1
|
Wang Y, Feng S, Huang Y, Peng R, Liang L, Wang W, Guo M, Zhu B, Zhang H, Liao J, Zhou J, Li H, Li X, Ning Y, Wu F, Wu K. Revealing multiple biological subtypes of schizophrenia through a data-driven approach. J Transl Med 2025; 23:505. [PMID: 40316994 PMCID: PMC12048963 DOI: 10.1186/s12967-025-06503-5] [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: 01/13/2025] [Accepted: 04/12/2025] [Indexed: 05/04/2025] Open
Abstract
INTRODUCTION The brain imaging subtypes of schizophrenia have been widely investigated using data-driven approaches. However, the heterogeneity of SZ in multiple biological data is largely unknown. METHODS A data-driven model was used to classify brain imaging, gut microbiota, and brain-gut fusion data obtained through a dot product fusion method, identifying significant subtypes and calculating their correlations with clinical symptoms and cognitive performance. RESULTS These subtypes remain relatively independent and demonstrate typical features and biomarkers, which are significantly associated with clinical symptoms and cognitive performance. Two brain subtypes with opposite structural and functional changes are identified: (1) a structural variant-dominant brain subtype with negative symptoms and cognitive deficits and (2) a functional alteration-dominant brain subtype with positive symptoms. The three gut subtypes include the following: (1) Collinsella-dominant; (2) Prevotella-dominant with positive symptoms; and (3) Streptococcus-dominant. Two brain-gut subtypes show different abnormalities in brain‒genus linkages: (1) strong connectivity of "brain function in the temporal and parietal lobes-Prevotella" with reduced attention scores and (2) strong connectivity of "brain structure and function in the frontal and parietal lobes-multiple genera" with positive symptoms. Notably, brain subtypes and brain-gut subtypes are most relevant to clinical symptoms, whereas gut subtypes reveal more cognitive biomarkers. CONCLUSION These findings show the potential to identify multiple biological subtypes with distinct biomarkers, thereby suggesting the possibility of personalized and precise treatment for SZ patients.
Collapse
Affiliation(s)
- Yuran Wang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, China
| | - Shixuan Feng
- Department of Psychiatry, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, 510370, China
| | - Yuanyuan Huang
- Department of Psychiatry, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, 510370, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, 510370, China
| | - Runlin Peng
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, China
| | - Liqin Liang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, China
| | - Wei Wang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, China
| | - Minxin Guo
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, China
| | - Baoyuan Zhu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, China
| | - Heng Zhang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, China
| | - Jianhao Liao
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, China
| | - Jing Zhou
- School of Material Science and Engineering, South China University of Technology, Guangzhou, 510006, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, 510370, China
- Guangdong Engineering Technology Research Center for Diagnosis and Rehabilitation of Dementia, Guangzhou, 510500, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China
| | - Hehua Li
- Department of Psychiatry, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, 510370, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, 510370, China
| | - Xiaobo Li
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Yuping Ning
- Department of Psychiatry, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, 510370, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, 510370, China
| | - Fengchun Wu
- Department of Psychiatry, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, 510370, China.
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, 510370, China.
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province, The Ministry of Education of China, Guangzhou Medical University, Guangzhou, 510370, China.
| | - Kai Wu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, China.
- Guangdong Province Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510006, China.
- Department of Nuclear Medicine and Radiology, Institute of Development, Aging and Cancer, Tohoku University, Sendai, 980-8575, Japan.
| |
Collapse
|
2
|
Cho MY, Eom JH, Choi EM, Yang SJ, Lee D, Kim YY, Kim HS, Hwang I. Recent advances in therapeutic probiotics: insights from human trials. Clin Microbiol Rev 2025:e0024024. [PMID: 40261032 DOI: 10.1128/cmr.00240-24] [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: 04/24/2025] Open
Abstract
SUMMARYRecent advances in therapeutic probiotics have shown promising results across various health conditions, reflecting a growing understanding of the human microbiome's role in health and disease. However, comprehensive reviews integrating the diverse therapeutic effects of probiotics in human subjects have been limited. By analyzing randomized controlled trials (RCTs) and meta-analyses, this review provides a comprehensive overview of key developments in probiotic interventions targeting gut, liver, skin, vaginal, mental, and oral health. Emerging evidence supports the efficacy of specific probiotic strains and combinations in treating a wide range of disorders, from gastrointestinal (GI) and liver diseases to dermatological conditions, bacterial vaginosis, mental disorders, and oral diseases. We discuss the expanding understanding of microbiome-organ connections underlying probiotic mechanisms of action. While many clinical trials demonstrate significant benefits, we acknowledge areas requiring further large-scale studies to establish definitive efficacy and optimal treatment protocols. The review addresses challenges in standardizing probiotic research methodologies and emphasizes the importance of considering individual variations in microbiome composition and host genetics. Additionally, we explore emerging concepts such as the oral-gut-brain axis and future directions, including high-resolution microbiome profiling, host-microbe interaction studies, organoid models, and artificial intelligence applications in probiotic research. Overall, this review offers a comprehensive update on the current state of therapeutic probiotics across multiple domains of human health, providing insights into future directions and the potential for probiotics to revolutionize preventive and therapeutic medicine.
Collapse
Affiliation(s)
- Mu-Yeol Cho
- Apple Tree Institute of Biomedical Science, Apple Tree Medical Foundation, Goyang-si, South Korea
| | - Je-Hyun Eom
- Apple Tree Institute of Biomedical Science, Apple Tree Medical Foundation, Goyang-si, South Korea
| | - Eun-Mi Choi
- Apple Tree Institute of Biomedical Science, Apple Tree Medical Foundation, Goyang-si, South Korea
| | | | - Dahye Lee
- Department of Orthodontics, Apple Tree Dental Hospital, Goyang-si, South Korea
| | - Young Youn Kim
- Department of Oral and Maxillofacial Surgery, Apple Tree Dental Hospital, Goyang-si, South Korea
| | - Hye-Sung Kim
- Department of Oral and Maxillofacial Surgery, Apple Tree Dental Hospital, Goyang-si, South Korea
| | - Inseong Hwang
- Apple Tree Institute of Biomedical Science, Apple Tree Medical Foundation, Goyang-si, South Korea
| |
Collapse
|
3
|
Kovynev A, Charchuta MM, Begtašević A, Ducarmon QR, Rensen PCN, Schönke M. Combination of dietary fiber and exercise training improves fat loss in mice but does not ameliorate MASLD more than exercise alone. Am J Physiol Gastrointest Liver Physiol 2025; 328:G399-G410. [PMID: 40033967 DOI: 10.1152/ajpgi.00317.2024] [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: 10/03/2024] [Revised: 01/02/2025] [Accepted: 02/20/2025] [Indexed: 03/05/2025]
Abstract
Lifestyle interventions, such as diet and exercise, are currently the main therapies against metabolic dysfunction-associated steatotic liver disease (MASLD). However, not much is known about the combined impact of fiber and exercise on the modulation of gut-liver axis and MASLD amelioration. Here, we studied the