue Li
- Division of Hepatobiliary Surgery, The Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi, China
| | - Zirui Liu
- Division of Hepatobiliary Surgery, The Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi, China
| | - Dongdong Zhang
- Division of Hepatobiliary Surgery, The Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi, China
| | - Zihan Liu
- Division of Hepatobiliary Surgery, The Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi, China
| | - Qing Wang
- Division of Hepatobiliary Surgery, The Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi, China
| | - Yi Gao
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi, China
| | - Biwen Mo
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Guilin Medical University, Guilin, 541002, Guangxi, China.
| | - Jiangfa Li
- Division of Hepatobiliary Surgery, The Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi, China.
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Ministry of Education, Nanning, 530021, Guangxi, China.
- Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Nanning, 530021, Guangxi, China.
| |
Collapse
|
12
|
Yaghmaei H, Bahanesteh A, Soltanipur M, Takaloo S, Rezaei M, Siadat SD. The Role of Gut Microbiota Modification in Nonalcoholic Fatty Liver Disease Treatment Strategies. Int J Hepatol 2024; 2024:4183880. [PMID: 39444759 PMCID: PMC11498984 DOI: 10.1155/2024/4183880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 08/25/2024] [Accepted: 10/05/2024] [Indexed: 10/25/2024] Open
Abstract
One of the most common chronic liver diseases is nonalcoholic fatty liver disease (NAFLD), which affects many people around the world. Gut microbiota (GM) dysbiosis seems to be an influential factor in the pathophysiology of NAFLD because changes in GM lead to fundamental changes in host metabolism. Therefore, the study of the effect of dysbiosis on the pathogenicity of NAFLD is important. European clinical guidelines state that the best advice for people with NAFLD is to lose weight and improve their lifestyle, but only 40% of people can achieve this goal. Accordingly, it is necessary to provide new treatment approaches for prevention and treatment. In addition to dietary interventions and lifestyle modifications, GM modification-based therapies are of interest. These therapies include probiotics, synbiotics, fecal microbiota transplantation (FMT), and next-generation probiotics. All of these treatments have had promising results in animal studies, and it can be imagined that acceptable results will be obtained in human studies as well. However, further investigations are required to generalize the outcomes of animal studies to humans.
Collapse
Affiliation(s)
- Hessam Yaghmaei
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
| | | | - Masood Soltanipur
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
- Cardiovascular Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sobhan Takaloo
- Biomedical Engineering Department, Hamedan University of Technology, Hamedan, Iran
| | - Mahdi Rezaei
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
- Cardiovascular Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Davar Siadat
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
- Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
13
|
Allen AM, Younossi ZM, Diehl AM, Charlton MR, Lazarus JV. Envisioning how to advance the MASH field. Nat Rev Gastroenterol Hepatol 2024; 21:726-738. [PMID: 38834817 DOI: 10.1038/s41575-024-00938-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/02/2024] [Indexed: 06/06/2024]
Abstract
Since 1980, the cumulative effort of scientists and health-care stakeholders has advanced the prerequisites to address metabolic dysfunction-associated steatotic liver disease (MASLD), a prevalent chronic non-communicable liver disease. This effort has led to, among others, the approval of the first drug specific for metabolic dysfunction-associated steatohepatitis (MASH; formerly known as nonalcoholic steatohepatitis). Despite substantial progress, MASLD is still a leading cause of advanced chronic liver disease, including primary liver cancer. This Perspective contextualizes the nomenclature change from nonalcoholic fatty liver disease to MASLD and proposes important considerations to accelerate further progress in the field, optimize patient-centric multidisciplinary care pathways, advance pharmacological, behavioural and diagnostic research, and address health disparities. Key regulatory and other steps necessary to optimize the approval and access to upcoming additional pharmacological therapeutic agents for MASH are also outlined. We conclude by calling for increased education and awareness, enhanced health system preparedness, and concerted action by policy-makers to further the public health and policy agenda to achieve at least parity with other non-communicable diseases and to aid in growing the community of practice to reduce the human and economic burden and end the public health threat of MASLD and MASH by 2030.
Collapse
Affiliation(s)
- Alina M Allen
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Zobair M Younossi
- Beatty Liver and Obesity Research Program, Inova Health System, Falls Church, VA, USA
- The Global NASH Council, Washington DC, USA
| | | | - Michael R Charlton
- Center for Liver Diseases, Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Jeffrey V Lazarus
- The Global NASH Council, Washington DC, USA.
- CUNY Graduate School of Public Health and Health Policy (CUNY SPH), New York, NY, USA.
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic, University of Barcelona, Barcelona, Spain.
- Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain.
| |
Collapse
|
14
|
Chen LZ, Jing XB, Chen X, Xie YC, Chen Y, Cai XB. Non-Invasive Serum Markers of Non-Alcoholic Fatty Liver Disease Fibrosis: Potential Tools for Detecting Patients with Cardiovascular Disease. Rev Cardiovasc Med 2024; 25:344. [PMID: 39355605 PMCID: PMC11440407 DOI: 10.31083/j.rcm2509344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 05/23/2024] [Accepted: 05/28/2024] [Indexed: 10/03/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD), one of the most common chronic liver diseases with a prevalence of 23%-25% globally, is an independent risk factor for cardiovascular diseases (CVDs). Growing evidence indicates that the development of NAFLD, ranging from non-alcoholic fatty liver (NAFL), non-alcoholic steatohepatitis (NASH), advanced fibrosis to cirrhosis, and even hepatocellular carcinoma, is at substantial risk for CVDs, which clinically contribute to increased cardiovascular morbidity and mortality. Non-invasive serum markers assessing liver fibrosis, such as fibrosis-4 (FIB-4) score, aspartate transaminase-to-platelet ratio index (APRI), and NAFLD fibrosis score (NFS), are expected to be useful tools for clinical management of patients with CVDs. This review aims to provide an overview of the evidence for the relationship between the progression of NAFLD and CVDs and the clinical application of non-invasive markers of liver fibrosis in managing patients with CVDs.
Collapse
Affiliation(s)
- Ling-Zi Chen
- Department of Gastroenterology, The First Affiliated Hospital of Shantou University Medical College, 515041 Shantou, Guangdong, China
| | - Xu-Bin Jing
- Department of Gastroenterology, The First Affiliated Hospital of Shantou University Medical College, 515041 Shantou, Guangdong, China
| | - Xiang Chen
- Department of Gastroenterology, The First Affiliated Hospital of Shantou University Medical College, 515041 Shantou, Guangdong, China
| | - Yan-Chun Xie
- Department of Endoscopy Center, Cancer Hospital of Shantou University Medical College, 515041 Shantou, Guangdong, China
| | - Yun Chen
- Department of Gastroenterology, The First Affiliated Hospital of Shantou University Medical College, 515041 Shantou, Guangdong, China
| | - Xian-Bin Cai
- Department of Gastroenterology, The First Affiliated Hospital of Shantou University Medical College, 515041 Shantou, Guangdong, China
| |
Collapse
|
15
|
Alam N, Jia L, Cheng A, Ren H, Fu Y, Ding X, Haq IU, Liu E. Global research trends on gut microbiota and metabolic dysfunction-associated steatohepatitis: Insights from bibliometric and scientometric analysis. Front Pharmacol 2024; 15:1390483. [PMID: 39070791 PMCID: PMC11273336 DOI: 10.3389/fphar.2024.1390483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 06/24/2024] [Indexed: 07/30/2024] Open
Abstract
Background Metabolic dysfunction-associated steatohepatitis (MASH) is an inflammatory subtype of metabolic dysfunction-associated steatotic liver disease (MASLD) has recently been proposed as a replacement term for NAFLD, a common, multifactorial and poorly understood liver disease whose incidence is increasing worldwide. In recent years, there has been increasing scientific interest in exploring the relationship between gut microbiota and MASH. To learn more about the gut microbiota in MASH, this study aims to provide a comprehensive analysis of the knowledge structure and research hotspots from a bibliometric perspective. Methods We searched the Web of Science Core Collection for articles and reviews that covered the connections between gut microbiota and MASH over the last decade. The Online Analysis Platforms, VOSviewer, CiteSpace, the R tool "bibliometrix" were used to analyzed existing publications trends and hotspots. Results A total of 4,069 documents related to the interaction between gut microbiota and MASH were retrieved from 2014 to 2023. The number of annual publications increased significantly over the last decade, particularly in the United States and China. The University of California-San Diego was the most productive institution, while researcher Rohit Loomba published the most papers in the field. Younossi ZM was ranked as the first co-cited author and largest contributor of highly cited articles in the field. Gastroenterology and hepatology were the most common specialty category. The most cited journal in the last decade was Hepatology. The Keyword Bursts analysis highlighted the importance of studying the association between gut microbiota and MASH, as well as related factors such as metabolic syndrome, insulin resistance, endotoxemia and overgrowth of gut bacteria. Keyword clusters with co-citation were used to illustrate important topics including intestinal permeability, insulin sensitivity and liver immunology. The most common keywords include insulin resistance, obesity, dysbiosis, inflammation and oxidative stress, which are current hotspots. Conclusion Our analysis highlights key aspects of this field and emphasizes multiorgan crosstalk in MASLD/MASH pathogenesis. In particular, the central role of the gut-liver axis and the significant influence of gut microbiota dysbiosis on disease progression are highlighted. Furthermore, our results highlight the transformative potential of microbiota-specific therapies and cover the way for innovative healthcare and pharmaceutical strategies.
Collapse
Affiliation(s)
- Naqash Alam
- Laboratory Animal Center, School of Basic Medical Sciences, Health Science Center, Xi’an Jiaotong University, Xi’an, China
| | - Linying Jia
- Laboratory Animal Center, School of Basic Medical Sciences, Health Science Center, Xi’an Jiaotong University, Xi’an, China
| | - Ao Cheng
- Laboratory Animal Center, School of Basic Medical Sciences, Health Science Center, Xi’an Jiaotong University, Xi’an, China
| | - Honghao Ren
- Laboratory Animal Center, School of Basic Medical Sciences, Health Science Center, Xi’an Jiaotong University, Xi’an, China
| | - Yu Fu
- Laboratory Animal Center, School of Basic Medical Sciences, Health Science Center, Xi’an Jiaotong University, Xi’an, China
| | - Xinhua Ding
- Laboratory Animal Center, School of Basic Medical Sciences, Health Science Center, Xi’an Jiaotong University, Xi’an, China
| | - Ihtisham Ul Haq
- Department of Neurobiology, School of Basic Medical Sciences, Health Science Center, Xi’an Jiaotong University, Xi’an, China
| | - Enqi Liu
- Laboratory Animal Center, School of Basic Medical Sciences, Health Science Center, Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
16
|
Yang Y, Yang L, Wu J, Hu J, Wan M, Bie J, Li J, Pan D, Sun G, Yang C. Optimal probiotic combinations for treating nonalcoholic fatty liver disease: A systematic review and network meta-analysis. Clin Nutr 2024; 43:1224-1239. [PMID: 38643738 DOI: 10.1016/j.clnu.2024.04.004] [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: 08/22/2023] [Revised: 03/20/2024] [Accepted: 04/01/2024] [Indexed: 04/23/2024]
Abstract
BACKGROUND Probiotic administration is a promising therapy for improving conditions in NAFLD patients. This network meta-analysis aimed to compare and estimate the relative effects of probiotic interventions and identify the optimal probiotic species for the treatment of NAFLD (Nonalcoholic fatty liver disease) patients. METHODS The PubMed, Web of Science, Embase, and Cochrane databases were searched from inception to 29 January 2024 to identify RCTs that were published in English. The GRADE framework was used to assess the quality of evidence contributing to each network estimate. RESULTS A total of 35 RCTs involving 2212 NAFLD patients were included in the analysis. For primary outcomes, Lactobacillus + Bifidobacterium + Streptococcus exhibited the highest probability of being the finest probiotic combination in terms of enhancing acceptability as well as reducing AST (SMD: -1.95 95% CI: -2.90, -0.99), ALT (SMD = -1.67, 95% CI: -2.48, -0.85), and GGT levels (SMD = -2.17, 95% CI: -3.27, -1.06). In terms of the secondary outcomes, Lactobacillus + Bifidobacterium + Streptococcus was also the best probiotic combination for reducing BMI (SMD = -0.45, 95% CI: -0.86, -0.04), LDL levels (SMD = -0.45, 95% CI: -0.87, -0.02), TC levels (SMD = -1.09, 95% CI: -1.89, -0.29), and TNF-α levels (SMD = -1.73, 95% CI: -2.72, -0.74). CONCLUSION This network meta-analysis revealed that Lactobacillus + Bifidobacterium + Streptococcus may be the most effective probiotic combination for the treatment of liver enzymes, lipid profiles, and inflammation factors. These findings can be used to guide the development of a probiotics-based treatment guideline for NAFLD since there are few direct comparisons between different therapies.
Collapse
Affiliation(s)
- Yafang Yang
- Department of Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214000, China
| | - Ligang Yang
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, and Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing 210009, China
| | - Jiale Wu
- Department of Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214000, China
| | - Jing Hu
- Department of Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214000, China
| | - Min Wan
- Department of Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214000, China
| | - Jindi Bie
- Department of Clinical Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214000, China
| | - Jiaxin Li
- Department of Clinical Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214000, China
| | - Da Pan
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, and Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing 210009, China
| | - Guiju Sun
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, and Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing 210009, China
| | - Chao Yang
- Department of Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214000, China.
| |
Collapse
|
17
|
Yu L, Gao F, Li Y, Su D, Han L, Li Y, Zhang X, Feng Z. Role of pattern recognition receptors in the development of MASLD and potential therapeutic applications. Biomed Pharmacother 2024; 175:116724. [PMID: 38761424 DOI: 10.1016/j.biopha.2024.116724] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 04/23/2024] [Accepted: 05/06/2024] [Indexed: 05/20/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) has become one of the most prevalent liver diseases worldwide, and its occurrence is strongly associated with obesity, insulin resistance (IR), genetics, and metabolic stress. Ranging from simple fatty liver to metabolic dysfunction-associated steatohepatitis (MASH), even to severe complications such as liver fibrosis and advanced cirrhosis or hepatocellular carcinoma, the underlying mechanisms of MASLD progression are complex and involve multiple cellular mediators and related signaling pathways. Pattern recognition receptors (PRRs) from the innate immune system, including Toll-like receptors (TLRs), C-type lectin receptors (CLRs), NOD-like receptors (NLRs), RIG-like receptors (RLRs), and DNA receptors, have been demonstrated to potentially contribute to the pathogenesis for MASLD. Their signaling pathways can induce inflammation, mediate oxidative stress, and affect the gut microbiota balance, ultimately resulting in hepatic steatosis, inflammatory injury and fibrosis. Here we review the available literature regarding the involvement of PRR-associated signals in the pathogenic and clinical features of MASLD, in vitro and in animal models of MASLD. We also discuss the emerging targets from PRRs for drug developments that involved agent therapies intended to arrest or reverse disease progression, thus enabling the refinement of therapeutic targets that can accelerate drug development.
Collapse
Affiliation(s)
- Lili Yu
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China; The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang Medical University, Xinxiang, Henan, China; Institute of Precision Medicine, Xinxiang Medical University, Xinxiang, Henan, China; Xinxiang Engineering Technology Research Center of immune checkpoint drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan 453000, P.R.China
| | - Feifei Gao
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China; Institute of Precision Medicine, Xinxiang Medical University, Xinxiang, Henan, China; Xinxiang Engineering Technology Research Center of immune checkpoint drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan 453000, P.R.China
| | - Yaoxin Li
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China; Institute of Precision Medicine, Xinxiang Medical University, Xinxiang, Henan, China; Xinxiang Engineering Technology Research Center of immune checkpoint drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan 453000, P.R.China
| | - Dan Su
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China; Institute of Precision Medicine, Xinxiang Medical University, Xinxiang, Henan, China; Xinxiang Engineering Technology Research Center of immune checkpoint drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan 453000, P.R.China
| | - Liping Han
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang Medical University, Xinxiang, Henan, China
| | - Yueming Li
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China; Institute of Precision Medicine, Xinxiang Medical University, Xinxiang, Henan, China; Xinxiang Engineering Technology Research Center of immune checkpoint drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan 453000, P.R.China
| | - Xuehan Zhang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China; Institute of Precision Medicine, Xinxiang Medical University, Xinxiang, Henan, China; Xinxiang Engineering Technology Research Center of immune checkpoint drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan 453000, P.R.China
| | - Zhiwei Feng
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China; Institute of Precision Medicine, Xinxiang Medical University, Xinxiang, Henan, China; Xinxiang Engineering Technology Research Center of immune checkpoint drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan 453000, P.R.China.
| |
Collapse
|
18
|
Liu D, Chen P. Binary Bacillus subtilis protects the intestinal mucosa barrier and alleviates nonalcoholic steatohepatitis. Animal Model Exp Med 2024; 7:362-366. [PMID: 37469297 PMCID: PMC11228086 DOI: 10.1002/ame2.12337] [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: 03/19/2023] [Accepted: 06/09/2023] [Indexed: 07/21/2023] Open
Abstract
BACKGROUND Nonalcoholic steatohepatitis (NASH) is characterized by liver steatosis, inflammation, and even fibrosis. NASH is likely to develop into cirrhosis and liver cancer, the major causes of liver related deaths. We aimed to study the effect of probiotics on NASH via the gut-liver axis. METHODS Thirty male Sprague-Dawley rats were divided into three groups. A control group of 10 rats was fed on a standard chow for 16 weeks. Twenty rats fed on a high-fat diet for 8 weeks were separated to two groups: a model group (10 rats) fed on vehicle for 8 weeks and a treatment group (10 rats) supplemented with binary Bacillus subtilis for 8 weeks. Hepatic expression of IL-6 and TNF-ɑ and ileum expression of IL-17 and occludin were measured. RESULTS The high-fat diet caused inflammation of the liver and ileum in rats. Binary Bacillus subtilis treatment reduces liver inflammation through the intestinal liver axis. Increased levels of IL-6 and TNF-α were detected in rats fed a high-fat diet, which were reduced to lower levels after treatment with binary Bacillus subtilis. In rats on the high-fat diet, elevated IL-17 levels and decreased occludin levels were observed. Treatment with Bacillus subtilis reduced IL-17 levels and restored the expression of occludin. CONCLUSION Binary Bacillus subtilis has a beneficial effect on liver inflammation and intestinal damage.
Collapse
Affiliation(s)
- Donglin Liu
- Department of Gastroenterology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Pengguo Chen
- Department of Gastroenterology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| |
Collapse
|
19
|
Popov J, Despot T, Avelar Rodriguez D, Khan I, Mech E, Khan M, Bojadzija M, Pai N. Implications of Microbiota and Immune System in Development and Progression of Metabolic Dysfunction-Associated Steatotic Liver Disease. Nutrients 2024; 16:1668. [PMID: 38892602 PMCID: PMC11175128 DOI: 10.3390/nu16111668] [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: 05/10/2024] [Revised: 05/23/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most prevalent type of liver disease worldwide. The exact pathophysiology behind MASLD remains unclear; however, it is thought that a combination of factors or "hits" act as precipitants for disease onset and progression. Abundant evidence supports the roles of diet, genes, metabolic dysregulation, and the intestinal microbiome in influencing the accumulation of lipids in hepatocytes and subsequent progression to inflammation and fibrosis. Currently, there is no cure for MASLD, but lifestyle changes have been the prevailing cornerstones of management. Research is now focusing on the intestinal microbiome as a potential therapeutic target for MASLD, with the spotlight shifting to probiotics, antibiotics, and fecal microbiota transplantation. In this review, we provide an overview of how intestinal microbiota interact with the immune system to contribute to the pathogenesis of MASLD and metabolic dysfunction-associated steatohepatitis (MASH). We also summarize key microbial taxa implicated in the disease and discuss evidence supporting microbial-targeted therapies in its management.
Collapse
Affiliation(s)
- Jelena Popov
- Boston Combined Residency Program, Boston Children’s Hospital & Boston Medical Center, Boston, MA 02115, USA;
| | - Tijana Despot
- College of Medicine and Health, University College Cork, T12 YN60 Cork, Ireland; (T.D.); (I.K.)
| | - David Avelar Rodriguez
- Department of Pediatric Gastroenterology, Hepatology & Nutrition, The Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1E8, Canada;
| | - Irfan Khan
- College of Medicine and Health, University College Cork, T12 YN60 Cork, Ireland; (T.D.); (I.K.)
| | - Eugene Mech
- School of Medicine, University College Dublin, D04 C1P1 Dublin, Ireland;
| | - Mahrukh Khan
- Department of Pediatrics, Faculty of Health Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada;
- Department of Medical Sciences, Faculty of Health Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Milan Bojadzija
- Department of Internal Medicine, Subotica General Hospital, 24000 Subotica, Serbia;
| | - Nikhil Pai
- Department of Pediatrics, Faculty of Health Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada;
- Division of Gastroenterology, Hepatology and Nutrition, McMaster Children’s Hospital, Hamilton, ON L8S 4L8, Canada
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Division of Gastroenterology, Hepatology, and Nutrition, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| |
Collapse
|
20
|
Wu T, Zeng Z, Yu Y. Role of Probiotics in Gut Microbiome and Metabolome in Non-Alcoholic Fatty Liver Disease Mouse Model: A Comparative Study. Microorganisms 2024; 12:1020. [PMID: 38792849 PMCID: PMC11124503 DOI: 10.3390/microorganisms12051020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 05/10/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most prevalent chronic liver condition worldwide. Numerous studies conducted recently have demonstrated a connection between the dysbiosis of the development of NAFLD and gut microbiota. Rebuilding a healthy gut ecology has been proposed as a strategy involving the use of probiotics. The purpose of this work is to investigate and compare the function of probiotics Akkermansia muciniphila (A. muciniphila) and VSL#3 in NAFLD mice. Rodent NAFLD was modeled using a methionine choline-deficient diet (MCD) with/without oral probiotic delivery. Subsequently, qPCR, histological staining, and liver function tests were conducted. Mass spectrometry-based analysis and 16S rDNA gene sequencing were used to investigate the liver metabolome and gut microbiota. We found that while both A. muciniphila and VSL#3 reduced hepatic fat content, A. muciniphila outperformed VSL#3. Furthermore, probiotic treatment restored the β diversity of the gut flora and A. muciniphila decreased the abundance of pathogenic bacteria such as Ileibacterium valens. These probiotics altered the metabolism in MCD mice, especially the glycerophospholipid metabolism. In conclusion, our findings distinguished the role of A. muciniphila and VSL#3 in NAFLD and indicated that oral-gavage probiotics remodel gut microbiota and improve metabolism, raising the possibility of using probiotics in the cure of NAFLD.
Collapse
Affiliation(s)
| | - Zheng Zeng
- Department of Infectious Diseases, Peking University First Hospital, Beijing 100034, China;
| | - Yanyan Yu
- Department of Infectious Diseases, Peking University First Hospital, Beijing 100034, China;
| |
Collapse
|
21
|
Linero PL, Castilla-Guerra L. Management of Cardiovascular Risk in the Non-alcoholic Fatty Liver Disease Setting. Eur Cardiol 2024; 19:e02. [PMID: 38807854 PMCID: PMC11131151 DOI: 10.15420/ecr.2023.19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 10/02/2023] [Indexed: 05/30/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is an overlooked and undetected pathology, which affects more than 32% of adults worldwide. NAFLD is becoming more common in Western industrialised countries, particularly in patients with central obesity, type 2 diabetes, dyslipidaemia and metabolic syndrome. Although NAFLD has traditionally been interpreted as a liver disease with a high risk of liver-related complications, NAFLD is an underappreciated and independent risk factor for atherosclerotic cardiovascular disease, which is the principal cause of death in patients with NAFLD. Treatment options to counteract both the progression and development of cardiovascular disease and NAFLD include lifestyle interventions, such as weight loss, increased physical activity and dietary modification, and optimal medical therapy of comorbid conditions; nevertheless, further studies are needed to define optimal treatment strategies for the prevention of both hepatic and cardiovascular complications of NAFLD.
Collapse
Affiliation(s)
- Paula Luque Linero
- Vascular Risk Unit, Department of Internal Medicine, Hospital Virgen MacarenaSeville, Spain
| | - Luis Castilla-Guerra
- Vascular Risk Unit, Department of Internal Medicine, Hospital Virgen MacarenaSeville, Spain
- Department of Medicine, University of SevilleSeville, Spain
| |
Collapse
|
22
|
Meyer M, Schwärzler J, Jukic A, Tilg H. Innate Immunity and MASLD. Biomolecules 2024; 14:476. [PMID: 38672492 PMCID: PMC11048298 DOI: 10.3390/biom14040476] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 04/08/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) has emerged as the most common liver disease worldwide in recent years. MASLD commonly presents as simple hepatic steatosis, but ~25% of patients develop liver inflammation, progressive fibrosis, liver cirrhosis and related hepatocellular carcinoma. Liver inflammation and the degree of fibrosis are key determinants of the prognosis. The pathophysiology of liver inflammation is incompletely understood and involves diverse factors and specifically innate and adaptive immune responses. More specifically, diverse mediators of innate immunity such as proinflammatory cytokines, adipokines, inflammasomes and various cell types like mononuclear cells, macrophages and natural killer cells are involved in directing the inflammatory process in MASLD. The activation of innate immunity is driven by various factors including excess lipids and lipotoxicity, insulin resistance and molecular patterns derived from gut commensals. Targeting pathways of innate immunity might therefore appear as an attractive therapeutic strategy in the future management of MASLD and possibly its complications.
Collapse
Affiliation(s)
| | | | | | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University Innsbruck, 6020 Innsbruck, Austria; (M.M.); (A.J.)
| |
Collapse
|
23
|
Song Y, Guo W, Wang J, Liu S, Li Z, Li Y. Probiotic consumption and hepatic steatosis: results from the NHANES 2011-2016 and Mendelian randomization study. Front Nutr 2024; 11:1334935. [PMID: 38650641 PMCID: PMC11033389 DOI: 10.3389/fnut.2024.1334935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 03/27/2024] [Indexed: 04/25/2024] Open
Abstract
Background Recent research showed that probiotics treatment may reduce insulin resistance, regulate lipid metabolism, raise liver enzyme levels, and ameliorate inflammation in individuals with metabolic associated fatty liver disease (MAFLD). However, the possible effects of probiotic use on the progression of hepatic steatosis (HS) have not been identified. The purpose of this study was to investigate this in a large population database. Methods The cross-sectional research was conducted among adults with complete data on probiotic yogurt consumption and HS in the 2011-2016 National Health and Nutrition Examination Survey (NHANES). Probiotic yogurt consumption was assessed using a dietary supplement questionnaire, while HS was evaluated with HS index (HSI). To explore their relationship, weighted univariate regression analysis, subgroup analysis, and interaction analysis were conducted. To evaluate the causal association between yogurt consumption and NAFLD, mendelian randomization analysis (MR) were performed. A restricted cubic spline (RCS) was used to analyze the relationship curve between the leves of yogurt consumption and hepatic steatosis. Results A total of 7,891 participants were included in the study represented 146.7 million non-institutionalized residents of the United States, of whom 4,322 (54.77%) were diagnosed with HS. Multivariable logistic regression showed probiotic yogurt consumption had significantly inverse relationship for HS (OR = 0.84, 95% CI: 0.72-0.97, p = 0.02) after adjusting for all covariates. Once more, the independent relationship between probiotic yogurt consumption and HS was verified by subgroup analysis and interaction analysis. The MR analysis results indicate that there is no causal relationship between yogurt consumption and NAFLD. The RCS model demonstrated a robust J-shaped link between yogurt consumption and HS, revealing a significant decrease in risk within the lower range of yogurt consumption, which attained the lowest risk close to 0.4 cup. Conclusion According to the NHANES data, the consumption of probiotics and yogurt has a beneficial effect on HS, whereas the MR results indicated it was not related to NAFLD. The RCS analysis indicates a J-shaped relationship between yogurt consumption and HS, which may account for the inconsistency in the results. Based on these findings, we recommend that adults take half a cup of yogurt daily.
Collapse
Affiliation(s)
- Yancheng Song
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Wencong Guo
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
- Laboratory of Nephrology & Department of Nephrology, The Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao, China
| | - Junke Wang
- Department of Cardiology, Qingdao Hiser Hospital Affiliated to Qingdao University, Qingdao, China
| | - Shuguang Liu
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zhaopeng Li
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yu Li
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
24
|
Schwärzler J, Grabherr F, Grander C, Adolph TE, Tilg H. The pathophysiology of MASLD: an immunometabolic perspective. Expert Rev Clin Immunol 2024; 20:375-386. [PMID: 38149354 DOI: 10.1080/1744666x.2023.2294046] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 12/08/2023] [Indexed: 12/28/2023]
Abstract
INTRODUCTION Metabolic-associated liver diseases have emerged pandemically across the globe and are clinically related to metabolic disorders such as obesity and type 2 diabetes. The new nomenclature and definition (i.e. metabolic dysfunction-associated steatotic liver disease - MASLD; metabolic dysfunction-associated steatohepatitis - MASH) reflect the nature of these complex systemic disorders, which are characterized by inflammation, gut dysbiosis and metabolic dysregulation. In this review, we summarize recent advantages in understanding the pathophysiology of MASLD, which we parallel to emerging therapeutic concepts. AREAS COVERED We summarize the pathophysiologic concepts of MASLD and its transition to MASH and subsequent advanced sequelae of diseases. Furthermore, we highlight how dietary constituents, microbes and associated metabolites, metabolic perturbations, and immune dysregulation fuel lipotoxicity, hepatic inflammation, liver injury, insulin resistance, and systemic inflammation. Deciphering the intricate pathophysiologic processes that contribute to the development and progression of MASLD is essential to develop targeted therapeutic approaches to combat this escalating burden for health-care systems. EXPERT OPINION The rapidly increasing prevalence of metabolic dysfunction-associated steatotic liver disease challenges health-care systems worldwide. Understanding pathophysiologic traits is crucial to improve the prevention and treatment of this disorder and to slow progression into advanced sequelae such as cirrhosis and hepatocellular carcinoma.
Collapse
Affiliation(s)
- Julian Schwärzler
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Felix Grabherr
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Christoph Grander
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Timon E Adolph
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
25
|
Abenavoli L, Gambardella ML, Scarlata GGM, Lenci I, Baiocchi L, Luzza F. The Many Faces of Metabolic Dysfunction-Associated Fatty Liver Disease Treatment: From the Mediterranean Diet to Fecal Microbiota Transplantation. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:563. [PMID: 38674209 PMCID: PMC11051743 DOI: 10.3390/medicina60040563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 03/22/2024] [Accepted: 03/28/2024] [Indexed: 04/28/2024]
Abstract
The gastrointestinal tract is inhabited by the gut microbiota. The main phyla are Firmicutes and Bacteroidetes. In non-alcoholic fatty liver disease, now renamed metabolic dysfunction-associated fatty liver disease (MAFLD), an alteration in Firmicutes and Bacteroidetes abundance promotes its pathogenesis and evolution into non-alcoholic steatohepatitis, liver cirrhosis, and hepatocellular carcinoma. For this reason, early treatment is necessary to counteract its progression. The aim of the present narrative review is to evaluate the different therapeutic approaches to MAFLD. The most important treatment for MAFLD is lifestyle changes. In this regard, the Mediterranean diet could be considered the gold standard in the prevention and treatment of MAFLD. In contrast, a Western diet should be discouraged. Probiotics and fecal microbiota transplantation seem to be valid, safe, and effective alternatives for MAFLD treatment. However, more studies with a longer follow-up and with a larger cohort of patients are needed to underline the more effective approaches to contrasting MAFLD.
Collapse
Affiliation(s)
- Ludovico Abenavoli
- Department of Health Sciences, University “Magna Graecia”, Viale Europa, 88100 Catanzaro, Italy; (M.L.G.); (G.G.M.S.); (F.L.)
| | - Maria Luisa Gambardella
- Department of Health Sciences, University “Magna Graecia”, Viale Europa, 88100 Catanzaro, Italy; (M.L.G.); (G.G.M.S.); (F.L.)
| | - Giuseppe Guido Maria Scarlata
- Department of Health Sciences, University “Magna Graecia”, Viale Europa, 88100 Catanzaro, Italy; (M.L.G.); (G.G.M.S.); (F.L.)
| | - Ilaria Lenci
- Hepatology and Liver Transplant Unit, University of Tor Vergata, Via Montpellier, 00133 Rome, Italy; (I.L.); (L.B.)
| | - Leonardo Baiocchi
- Hepatology and Liver Transplant Unit, University of Tor Vergata, Via Montpellier, 00133 Rome, Italy; (I.L.); (L.B.)
| | - Francesco Luzza
- Department of Health Sciences, University “Magna Graecia”, Viale Europa, 88100 Catanzaro, Italy; (M.L.G.); (G.G.M.S.); (F.L.)
| |
Collapse
|
26
|
Shehata AI, Soliman AA, Ahmed HA, Gewaily MS, Amer AA, Shukry M, Abdel-Latif HMR. Evaluation of different probiotics on growth, body composition, antioxidant capacity, and histoarchitecture of Mugil capito. Sci Rep 2024; 14:7379. [PMID: 38548786 PMCID: PMC10978984 DOI: 10.1038/s41598-024-57489-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 03/19/2024] [Indexed: 04/01/2024] Open
Abstract
We investigated the dietary effects of the single application of Saccharomyces cerevisiae, Lactobacillus bulgaricus, and their combination on growth, proximate composition of whole fish body, antioxidant defense, and histoarchitecture of hapa-reared Mugil capito. Healthy fish (Fish weighed = 10.30 ± 0.10 g at first) were randomly allocated into 4 equal groups, each with three replicates. These groups were designed as follows: (1) a group fed a basal diet without probiotics (control), (2) a group fed a diet containing S. cerevisiae (4 g/kg diet), (3) a group fed a diet containing L. bulgaricus (2 g/kg diet), and (4) the last group fed a diet containing a combination of both, all for a duration of 60 days. Probiotic-treated groups showed significantly better growth and nutrition utilization than the control group. Significant differences were observed in the crude fat and crude protein contents among the groups, with the combination group exhibiting the highest levels. However, there were no significant variations in ash content across all groups. The highest hepatic antioxidant capacity (superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GPX) enzyme activities) was observed in the combination group. Thiobarbituric acid reactive substance (TBARS) concentrations were decreased significantly in all probiotic groups, suggesting improved oxidative stress resilience in these groups. The histomorphological analysis of the hepatopancreatic tissues revealed well-arranged parenchyma, increased glycogen storage, and melanomacrophage centers in probiotic-treated groups, particularly the combined probiotics group. Furthermore, the probiotic supplementation improved the histoarchitecture of the intestinal villi compared to the control group. To put it briefly, combined dietary administration of these probiotics improved growth, body composition, antioxidant defenses, and hepatic and intestinal health in hapa-reared M. capito, highlighting their promising role in promoting welfare and productivity.
Collapse
Affiliation(s)
- Akram Ismael Shehata
- Department of Animal and Fish Production, Faculty of Agriculture (Saba Basha), Alexandria University, Alexandria, 21531, Egypt.
| | - Ali A Soliman
- National Institute of Oceanography and Fisheries (NIOF), Alexandria, Egypt
| | - Hamada A Ahmed
- Department of Nutrition and Veterinary Clinical Nutrition, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511, Egypt
| | - Mahmoud S Gewaily
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh, 33516, Egypt
| | - Asem A Amer
- Department of Fish Nutrition and Feed Technology, Central Laboratory for Aquaculture Research, Agricultural Research Center, Abbassa, Abo-Hammad, Sharqia, 44662, Egypt
| | - Mustafa Shukry
- Department of Physiology, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh, 33516, Egypt
| | - Hany M R Abdel-Latif
- Department of Poultry and Fish Diseases, Faculty of Veterinary Medicine, Alexandria University, Alexandria, 22758, Egypt.
| |
Collapse
|
27
|
Zhao T, Lun S, Yan M, Park J, Wang S, Chen C. 6,7-Dimethoxycoumarin, Gardenoside and Rhein combination improves non-alcoholic fatty liver disease in rats. JOURNAL OF ETHNOPHARMACOLOGY 2024; 322:117646. [PMID: 38135236 DOI: 10.1016/j.jep.2023.117646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 12/14/2023] [Accepted: 12/19/2023] [Indexed: 12/24/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE This study explores the potential therapeutic benefits of using a three-component DGR (composed of specific compounds) to target the NLRP3 inflammasome in the context of non-alcoholic fatty liver disease (NAFLD). AIM OF THE STUDY To assess the impact of a three-component DGR on NAFLD, specifically examining its effects on liver lipid accumulation, inflammation, and the diversity of intestinal microbial communities. METHODS NAFLD was induced in 8-week-old Sprague Dawley rats by feeding them a high-fat emulsion diet every morning for 8 consecutive weeks. Oral administration of DGR or its constituent equivalents in the afternoon. The pharmacological effects of DGR were evaluated using H&E, ORO and ELISA methods to determine the changes in serum and liver tissue indexes of rat-models. Immunohistochemical staining and Western blot were used to assess the interaction between DGR, NLRP3 and IL-1β. RESULTS The induction of NAFLD resulted in elevated hepatic triglycerides (TG), total cholesterol (TC), and free fatty acids (FFA). However, these alterations were ameliorated upon administration of DGR. It is noteworthy that DGR exhibited superior efficacy in comparison to its constituent compounds, manifesting augmented antioxidant activity, diminished hepatic damage, and the attenuation of pro-inflammatory factors. Both DGR and its individual monomeric constituents exhibited the capacity to attenuate the activation of the NLRP3 inflammasome in the liver, leading to an amelioration of the pathological characteristics associated with NAFLD. An analysis of the intestinal flora unveiled an elevated abundance of p_Firmicutes (1.1-fold), p_Cyanobacteria (5.76-fold), and p_Verrucomicrobia (5.2-fold), accompanied by a heightened p_Firmicutes to p_Bacteroidetes ratio (5.49-fold). CONCLUSIONS In the non-alcoholic fatty liver disease (NAFLD) rat model, the concurrent administration of three-component DGR effectively regulated lipid deposition, suppressed liver inflammation, and restored balance in the intestinal flora, thereby improving NAFLD pathology. These findings propose a promising therapeutic strategy for NAFLD, centered on inhibiting the NLRP3 inflammasome through the use of the three-component DGR.
Collapse
Affiliation(s)
- Tianyi Zhao
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun, 130117, PR China
| | - Shiyi Lun
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun, 130117, PR China
| | - Maoying Yan
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun, 130117, PR China
| | - JongPil Park
- Department of Pharmaceutical Engineering, Daegu Haany University, Gyeongsan, 38610, Republic of Korea
| | - Shumin Wang
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun, 130117, PR China.
| | - Changbao Chen
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun, 130117, PR China.
| |
Collapse
|
28
|
Li X, Pan C, Ma W, Yang T, Wang C, Han W, Zhang W, Li H, Li Z, Zhao T, Guo XF, Li D. Effects of dietary supplementation of fish oil plus vitamin D 3 on gut microbiota and fecal metabolites, and their correlation with nonalcoholic fatty liver disease risk factors: a randomized controlled trial. Food Funct 2024; 15:2616-2627. [PMID: 38356413 DOI: 10.1039/d3fo02319b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2024]
Abstract
We previously reported that fish oil plus vitamin D3 (FO + D) could ameliorate nonalcoholic fatty liver disease (NAFLD). However, it is unclear whether the beneficial effects of FO + D on NAFLD are associated with gut microbiota and fecal metabolites. In this study, we investigated the effects of dietary supplementation of FO + D on gut microbiota and fecal metabolites and their correlation with NAFLD risk factors. Methods: A total of 61 subjects were randomly divided into three groups: FO + D group (2.34 g day-1 of eicosatetraenoic acid (EPA) + docosahexaenoic acid (DHA) + 1680 IU vitamin D3), FO group (2.34 g day-1 of EPA + DHA), and corn oil (CO) group (1.70 g d-1 linoleic acid). Blood and fecal samples were collected at the baseline and day 90. Gut microbiota were analyzed through 16S rRNA PCR analysis, and fecal co-metabolites were determined via untargeted ultraperformance liquid chromatography-quadrupole time-of-flight mass spectrometry (UPLC-Q-TOF-MS). Results: The relative abundance of Eubacterium (p = 0.03) and Lactobacillus (p = 0.05) increased, whereas that of Streptococcus (p = 0.02) and Dialister (p = 0.04) decreased in the FO + D group compared with the CO group. Besides, changes in tetracosahexaenoic acid (THA, C24:6 n-3) (p = 0.03) levels were significantly enhanced, whereas 8,9-DiHETrE levels (p < 0.05) were reduced in the FO + D group compared with the CO group. The changes in 1,25-dihydroxyvitamin D3 levels in the fecal samples were inversely associated with insulin resistance, which was determined using the homeostatic model assessment model (HOMA-IR, r = -0.29, p = 0.02), and changes in 8,9-DiHETrE levels were positively associated with adiponectin levels (r = -0.43, p < 0.05). Conclusion: The present results indicate that the beneficial effects of FO + D on NAFLD may be partially attributed to the impact on gut microbiota and fecal metabolites.
Collapse
Affiliation(s)
- Xueqi Li
- Institute of Nutrition & Health, Qingdao University, Qingdao, China.
- School of Public Health, Qingdao University, Qingdao, China
- Binzhou Center for Disease Control and Prevention, Binzhou, China
| | - Chi Pan
- Institute of Nutrition & Health, Qingdao University, Qingdao, China.
- School of Public Health, Qingdao University, Qingdao, China
| | - Wenjun Ma
- Institute of Nutrition & Health, Qingdao University, Qingdao, China.
- School of Public Health, Qingdao University, Qingdao, China
| | - Ting Yang
- Institute of Nutrition & Health, Qingdao University, Qingdao, China.
- School of Public Health, Qingdao University, Qingdao, China
| | - Chong Wang
- Institute of Nutrition & Health, Qingdao University, Qingdao, China.
- School of Public Health, Qingdao University, Qingdao, China
| | - Weiwei Han
- Institute of Nutrition & Health, Qingdao University, Qingdao, China.
- School of Public Health, Qingdao University, Qingdao, China
| | - Wei Zhang
- Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hui Li
- Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zhongxia Li
- Byhealth Institute of Nutrition & Health, Guangzhou, China
| | - Ting Zhao
- Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiao-Fei Guo
- Institute of Nutrition & Health, Qingdao University, Qingdao, China.
- School of Public Health, Qingdao University, Qingdao, China
| | - Duo Li
- Institute of Nutrition & Health, Qingdao University, Qingdao, China.
- School of Public Health, Qingdao University, Qingdao, China
| |
Collapse
|
29
|
Fan C, He Y, Yang J, Da M. Association Between Live Microbe Intake and NAFLD: Evidence From NHANES 2003-2018. JOURNAL OF THE AMERICAN NUTRITION ASSOCIATION 2024; 43:272-278. [PMID: 37930261 DOI: 10.1080/27697061.2023.2270537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 10/10/2023] [Indexed: 11/07/2023]
Abstract
OBJECTIVE This study aimed to shed light on the potential relationship between live microbe intake and nonalcoholic fatty liver disease (NAFLD). METHOD By using a cross-sectional study design, the researchers were able to investigate the possible causal association between the two variables in a rigorous and systematic manner. RESULTS Our study investigated the correlation between the intake of live microbe-containing foods and NAFLD in a representative sample of adults. The study found that the intake of live microbe-containing foods was associated with lower blood pressure, plasma glucose, NAFLD, body mass index, glycated hemoglobin, alanine aminotransferase, aspartate aminotransferase, gamma-glutamyl transpeptidase, and low-density lipoprotein cholesterol, as well as higher high-density lipoprotein cholesterol levels (p < 0.05). In univariate logistic regression, high dietary live microbe intake was associated with lower NAFLD prevalence than low intake (OR = 0.830; 95% CI, 0.759 to 0.908; p < 0.001). After adjusting for multiple variables, the same conclusion was supported (p < 0.05). In subgroup analyses, there was a significant difference in the race and smoking groups, with p for interaction of 0.01 and 0.02, respectively. This study's findings serve to augment the existing body of evidence linking live microbes with favorable health outcomes. CONCLUSIONS Our study revealed a robust correlation between dietary intake of live microbes and the prevalence of NAFLD in a cross-sectional analysis. Our findings offer a novel perspective on NAFLD research, highlighting the potential of targeted modulation of specific bacterial taxa, including the promotion of beneficial bacteria and suppression of harmful ones, as a promising strategy for preventing and treating NAFLD.
Collapse
Affiliation(s)
- Chuanlei Fan
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Yang He
- The First Clinical Medical College, Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu, China
| | - Jian Yang
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Mingxu Da
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
- Department of Surgical Oncology, Gansu Provincial Hospital, Lanzhou, China
| |
Collapse
|
30
|
Hsieh RH, Chien YJ, Lan WY, Lin YK, Lin YH, Chiang CF, Yang MT. Bacillus coagulans TCI711 Supplementation Improved Nonalcoholic Fatty Liver by Modulating Gut Microbiota: A Randomized, Placebo-Controlled, Clinical Trial. Curr Dev Nutr 2024; 8:102083. [PMID: 38510931 PMCID: PMC10951533 DOI: 10.1016/j.cdnut.2024.102083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 12/21/2023] [Accepted: 01/10/2024] [Indexed: 03/22/2024] Open
Abstract
Background Nonalcoholic fatty liver disease (NAFLD) has become one of the major problems of chronic liver disease worldwide. It not only causes damage to the liver but also engenders chronic hepatitis and cirrhosis. Recent studies have shown that regulating Bacillus coagulans can improve NAFLD. Objectives This trial explores whether B. coagulans TCI711 (BCT) could ameliorate NAFLD. Methods A total of 57 patients with NAFLD were recruited through FibroScan liver fibrosis scanner and divided into placebo (n = 28) and BCT-supplemented groups (n = 29). Specifically, 1 BCT probiotic capsule was supplemented daily for 8 wk. Furthermore, the blood, stool, and fatty liver content were then examined. Results Parameters evaluated for liver and kidney indicators showed no side effects after supplementing BCT. A significant reduction of 8.7% in the fatty liver was achieved by effectively suppressing the grade of fatty liver as revealed by controlled attenuation parameter. BCT also regulated gut microbiota profiles, with significant increases observed in Bifidobacterium, Eubacterium, Ruminococcaceae, and Sellimonas compared with the baseline. Conclusions BCT may improve NAFLD by regulating gut microbiota, and parameters evaluated for liver and kidney indicate no side effects.
Collapse
Affiliation(s)
- Rong-Hong Hsieh
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei, Taiwan
| | - Yu-Ju Chien
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei, Taiwan
| | - Wen-Yi Lan
- Center for General Education, Taipei Medical University, Taipei, Taiwan
| | - Yung-Kai Lin
- Institute of Food Safety and Risk Management, National Taiwan Ocean University, Keelung, Taiwan
- Department of Food Science, National Taiwan Ocean University, Keelung, Taiwan
- Graduate Institute of Biomedical Engineering, National Chung Hsing University, Taichung, Taiwan
| | | | - Chi-Fu Chiang
- Research & Design Center, TCI Co., Ltd., Taipei, Taiwan
| | - Ming-Ta Yang
- Center for General Education, Taipei Medical University, Taipei, Taiwan
- Clinical Research Center, Taipei Medical University Hospital, Taipei, Taiwan
| |
Collapse
|
31
|
Portincasa P, Khalil M, Graziani A, Frühbeck G, Baffy G, Garruti G, Di Ciaula A, Bonfrate L. Gut microbes in metabolic disturbances. Promising role for therapeutic manipulations? Eur J Intern Med 2024; 119:13-30. [PMID: 37802720 DOI: 10.1016/j.ejim.2023.10.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 08/30/2023] [Accepted: 10/02/2023] [Indexed: 10/08/2023]
Abstract
The prevalence of overweight, obesity, type 2 diabetes, metabolic syndrome and steatotic liver disease is rapidly increasing worldwide with a huge economic burden in terms of morbidity and mortality. Several genetic and environmental factors are involved in the onset and development of metabolic disorders and related complications. A critical role also exists for the gut microbiota, a complex polymicrobial ecology at the interface of the internal and external environment. The gut microbiota contributes to food digestion and transformation, caloric intake, and immune response of the host, keeping the homeostatic control in health. Mechanisms of disease include enhanced energy extraction from the non-digestible dietary carbohydrates, increased gut permeability and translocation of bacterial metabolites which activate a chronic low-grade systemic inflammation and insulin resistance, as precursors of tangible metabolic disorders involving glucose and lipid homeostasis. The ultimate causative role of gut microbiota in this respect remains to be elucidated, as well as the therapeutic value of manipulating the gut microbiota by diet, pre- and pro- synbiotics, or fecal microbial transplantation.
Collapse
Affiliation(s)
- Piero Portincasa
- Clinica Medica "A. Murri", Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari "Aldo Moro" Medical School, Policlinico Hospital, Piazza G. Cesare 11, Bari 70124, Italy.
| | - Mohamad Khalil
- Clinica Medica "A. Murri", Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari "Aldo Moro" Medical School, Policlinico Hospital, Piazza G. Cesare 11, Bari 70124, Italy
| | - Annarita Graziani
- Institut AllergoSan Pharmazeutische Produkte Forschungs- und Vertriebs GmbH, Graz, Austria
| | - Gema Frühbeck
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), ISCIII, Pamplona, Spain; Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain; Department of Endocrinology & Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
| | - Gyorgy Baffy
- Department of Medicine, VA Boston Healthcare System and Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02130, USA
| | - Gabriella Garruti
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari Medical School, Bari 70124, Italy
| | - Agostino Di Ciaula
- Clinica Medica "A. Murri", Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari "Aldo Moro" Medical School, Policlinico Hospital, Piazza G. Cesare 11, Bari 70124, Italy.
| | - Leonilde Bonfrate
- Clinica Medica "A. Murri", Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari "Aldo Moro" Medical School, Policlinico Hospital, Piazza G. Cesare 11, Bari 70124, Italy
| |
Collapse
|
32
|
Xiao J, Xiang H, Xiang H, Sun Z, Xu J, Ren H, Hu P, Peng M. GW9662 ameliorates nonalcoholic steatohepatitis by inhibiting the PPARγ/CD36 pathway and altering the gut microbiota. Eur J Pharmacol 2023; 960:176113. [PMID: 37838102 DOI: 10.1016/j.ejphar.2023.176113] [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: 08/27/2023] [Revised: 10/04/2023] [Accepted: 10/11/2023] [Indexed: 10/16/2023]
Abstract
BACKGROUND & AIMS Peroxisome proliferator-activated receptors (PPARs) are currently among the most focused-on therapeutic targets for non-alcoholic steatohepatitis (NASH), although no clinical transformation has been achieved to date. In this study, we aimed to evaluate the effects of GW9662 on choline-deficient, L-amino acid-defined high-fat diet (CDAA-HFD)-induced NASH mice and reveal the mechanism underlying this effect. METHODS GW9662 (1 mg/kg) was administered in CDAA-HFD mouse model of NASH. The effect of GW9662 on hepatic lipid metabolism was investigated using liver RNA-seq and HepG2 cells induced by oleic acid and palmitic acid. In addition, 16S rRNA gene sequencing was performed to analyze the effects of GW9662 on the composition and function of the fecal microbiota. RESULTS GW9662 improved the CDAA-HFD caused elevation in the levels of ALT, AST, hepatic free fatty acids and triglycerides. The liver pathological analysis indicated that GW9662 alleviated the hepatic steatosis and fibrosis. The NAFLD activity score and RNA-Seq revealed that GW9662 mainly regulated the fatty acids transport and lipid synthesis by inhibiting PPARγ, CD36, FABP1, FASN, and SCD1, and through the up-regulation of PPARα. Moreover, GW9662 reduced the epididymal fat weight. GW9662 reversed the gut microbiota disorder by increasing the abundance of the beneficial bacteria Dubosiella and Lactobacillus and decreasing the abundance of harmful bacteria Lachnospiraceae_NK4A136_group, Helicobacteraceae, Desulfovibriaceae, and Rickenaceae. CONCLUSIONS GW9662 ameliorated lipid metabolism by inhibiting the PPARγ/CD36 pathway and altering the composition of the gut microbiota in NASH mice. Therefore, the PPARγ antagonist GW9662 deserves more attention as a potential therapeutic agent for NASH.
Collapse
Affiliation(s)
- Jing Xiao
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Huanyu Xiang
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Hongyan Xiang
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Zilin Sun
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Jing Xu
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Hong Ren
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Peng Hu
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China.
| | - Mingli Peng
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
33
|
Zakaria Z, Othman ZA, Nna VU, Mohamed M. The promising roles of medicinal plants and bioactive compounds on hepatic lipid metabolism in the treatment of non-alcoholic fatty liver disease in animal models: molecular targets. Arch Physiol Biochem 2023; 129:1262-1278. [PMID: 34153200 DOI: 10.1080/13813455.2021.1939387] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 06/01/2021] [Indexed: 12/13/2022]
Abstract
Imbalance in hepatic lipid metabolism can lead to an abnormal triglycerides deposition in the hepatocytes which can cause non-alcoholic fatty liver disease (NAFLD). Four main mechanisms responsible for regulating hepatic lipid metabolism are fatty acid uptake, de novo lipogenesis, lipolysis and fatty acid oxidation. Controlling the expression of transcription factors at molecular level plays a crucial role in NAFLD management. This paper reviews various medicinal plants and their bioactive compounds emphasising mechanisms involved in hepatic lipid metabolism, other important NAFLD pathological features, and their promising roles in managing NAFLD through regulating key transcription factors. Although there are many medicinal plants popularly investigated for NAFLD treatment, there is still little information and scientific evidence available and there has been no research on clinical trials scrutinised on this matter. This review also aims to provide molecular information of medicinal plants in NALFD treatment that might have potentials for future scientifically controlled studies.
Collapse
Affiliation(s)
- Zaida Zakaria
- Department of Physiology, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, Kelantan, Malaysia
| | - Zaidatul Akmal Othman
- Department of Physiology, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, Kelantan, Malaysia
- Unit of Physiology, Faculty of Medicine, Universiti Sultan Zainal Abidin, Kuala Terengganu, Malaysia
| | - Victor Udo Nna
- Department of Physiology, Faculty of Basic Medical Sciences, College of Medical Sciences, University of Calabar, Calabar, Nigeria
| | - Mahaneem Mohamed
- Department of Physiology, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, Kelantan, Malaysia
- Unit of Integrative Medicine, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, Kelantan, Malaysia
| |
Collapse
|
34
|
Tilg H, Adolph TE, Tacke F. Therapeutic modulation of the liver immune microenvironment. Hepatology 2023; 78:1581-1601. [PMID: 37057876 DOI: 10.1097/hep.0000000000000386] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 03/14/2023] [Indexed: 04/15/2023]
Abstract
Inflammation is a hallmark of progressive liver diseases such as chronic viral or immune-mediated hepatitis, alcohol-associated liver disease, and NAFLD. Preclinical and clinical studies have provided robust evidence that cytokines and related cellular stress sensors in innate and adaptive immunity orchestrate hepatic disease processes. Unresolved inflammation and liver injury result in hepatic scarring, fibrosis, and cirrhosis, which may culminate in HCC. Liver diseases are accompanied by gut dysbiosis and a bloom of pathobionts, fueling hepatic inflammation. Anti-inflammatory strategies are extensively used to treat human immune-mediated conditions beyond the liver, while evidence for immunomodulatory therapies and cell therapy-based strategies in liver diseases is only emerging. The development and establishment of novel immunomodulatory therapies for chronic liver diseases has been dampened by several clinical challenges, such as invasive monitoring of therapeutic efficacy with liver biopsy in clinical trials and risk of DILI in several studies. Such aspects prevented advancements of novel medical therapies for chronic inflammatory liver diseases. New concepts modulating the liver immune environment are studied and eagerly awaited to improve the management of chronic liver diseases in the future.
Collapse
Affiliation(s)
- Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, & Metabolism, Medical University Innsbruck, Innsbruck, Austria
| | - Timon E Adolph
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, & Metabolism, Medical University Innsbruck, Innsbruck, Austria
| | - Frank Tacke
- Department of Hepatology & Gastroenterology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany
| |
Collapse
|
35
|
Zhu J, Song Y, Xiao Y, Ma L, Hu C, Yang H, Wang X, Lyu W. Metagenomic reconstructions of caecal microbiome in Landes, Roman and Zhedong White geese. Br Poult Sci 2023; 64:565-576. [PMID: 37493577 DOI: 10.1080/00071668.2023.2239172] [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: 01/11/2023] [Revised: 06/10/2023] [Accepted: 06/16/2023] [Indexed: 07/27/2023]
Abstract
1. The caecal microbiota in geese play a crucial role in determining the host's health, disease status and behaviour, as evidenced by extensive epidemiological data. The present investigation conducted 10× metagenomic sequencing of caecal content samples obtained from three distinct goose species, namely Landes geese, Roman geese and Zhedong White geese (n = 5), to explore the contribution of the gut microbiome to carbohydrate metabolism.2. In total, 337GB of Illumina data were generated, which identified 1,048,575 complete genes and construction of 331 metagenomic bins, encompassing 78 species from nine phyla. Firmicutes, Bacteroidetes, Actinobacteria, Proteobacteria and Bacteria were identified as the dominant phyla while Prevotella, Bacteroides, Streptococcus, and Subdoligranulum were the most abundant genera in the caecum of geese.3. The genes were allocated to 375 pathways using the Kyoto Encyclopedia of Genes and Genome (KEGG) analysis. The most abundant classes in the caecum of geese were confirmed to be glycoside hydrolases (GHs), glycosyl transferases (GTs), as identified through the carbohydrate-active enzyme (CAZyme) database mapping. Subdoligranulum variabile and Mediterraneibacter glycyrrhizinilyticus were discovered to potentially facilitate carbohydrate digestion in geese.4. Notwithstanding, further investigation and validation are required to establish a connection between these species and CAZymes. Based on binning analysis, Mediterraneibacter glycyrrhizinilyticus and Ruminococcus sp. CAG:177 are potential species in LD geese that contribute to the production of fatty liver.
Collapse
Affiliation(s)
- J Zhu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Agro-Product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
- College of Animal Science, Zhejiang University, Hangzhou, China
- Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, China
| | - Y Song
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Agro-Product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Y Xiao
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Agro-Product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - L Ma
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Agro-Product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - C Hu
- College of Animal Science, Zhejiang University, Hangzhou, China
- Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, China
| | - H Yang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Agro-Product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - X Wang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Agro-Product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - W Lyu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Agro-Product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| |
Collapse
|
36
|
Tilg H, Byrne CD, Targher G. NASH drug treatment development: challenges and lessons. Lancet Gastroenterol Hepatol 2023; 8:943-954. [PMID: 37597527 DOI: 10.1016/s2468-1253(23)00159-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/19/2023] [Accepted: 05/19/2023] [Indexed: 08/21/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) has become the most common chronic liver disease worldwide. Although NAFLD is tightly linked to obesity and type 2 diabetes, this liver disease also affects individuals who do not have obesity. NAFLD increases the risk of developing cardiovascular disease, chronic kidney disease, and certain extrahepatic cancers. There is currently no licensed pharmacotherapy for NAFLD, despite numerous clinical trials in the past two decades. Currently, the reason so few drugs have been successful in the treatment of NAFLD in a trial setting is not fully understood. As cardiovascular disease is the predominant cause of mortality in people with NAFLD, future pharmacotherapies for NAFLD must consider associated cardiometabolic risk factors. The successful use of glucose-lowering drugs in the treatment of type 2 diabetes in patients with NAFLD indicates that this strategy is important, and worth developing further. Greater public awareness of NAFLD is needed because collaboration between all stakeholders is vital to enable a holistic approach to successful treatment.
Collapse
Affiliation(s)
- Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University Innsbruck, Innsbruck, Austria.
| | - Christopher D Byrne
- National Institute for Health and Care Research, Southampton Biomedical Research Centre, University Hospital Southampton and University of Southampton, Southampton, UK
| | - Giovanni Targher
- Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Verona, Verona, Italy; IRCCS Ospedale Sacro Cuore Don Calabria, Negrar di Valpolicella, Italy
| |
Collapse
|
37
|
Burger K, Jung F, Baumann A, Brandt A, Staltner R, Sánchez V, Bergheim I. TNFα is a key trigger of inflammation in diet-induced non-obese MASLD in mice. Redox Biol 2023; 66:102870. [PMID: 37683301 PMCID: PMC10493600 DOI: 10.1016/j.redox.2023.102870] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 08/28/2023] [Accepted: 08/31/2023] [Indexed: 09/10/2023] Open
Abstract
Tumor necrosis factor alpha (TNFα) is thought to be a critical factor in the development of metabolic dysfunction-associated steatotic liver disease (MASLD). Here, we determined the effects of a treatment with the anti-TNFα antibody infliximab and a genetic deletion of TNFα, respectively, in the development of non-obese diet-induced early metabolic dysfunction-associated steatohepatitis (MASH) in mice. The treatment with infliximab improved markers of liver damage in mice with pre-existing early MASH. In TNFα-/- mice, the development of early signs of MASH and insulin resistance was significantly attenuated compared to wild-type animals. While mRNA expression of proinflammatory cytokines like interleukin 1β (Il1b) and interleukin 6 (Il6) were significantly lower in livers of MASH-diet-fed TNFα-/- mice compared to wild-type mice with early MASH, markers of intestinal barrier function were similarly impaired in both MASH-diet-fed groups compared to controls. Our data suggest that TNFα is a key regulator of hepatic inflammation and insulin resistance associated with the development of early non-obese MASH.
Collapse
Affiliation(s)
- Katharina Burger
- Department of Nutritional Sciences, Molecular Nutritional Science, University of Vienna, Vienna, Austria
| | - Finn Jung
- Department of Nutritional Sciences, Molecular Nutritional Science, University of Vienna, Vienna, Austria
| | - Anja Baumann
- Department of Nutritional Sciences, Molecular Nutritional Science, University of Vienna, Vienna, Austria
| | - Annette Brandt
- Department of Nutritional Sciences, Molecular Nutritional Science, University of Vienna, Vienna, Austria
| | - Raphaela Staltner
- Department of Nutritional Sciences, Molecular Nutritional Science, University of Vienna, Vienna, Austria
| | - Victor Sánchez
- Department of Nutritional Sciences, Molecular Nutritional Science, University of Vienna, Vienna, Austria
| | - Ina Bergheim
- Department of Nutritional Sciences, Molecular Nutritional Science, University of Vienna, Vienna, Austria.
| |
Collapse
|
38
|
Moreira ALG, Silva GA, Silva PHF, Salvador SL, Vicente RM, Ferreira GC, Tanus-Santos JE, Mayer MPA, Ishikawa KH, de Souza SLS, Furlaneto FAC, Messora MR. Bifidobacterium animalis subspecies lactis HN019 can reduce the sequelae of experimental periodontitis in rats modulating intestinal parameters, expression of lipogenic genes, and levels of hepatic steatosis. J Periodontal Res 2023; 58:1006-1019. [PMID: 37482954 DOI: 10.1111/jre.13163] [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: 12/09/2022] [Revised: 05/15/2023] [Accepted: 07/07/2023] [Indexed: 07/25/2023]
Abstract
OBJECTIVE To determine whether Bifidobacterium animalis subspecies lactis HN019 (B. lactis HN019) can reduce the sequelae of experimental periodontitis (EP) in rats modulating systemic parameters. BACKGROUND This study evaluated the effects of probiotic therapy (PROB) in the prevention of local and systemic damage resulting from EP. METHODS Forty-eight rats were allocated into four groups: C (control), PROB, EP, and EP-PROB. PROB (1 × 1010 CFU/mL) administration lasted 8 weeks and PE was induced on the 7th week by placing ligature on the animals' lower first molars. All animals were euthanized in the 9th week of the experiment. Biomolecular analyses, RT-PCR, and histomorphometric analyses were performed. The data obtained were analyzed statistically (ANOVA, Tukey, p < .05). RESULTS The EP group had higher dyslipidemia when compared to the C group, as well as higher levels of insulin resistance, proteinuria levels, percentages of systolic blood pressure, percentage of fatty hepatocytes in the liver, and expression of adipokines was up-regulated (LEPR, NAMPT, and FABP4). All these parameters (except insulin resistance, systolic blood pressure, LEPR and FABP4 gene expression) were reduced in the EP-PROB group when compared to the EP group. The EP group had lower villus height and crypt depth, as well as a greater reduction in Bacteroidetes and a greater increase in Firmicutes when compared to the EP-PROB group. Greater alveolar bone loss was observed in the EP group when compared to the EP-PROB group. CONCLUSION Bifidobacterium lactis HN019 can reduce the sequelae of EP in rats modulating intestinal parameters, attenuating expression of lipogenic genes and hepatic steatosis.
Collapse
Affiliation(s)
- André L G Moreira
- Department of Oral and Maxillofacial Surgery and Periodontology, School of Dentistry of Ribeirão Preto, University of São Paulo - USP, Ribeirão Preto, São Paulo, Brazil
| | - Giselle A Silva
- Department of Oral and Maxillofacial Surgery and Periodontology, School of Dentistry of Ribeirão Preto, University of São Paulo - USP, Ribeirão Preto, São Paulo, Brazil
| | - Pedro H F Silva
- Department of Oral and Maxillofacial Surgery and Periodontology, School of Dentistry of Ribeirão Preto, University of São Paulo - USP, Ribeirão Preto, São Paulo, Brazil
| | - Sérgio L Salvador
- Department of Clinical Analyses, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo - USP, Ribeirão Preto, São Paulo, Brazil
| | - Raphael M Vicente
- Department of Oral and Maxillofacial Surgery and Periodontology, School of Dentistry of Ribeirão Preto, University of São Paulo - USP, Ribeirão Preto, São Paulo, Brazil
| | - Graziele C Ferreira
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo - USP, Ribeirão Preto, São Paulo, Brazil
| | - Jose E Tanus-Santos
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo - USP, Ribeirão Preto, São Paulo, Brazil
| | - Marcia P A Mayer
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Karin H Ishikawa
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Sérgio Luís Scombatti de Souza
- Department of Oral and Maxillofacial Surgery and Periodontology, School of Dentistry of Ribeirão Preto, University of São Paulo - USP, Ribeirão Preto, São Paulo, Brazil
| | - Flávia A C Furlaneto
- Department of Oral and Maxillofacial Surgery and Periodontology, School of Dentistry of Ribeirão Preto, University of São Paulo - USP, Ribeirão Preto, São Paulo, Brazil
| | - Michel R Messora
- Department of Oral and Maxillofacial Surgery and Periodontology, School of Dentistry of Ribeirão Preto, University of São Paulo - USP, Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
39
|
Hayashi H, Sawada K, Tanaka H, Muro K, Hasebe T, Nakajima S, Okumura T, Fujiya M. The effect of heat-killed Lactobacillus brevis SBL88 on improving selective hepatic insulin resistance in non-alcoholic fatty liver disease mice without altering the gut microbiota. J Gastroenterol Hepatol 2023; 38:1847-1854. [PMID: 37646384 DOI: 10.1111/jgh.16337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 08/07/2023] [Accepted: 08/08/2023] [Indexed: 09/01/2023]
Abstract
BACKGROUND AND AIM There have been several reports that some probiotics improve non-alcoholic fatty liver disease (NAFLD); however, many studies have involved cocktail therapies. We evaluated whether heat-killed Lactobacillus brevis SBL88 (L. brevis SBL88) monotherapy improves the clinical features of NAFLD. METHODS The NAFLD model was induced in mice fed a high-fat diet (HFD) (HFD mice) or HFD + 1% heat-killed L. brevis SBL88 (SBL mice) for 16 weeks. Histopathological liver findings were analyzed. To evaluate the gut microbiota, a modified terminal restriction fragment length polymorphism analysis of the feces was performed. RNA sequencing in the liver was performed with Ion Proton™. To investigate the direct effects of heat-killed L. brevis SBL88, an in vitro study was performed. RESULTS Histopathological findings revealed that fat droplets in the liver were significantly reduced in SBL mice; however, terminal restriction fragment length polymorphism did not show alterations in the gut microbiota between HFD mice and SBL mice. RNA sequencing and pathway analysis revealed that the regulation of lipid and insulin metabolism was affected. The mRNA expression of insulin receptor substrate 2 (IRS-2) was significantly higher in SBL mice, whereas the expression of IRS-1 was not significantly different. Phospho-IRS-2 expression was also significantly increased in SBL mice. In addition, an in vitro study revealed significant alterations in IRS-2 and forkhead box protein O1 expression levels. CONCLUSION SBL mice exhibited partially improved selective hepatic insulin resistance. Our data suggest that heat-killed L. brevis SBL88 could attenuate the clinical features of NAFLD that are not mediated by alterations in the gut microbiota.
Collapse
Affiliation(s)
- Hidemi Hayashi
- Division of Metabolism and Biosystemic Science, Gastroenterology, and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Koji Sawada
- Division of Metabolism and Biosystemic Science, Gastroenterology, and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Hiroki Tanaka
- Division of Tumor Pathology, Department of Pathology, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Kazuki Muro
- Division of Metabolism and Biosystemic Science, Gastroenterology, and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Takumu Hasebe
- Division of Metabolism and Biosystemic Science, Gastroenterology, and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Shunsuke Nakajima
- Division of Metabolism and Biosystemic Science, Gastroenterology, and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Toshikatsu Okumura
- Division of Metabolism and Biosystemic Science, Gastroenterology, and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Mikihiro Fujiya
- Gastroenterology and Endoscopy, Division of Metabolism and Biosystemic Science, Gastroenterology, and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| |
Collapse
|
40
|
Liu L, Deng L, Wei W, Li C, Lu Y, Bai J, Li L, Zhang H, Jin N, Li C, Zhao C. Lactiplantibacillus plantarum LPJZ-658 Improves Non-Alcoholic Steatohepatitis by Modulating Bile Acid Metabolism and Gut Microbiota in Mice. Int J Mol Sci 2023; 24:13997. [PMID: 37762300 PMCID: PMC10531215 DOI: 10.3390/ijms241813997] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/05/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
Non-alcoholic steatohepatitis (NASH) is one of the most prevalent diseases worldwide; it is characterized by hepatic lipid accumulation, inflammation, and progressive fibrosis. Here, a Western diet combined with low-dose weekly carbon tetrachloride was fed to C57BL/6J mice for 12 weeks to build a NASH model to investigate the attenuating effects and possible mechanisms of Lactiplantibacillus plantarum LPJZ-658. Hepatic pathology, lipid profiles, and gene expression were assessed. The metabolomic profiling of the serum was performed. The composition structure of gut microbiota was profiled using 16s rRNA sequencing. The results show that LPJZ-658 treatment significantly attenuated liver injury, steatosis, fibrosis, and inflammation in NASH mice. Metabolic pathway analysis revealed that several pathways, such as purine metabolism, glycerophospholipid metabolism, linoleic acid metabolism, and primary bile acid biosynthesis, were associated with NASH. Notably, we found that treatment with LPJZ-658 regulated the levels of bile acids (BAs) in the serum. Moreover, LPJZ-658 restored NASH-induced gut microbiota dysbiosis. The correlation analysis deduced obvious interactions between BAs and gut microbiota. The current study indicates that LPJZ-658 supplementation protects against NASH progression, which is accompanied by alternating BA metabolic and modulating gut microbiota.
Collapse
Affiliation(s)
- Liming Liu
- College of Animal Science and Technology, Jilin Agricultural Science and Technology University, Jilin 132101, China; (L.L.); (C.L.); (Y.L.)
| | - Liquan Deng
- School of Public Health, Jilin University, Changchun 130021, China;
| | - Wei Wei
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China;
| | - Chunhua Li
- College of Animal Science and Technology, Jilin Agricultural Science and Technology University, Jilin 132101, China; (L.L.); (C.L.); (Y.L.)
| | - Yuting Lu
- College of Animal Science and Technology, Jilin Agricultural Science and Technology University, Jilin 132101, China; (L.L.); (C.L.); (Y.L.)
| | - Jieying Bai
- College of Future Technology, Peking University, Beijing 100871, China;
| | - Letian Li
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China; (L.L.); (N.J.)
| | - Heping Zhang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot 010018, China;
| | - Ningyi Jin
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China; (L.L.); (N.J.)
| | - Chang Li
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China; (L.L.); (N.J.)
| | - Cuiqing Zhao
- College of Animal Science and Technology, Jilin Agricultural Science and Technology University, Jilin 132101, China; (L.L.); (C.L.); (Y.L.)
| |
Collapse
|
41
|
Vachliotis ID, Polyzos SA. The Role of Tumor Necrosis Factor-Alpha in the Pathogenesis and Treatment of Nonalcoholic Fatty Liver Disease. Curr Obes Rep 2023; 12:191-206. [PMID: 37407724 PMCID: PMC10482776 DOI: 10.1007/s13679-023-00519-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/20/2023] [Indexed: 07/07/2023]
Abstract
PURPOSE OF REVIEW To summarize experimental and clinical evidence on the association between tumor necrosis factor-α (TNF-α) and nonalcoholic fatty liver disease (NAFLD) and discuss potential treatment considerations. RECENT FINDINGS Experimental evidence suggests that TNF-α is a cytokine with a critical role in the pathogenesis of NAFLD. Although, the production of TNF-α may be an early event during the course of nonalcoholic fatty liver (NAFL), TNF-α may play a more substantial role in the pathogenesis of nonalcoholic steatohepatitis (NASH) and NAFLD-associated fibrosis. Moreover, TNF-α may potentiate hepatic insulin resistance, thus interconnecting inflammatory with metabolic signals and possibly contributing to the development of NAFLD-related comorbidities, including cardiovascular disease, hepatocellular carcinoma, and extra-hepatic malignancies. In clinical terms, TNF-α is probably associated with the severity of NAFLD; circulating TNF-α gradually increases from controls to patients with NAFL, and then, to patients with NASH. Given this potential association, various therapeutic interventions (obeticholic acid, peroxisome proliferator-activated receptors, sodium-glucose co-transporter 2 inhibitors, glucagon-like peptide-1 receptor agonists, probiotics, synbiotics, rifaximin, vitamin E, pentoxifylline, ursodeoxycholic acid, fibroblast growth factor-21, n-3 polyunsaturated fatty acids, statins, angiotensin receptor blockers) have been evaluated for their effect on TNF-α and NAFLD. Interestingly, anti-TNF biologics have shown favorable metabolic and hepatic effects, which may open a possible therapeutic window for the management of advanced NAFLD. The potential key pathogenic role of TNF-α in NAFLD warrants further investigation and may have important diagnostic and therapeutic implications.
Collapse
Affiliation(s)
- Ilias D. Vachliotis
- First Department of Pharmacology, Medical School, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
- Second Department of Internal Medicine, 424 General Military Hospital, Thessaloniki, Greece
| | - Stergios A. Polyzos
- First Department of Pharmacology, Medical School, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| |
Collapse
|
42
|
Zhao Y, Li C, Luan Z, Chen J, Wang C, Jing Y, Qi S, Zhao Z, Zhang H, Wu J, Chen Y, Li Z, Zhao B, Wang S, Yang Y, Sun G. Lactobacillus oris improves non-alcoholic fatty liver in mice and inhibits endogenous cholesterol biosynthesis. Sci Rep 2023; 13:12946. [PMID: 37558739 PMCID: PMC10412569 DOI: 10.1038/s41598-023-38530-x] [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: 09/01/2022] [Accepted: 07/10/2023] [Indexed: 08/11/2023] Open
Abstract
We previously confirmed that a strain of Lactobacillus oris isolated from the fecal samples of healthy Hainan centenarian having potent lipid-lowering ability in HepG2 cells; and this study was to investigate the effect of the stain on non-alcoholic fatty liver in mice in vivio. The Lactobacillus oris strain isolated from Hainan centenarian fecal samples were frozen stored in our laboratory. Thirty ob/ob mice (10 in each group) were orally gavaged with Lactobacillus oris (Lactobacillus, 5 × 109 cfu), mixed probiotics (Mixed, 5 × 109 cfu, a mixture with known lipid-lowering ability), or culture medium (Control) respectively. Lactobacillus oris isolated from fecal samples of Hainan centenarians showed significantly in vivo lipid lowering ability compared with the controls, and the ability was comparable with mixed probiotics strains in mice The possible mechanisms of lipid-lowering of probiotics and Lactobacillus oris may be associated with HMGR inhibition to suppress the synthesis of endogenous cholesterol; bile acids reabsorption, and intestinal FXR-FGF15 signaling pathways promoting the cholesterol conversion into bile acids secretion.
Collapse
Affiliation(s)
- Yiming Zhao
- Department of Gastroenterology and Hepatology, Hainan Hospital of PLA General Hospital, Sanya, 572013, China
| | - Congyong Li
- Sixth Health Care Department, Second Medical Center of PLA General Hospital, Beijing, 100853, China
| | - Zhe Luan
- Department of Gastroenterology and Hepatology, First Medical Center of PLA General Hospital, Beijing, 100853, China
| | - Jun Chen
- Unit 91917, Beijing, 102401, China
| | - Cong Wang
- Emergency Department, Tianjin First Central Hospital, Tianjin, 300192, China
| | - Yujia Jing
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100193, China
| | - Shirui Qi
- Emergency Department, Tianjin First Central Hospital, Tianjin, 300192, China
| | - Zhizhuang Zhao
- Department of Gastroenterology and Hepatology, Hainan Hospital of PLA General Hospital, Sanya, 572013, China
| | - Hanwen Zhang
- Department of Gastroenterology and Hepatology, First Medical Center of PLA General Hospital, Beijing, 100853, China
| | - Junling Wu
- Department of Gastroenterology and Hepatology, First Medical Center of PLA General Hospital, Beijing, 100853, China
| | - Yi Chen
- Department of Gastroenterology and Hepatology, First Medical Center of PLA General Hospital, Beijing, 100853, China
| | - Zhuanyu Li
- Beijing QuantiHealth Technology Co., Ltd., Beijing, 100070, China
| | - Bowen Zhao
- Beijing QuantiHealth Technology Co., Ltd., Beijing, 100070, China
| | - Shufang Wang
- Department of Gastroenterology and Hepatology, First Medical Center of PLA General Hospital, Beijing, 100853, China.
| | - Yunsheng Yang
- Department of Gastroenterology and Hepatology, First Medical Center of PLA General Hospital, Beijing, 100853, China.
| | - Gang Sun
- Department of Gastroenterology and Hepatology, First Medical Center of PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
43
|
Grander C, Grabherr F, Tilg H. Non-alcoholic fatty liver disease: pathophysiological concepts and treatment options. Cardiovasc Res 2023; 119:1787-1798. [PMID: 37364164 PMCID: PMC10405569 DOI: 10.1093/cvr/cvad095] [Citation(s) in RCA: 64] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/30/2022] [Accepted: 06/23/2023] [Indexed: 06/28/2023] Open
Abstract
The prevalence of non-alcoholic fatty liver disease (NAFLD) is continually increasing due to the global obesity epidemic. NAFLD comprises a systemic metabolic disease accompanied frequently by insulin resistance and hepatic and systemic inflammation. Whereas simple hepatic steatosis is the most common disease manifestation, a more progressive disease course characterized by liver fibrosis and inflammation (i.e. non-alcoholic steatohepatitis) is present in 10-20% of affected individuals. NAFLD furthermore progresses in a substantial number of patients towards liver cirrhosis and hepatocellular carcinoma. Whereas this disease now affects almost 25% of the world's population and is mainly observed in obesity and type 2 diabetes, NAFLD also affects lean individuals. Pathophysiology involves lipotoxicity, hepatic immune disturbances accompanied by hepatic insulin resistance, a gut dysbiosis, and commonly hepatic and systemic insulin resistance defining this disorder a prototypic systemic metabolic disorder. Not surprisingly many affected patients have other disease manifestations, and indeed cardiovascular disease, chronic kidney disease, and extrahepatic malignancies are all contributing substantially to patient outcome. Weight loss and lifestyle change reflect the cornerstone of treatment, and several medical treatment options are currently under investigation. The most promising treatment strategies include glucagon-like peptide 1 receptor antagonists, sodium-glucose transporter 2 inhibitors, Fibroblast Growth Factor analogues, Farnesoid X receptor agonists, and peroxisome proliferator-activated receptor agonists. Here, we review epidemiology, pathophysiology, and therapeutic options for NAFLD.
Collapse
Affiliation(s)
- Christoph Grander
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University Innsbruck, Anichstrasse 35, Innsbruck 6020, Austria
| | - Felix Grabherr
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University Innsbruck, Anichstrasse 35, Innsbruck 6020, Austria
| | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University Innsbruck, Anichstrasse 35, Innsbruck 6020, Austria
| |
Collapse
|
44
|
Yang X, Li D, Zhang M, Feng Y, Jin X, Liu D, Guo Y, Hu Y. Ginkgo biloba extract alleviates fatty liver hemorrhagic syndrome in laying hens via reshaping gut microbiota. J Anim Sci Biotechnol 2023; 14:97. [PMID: 37533076 PMCID: PMC10399048 DOI: 10.1186/s40104-023-00900-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 05/31/2023] [Indexed: 08/04/2023] Open
Abstract
BACKGROUND Ginkgo biloba extract (GBE) is evidenced to be effective in the prevention and alleviation of metabolic disorders, including obesity, diabetes and fatty liver disease. However, the role of GBE in alleviating fatty liver hemorrhagic syndrome (FLHS) in laying hens and the underlying mechanisms remain to be elucidated. Here, we investigated the effects of GBE on relieving FLHS with an emphasis on the modulatory role of GBE in chicken gut microbiota. RESULTS The results showed that GBE treatment ameliorated biochemical blood indicators in high-fat diet (HFD)-induced FLHS laying hen model by decreasing the levels of TG, TC, ALT and ALP. The lipid accumulation and pathological score of liver were also relieved after GBE treatment. Moreover, GBE treatment enhanced the antioxidant activity of liver and serum by increasing GSH, SOD, T-AOC, GSH-PX and reducing MDA, and downregulated the expression of genes related to lipid synthesis (FAS, LXRα, GPAT1, PPARγ and ChREBP1) and inflammatory cytokines (TNF-α, IL-6, TLR4 and NF-κB) in the liver. Microbial profiling analysis revealed that GBE treatment reshaped the HFD-perturbed gut microbiota, particularly elevated the abundance of Megasphaera in the cecum. Meanwhile, targeted metabolomic analysis of SCFAs revealed that GBE treatment significantly promoted the production of total SCFAs, acetate and propionate, which were positively correlated with the GBE-enriched gut microbiota. Finally, we confirmed that the GBE-altered gut microbiota was sufficient to alleviate FLHS by fecal microbiota transplantation (FMT). CONCLUSIONS We provided evidence that GBE alleviated FLHS in HFD-induced laying hens through reshaping the composition of gut microbiota. Our findings shed light on mechanism underlying the anti-FLHS efficacy of GBE and lay foundations for future use of GBE as additive to prevent and control FLHS in laying hen industry.
Collapse
Affiliation(s)
- Xinyue Yang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Depeng Li
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Meihong Zhang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Yuqing Feng
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Xiaolu Jin
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Dan Liu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Yuming Guo
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Yongfei Hu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
45
|
Avelar-Rodríguez D, Peña-Vélez R, Popov J, Hill L, Ryan PM. Probiotics and non-alcoholic fatty liver disease in children and adolescents: a systematic review. REVISTA ESPANOLA DE ENFERMEDADES DIGESTIVAS 2023; 115:418-427. [PMID: 36412490 DOI: 10.17235/reed.2022.8796/2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/05/2023]
Abstract
BACKGROUND non-alcoholic fatty liver disease (NAFLD) in childhood is an increasing global public health issue with significant long-term consequences. NAFLD management mainly consists of lifestyle modifications, however, adjunct pharmacological therapies are currently lacking. Gut microbiota manipulation via probiotics may alter the course of pediatric NAFLD. The objective of this systematic review was to synthesize all the available literature on the use of probiotics in children and adolescents with NAFLD. METHODS PubMed, EBSCOhost, Scopus, Web of Science, and Cochrane Library were systematically searched for trials on the use of probiotics in pediatric NAFLD. A quantitative DerSimonian Laird random effects meta-analysis was performed when possible; otherwise, a narrative summary of the study outcomes was presented and discussed. A separate search was completed to include all the ongoing registered trials on probiotics use in pediatric NAFLD. RESULTS five randomized controlled trials met the inclusion criteria. Of these, four trials were included in the final quantitative analysis. Probiotic therapy significantly reduced the levels of alanine aminotransferase (ALT) (mean difference: -10.39 [-19.85, -0.93]), however significant heterogeneity between studies was identified (I2, 93 %). CONCLUSIONS there is insufficient evidence to support probiotics in the treatment of pediatric NAFLD given the substantial degree of discordance amongst the available trials. Lifestyle modifications focusing on maintaining a normal BMI and regular exercise continue to be the gold standard approach to treating NAFLD in children.
Collapse
Affiliation(s)
| | - Rubén Peña-Vélez
- Gastroenterología y Nutrición, Hospital General de Puebla "Dr. Eduardo Vázquez N", México
| | - Jelena Popov
- College of Medicine and Health. University College Cork
| | | | | |
Collapse
|
46
|
El-Baz AM, El-Ganiny AM, Hellal D, Anwer HM, El-Aziz HAA, Tharwat IE, El-Adawy MA, Helal SEDM, Mohamed MTA, Azb TM, Elshafaey HM, Shalata AA, Elmeligi SM, Abdelbary NH, El-Kott AF, Al-Saeed FA, Salem ET, El-Sokkary MMA, Shata A, Shabaan AA. Valuable effects of lactobacillus and citicoline on steatohepatitis: role of Nrf2/HO-1 and gut microbiota. AMB Express 2023; 13:57. [PMID: 37291355 DOI: 10.1186/s13568-023-01561-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 05/16/2023] [Indexed: 06/10/2023] Open
Abstract
Non-alcoholic steatohepatitis (NASH) is a more dangerous form of chronic non-alcoholic fatty liver disease (NAFLD). In the current investigation, the influence of citicoline on high-fat diet (HFD)-induced NASH was examined, both alone and in combination with Lactobacillus (probiotic). NASH was induced by feeding HFD (10% sugar, 10% lard stearin, 2% cholesterol, and 0.5% cholic acid) to rats for 13 weeks and received single i.p. injection of streptozotocin (STZ, 30 mg/kg) after 4 weeks. Citicoline was given at two dose levels (250 mg and 500 mg, i.p.) at the beginning of the sixth week, and in combination with an oral suspension of Lactobacillus every day for eight weeks until the study's conclusion. HFD/STZ induced steatohepatitis as shown by histopathological changes, elevated serum liver enzymes, serum hyperlipidemia and hepatic fat accumulation. Moreover, HFD convinced oxidative stress by increased lipid peroxidation marker (MDA) and decreased antioxidant enzymes (GSH and TAC). Upregulation of TLR4/NF-kB and the downstream inflammatory cascade (TNF-α, and IL-6) as well as Pentaraxin, fetuin-B and apoptotic markers (caspase-3 and Bax) were observed. NASH rats also had massive increase in Bacteroides spp., Fusobacterium spp., E. coli, Clostridium spp., Providencia spp., Prevotella interrmedia, and P. gingivalis while remarkable drop in Bifidobacteria spp. and Lactobacillus spp. Co-treatment with citicoline alone and with Lactobacillus improve histopathological NASH outcomes and reversed all of these molecular pathological alterations linked to NASH via upregulating the expression of Nrf2/HO-1 and downregulating TLR4/NF-kB signaling pathways. These results suggest that citicoline and lactobacillus may represent new hepatoprotective strategies against NASH progression.
Collapse
Affiliation(s)
- Ahmed M El-Baz
- Department of Microbiology and Biotechnology, Faculty of Pharmacy, Delta University for Science and Technology, 11152, Gamasa, Egypt.
- Department of Microbiology and Biotechnology, Faculty of Pharmacy, Delta University for Science and Technology, International Coastal Road, Gamasa City, Mansoura, Dakahlia, P.O. Box +11152, Egypt.
| | - Amira M El-Ganiny
- Department of Microbiology and Immunology, Faculty of Pharmacy, Zagazig University, 44519, Zagazig, Egypt
| | - Doaa Hellal
- Department of Clinical Pharmacology, Faculty of Medicine, Mansoura University, 35516, Mansoura, Egypt
| | - Hala M Anwer
- Department of Physiology, Faculty of Medicine, Benha University, Benha, Egypt
| | - Hend A Abd El-Aziz
- Department of Clinical Pharmacy, Faculty of Pharmacy, Delta University for Science and Technology, 11152, Gamasa, Egypt
| | - Ibrahim E Tharwat
- Department of Clinical Pharmacy, Faculty of Pharmacy, Delta University for Science and Technology, 11152, Gamasa, Egypt
| | - Mohamed A El-Adawy
- Department of Clinical Pharmacy, Faculty of Pharmacy, Delta University for Science and Technology, 11152, Gamasa, Egypt
| | - Shehab El-Din M Helal
- Department of Clinical Pharmacy, Faculty of Pharmacy, Delta University for Science and Technology, 11152, Gamasa, Egypt
| | - Menna Tallah A Mohamed
- Department of Clinical Pharmacy, Faculty of Pharmacy, Delta University for Science and Technology, 11152, Gamasa, Egypt
| | - Tassnim M Azb
- Department of Clinical Pharmacy, Faculty of Pharmacy, Delta University for Science and Technology, 11152, Gamasa, Egypt
| | - Hanya M Elshafaey
- Department of Clinical Pharmacy, Faculty of Pharmacy, Delta University for Science and Technology, 11152, Gamasa, Egypt
| | - AbdulRahman A Shalata
- Department of Clinical Pharmacy, Faculty of Pharmacy, Delta University for Science and Technology, 11152, Gamasa, Egypt
| | - Sahar M Elmeligi
- Department of Clinical Pharmacy, Faculty of Pharmacy, Delta University for Science and Technology, 11152, Gamasa, Egypt
| | - Noran H Abdelbary
- Department of Clinical Pharmacy, Faculty of Pharmacy, Delta University for Science and Technology, 11152, Gamasa, Egypt
| | - Attalla F El-Kott
- Department of Biology, College of Science, King Khalid University, 61421, Abha, Saudi Arabia
- Department of Zoology, College of Science, Damanhour University, 22511, Damanhour, Egypt
| | - Fatimah A Al-Saeed
- Department of Biology, College of Science, King Khalid University, 61421, Abha, Saudi Arabia
| | - Eman T Salem
- Department of Basic Science, Faculty of Physical Therapy, Horus University-Egypt, 34518, Horus, New Damietta, Egypt
| | | | - Ahmed Shata
- Department of Clinical Pharmacology, Faculty of Medicine, Mansoura University, 35516, Mansoura, Egypt
- Department of Clinical Pharmacy, Faculty of Pharmacy, Delta University for Science and Technology, 11152, Gamasa, Egypt
| | - Ahmed A Shabaan
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, 11152, Gamasa, Egypt
- Department of Pharmacology and Toxicology, Faculty of pharmacy, Mansoura University, 35516, Mansoura, Egypt
| |
Collapse
|
47
|
El-Baz AM, El-Ganiny AM, Hellal D, Anwer HM, El-Aziz HAA, Tharwat IE, El-Adawy MA, Helal SEDM, Mohamed MTA, Azb TM, Elshafaey HM, Shalata AA, Elmeligi SM, Abdelbary NH, El-kott AF, Al-Saeed FA, Salem ET, El-Sokkary MMA, Shata A, Shabaan AA. Valuable effects of lactobacillus and citicoline on steatohepatitis: role of Nrf2/HO-1 and gut microbiota. AMB Express 2023; 13:57. [DOI: https:/doi.org/10.1186/s13568-023-01561-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 05/16/2023] [Indexed: 05/14/2025] Open
Abstract
AbstractNon-alcoholic steatohepatitis (NASH) is a more dangerous form of chronic non-alcoholic fatty liver disease (NAFLD). In the current investigation, the influence of citicoline on high-fat diet (HFD)-induced NASH was examined, both alone and in combination with Lactobacillus (probiotic). NASH was induced by feeding HFD (10% sugar, 10% lard stearin, 2% cholesterol, and 0.5% cholic acid) to rats for 13 weeks and received single i.p. injection of streptozotocin (STZ, 30 mg/kg) after 4 weeks. Citicoline was given at two dose levels (250 mg and 500 mg, i.p.) at the beginning of the sixth week, and in combination with an oral suspension of Lactobacillus every day for eight weeks until the study’s conclusion. HFD/STZ induced steatohepatitis as shown by histopathological changes, elevated serum liver enzymes, serum hyperlipidemia and hepatic fat accumulation. Moreover, HFD convinced oxidative stress by increased lipid peroxidation marker (MDA) and decreased antioxidant enzymes (GSH and TAC). Upregulation of TLR4/NF-kB and the downstream inflammatory cascade (TNF-α, and IL-6) as well as Pentaraxin, fetuin-B and apoptotic markers (caspase-3 and Bax) were observed. NASH rats also had massive increase in Bacteroides spp., Fusobacterium spp., E. coli, Clostridium spp., Providencia spp., Prevotella interrmedia, and P. gingivalis while remarkable drop in Bifidobacteria spp. and Lactobacillus spp. Co-treatment with citicoline alone and with Lactobacillus improve histopathological NASH outcomes and reversed all of these molecular pathological alterations linked to NASH via upregulating the expression of Nrf2/HO-1 and downregulating TLR4/NF-kB signaling pathways. These results suggest that citicoline and lactobacillus may represent new hepatoprotective strategies against NASH progression.
Collapse
|
48
|
Adori M, Bhat S, Gramignoli R, Valladolid-Acebes I, Bengtsson T, Uhlèn M, Adori C. Hepatic Innervations and Nonalcoholic Fatty Liver Disease. Semin Liver Dis 2023; 43:149-162. [PMID: 37156523 PMCID: PMC10348844 DOI: 10.1055/s-0043-57237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disorder. Increased sympathetic (noradrenergic) nerve tone has a complex role in the etiopathomechanism of NAFLD, affecting the development/progression of steatosis, inflammation, fibrosis, and liver hemodynamical alterations. Also, lipid sensing by vagal afferent fibers is an important player in the development of hepatic steatosis. Moreover, disorganization and progressive degeneration of liver sympathetic nerves were recently described in human and experimental NAFLD. These structural alterations likely come along with impaired liver sympathetic nerve functionality and lack of adequate hepatic noradrenergic signaling. Here, we first overview the anatomy and physiology of liver nerves. Then, we discuss the nerve impairments in NAFLD and their pathophysiological consequences in hepatic metabolism, inflammation, fibrosis, and hemodynamics. We conclude that further studies considering the spatial-temporal dynamics of structural and functional changes in the hepatic nervous system may lead to more targeted pharmacotherapeutic advances in NAFLD.
Collapse
Affiliation(s)
- Monika Adori
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Sadam Bhat
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Roberto Gramignoli
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Ismael Valladolid-Acebes
- Department of Molecular Medicine and Surgery, The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Stockholm, Sweden
| | - Tore Bengtsson
- Department of Molecular Biosciences, The Wenner-Gren Institute (MBW), Stockholm University, Stockholm, Sweden
| | - Mathias Uhlèn
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Science for Life Laboratory, Royal Institute of Technology, Stockholm, Sweden
| | - Csaba Adori
- Department of Molecular Biosciences, The Wenner-Gren Institute (MBW), Stockholm University, Stockholm, Sweden
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
49
|
Pezzino S, Sofia M, Mazzone C, Castorina S, Puleo S, Barchitta M, Agodi A, Gallo L, La Greca G, Latteri S. Gut Microbiome in the Progression of NAFLD, NASH and Cirrhosis, and Its Connection with Biotics: A Bibliometric Study Using Dimensions Scientific Research Database. BIOLOGY 2023; 12:biology12050662. [PMID: 37237476 DOI: 10.3390/biology12050662] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/30/2023] [Accepted: 04/19/2023] [Indexed: 05/28/2023]
Abstract
There is growing evidence that gut microbiota dysbiosis is linked to the etiopathogenesis of nonalcoholic fatty liver disease (NAFLD), from the initial stage of disease until the progressive stage of nonalcoholic steatohepatitis (NASH) and the final stage of cirrhosis. Conversely, probiotics, prebiotics, and synbiotics have shown promise in restoring dysbiosis and lowering clinical indicators of disease in a number of both preclinical and clinical studies. Additionally, postbiotics and parabiotics have recently garnered some attention. The purpose of this bibliometric analysis is to assess recent publishing trends concerning the role of the gut microbiome in the progression of NAFLD, NASH and cirrhosis and its connection with biotics. The free access version of the Dimensions scientific research database was used to find publications in this field from 2002 to 2022. VOSviewer and Dimensions' integrated tools were used to analyze current research trends. Research into the following topics is expected to emerge in this field: (1) evaluation of risk factors which are correlated with the progression of NAFLD, such as obesity and metabolic syndrome; (2) pathogenic mechanisms, such as liver inflammation through toll-like receptors activation, or alteration of short-chain fatty acids metabolisms, which contribute to NAFLD development and its progression in more severe forms, such as cirrhosis; (3) therapy for cirrhosis through dysbiosis reduction, and research on hepatic encephalopathy a common consequence of cirrhosis; (4) evaluation of diversity, and composition of gut microbiome under NAFLD, and as it varies under NASH and cirrhosis by rRNA gene sequencing, a tool which can also be used for the development of new probiotics and explore into the impact of biotics on the gut microbiome; (5) treatments to reduce dysbiosis with new probiotics, such as Akkermansia, or with fecal microbiome transplantation.
Collapse
Affiliation(s)
- Salvatore Pezzino
- Department of Surgical Sciences and Advanced Technologies "G. F. Ingrassia", Cannizzaro Hospital, University of Catania, 95123 Catania, Italy
| | - Maria Sofia
- Department of Surgical Sciences and Advanced Technologies "G. F. Ingrassia", Cannizzaro Hospital, University of Catania, 95123 Catania, Italy
| | - Chiara Mazzone
- Department of Surgical Sciences and Advanced Technologies "G. F. Ingrassia", Cannizzaro Hospital, University of Catania, 95123 Catania, Italy
| | - Sergio Castorina
- Department of Surgical Sciences and Advanced Technologies "G. F. Ingrassia", Cannizzaro Hospital, University of Catania, 95123 Catania, Italy
| | - Stefano Puleo
- Department of Surgical Sciences and Advanced Technologies "G. F. Ingrassia", Cannizzaro Hospital, University of Catania, 95123 Catania, Italy
| | - Martina Barchitta
- Department of Surgical Sciences and Advanced Technologies "G. F. Ingrassia", Cannizzaro Hospital, University of Catania, 95123 Catania, Italy
| | - Antonella Agodi
- Department of Surgical Sciences and Advanced Technologies "G. F. Ingrassia", Cannizzaro Hospital, University of Catania, 95123 Catania, Italy
| | - Luisa Gallo
- Department of Surgical Sciences and Advanced Technologies "G. F. Ingrassia", Cannizzaro Hospital, University of Catania, 95123 Catania, Italy
| | - Gaetano La Greca
- Department of Surgical Sciences and Advanced Technologies "G. F. Ingrassia", Cannizzaro Hospital, University of Catania, 95123 Catania, Italy
| | - Saverio Latteri
- Department of Surgical Sciences and Advanced Technologies "G. F. Ingrassia", Cannizzaro Hospital, University of Catania, 95123 Catania, Italy
| |
Collapse
|
50
|
St-Amant A, Bergdahl A. A systematic review and meta-analysis of randomized controlled trials investigating the effects of probiotics on oxidative stress in healthy adults. Clin Nutr ESPEN 2023; 54:180-186. [PMID: 36963861 DOI: 10.1016/j.clnesp.2023.01.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 11/06/2022] [Accepted: 01/16/2023] [Indexed: 01/22/2023]
Abstract
BACKGROUND The oxidative stress (OS) theory of disease stipulates that a chronic imbalance in the ratio of oxidants to antioxidants in the cellular environment leads to a variety of debilitating conditions, including type 2 diabetes, cardiovascular and liver diseases. Metabolites in the gut microbiome have been associated with increases in reactive oxygen species (ROS). Many randomized controlled trials (RCTs) have thus investigated the potential of probiotics as a nutraceutical intervention to improve parameters of OS. AIM The objective of this paper is to review relevant human RCTs exploring the potential of probiotic supplementation to prevent OS in metabolically healthy individuals. METHODS This systematic review and meta-analysis was registered on PROSPERO (CRD42021297210). The PubMed database was searched using keywords related to probiotics and OS. In total, out of the 652 studies were screened, 9 respected the inclusion criteria. RESULTS Total antioxidant capacity (TAC) (SMD: 0.83 mmol/L, 95% CI: 0.25-1.40, p = 0.005) and glutathione (GSH) (SMD: 0.45, 95% CI: 0.13-0.77, p = 0.006) are improved with probiotic ingestion, although there are no alterations in superoxide dismutase (SOD) (SMD: 0.33, 95% CI: -0.27-0.93, p = 0.28). Decreases in plasma concentrations of the OS biomarker malondialdehyde (MDA) (SMD: -0.55, 95% CI: -1.11-0.00, p = 0.05) are also detected. CONCLUSION Probiotics improve AS and OS in metabolically healthy individuals. However, more studies are needed to address the moderate to high degree of heterogeneity in methodology.
Collapse
Affiliation(s)
- Antoine St-Amant
- Concordia University, Montreal, QC, Canada, H4B 1R6, Department of Health, Kinesiology and Applied Physiology.
| | - Andreas Bergdahl
- Concordia University, Montreal, QC, Canada, H4B 1R6, Department of Health, Kinesiology and Applied Physiology.
| |
Collapse
|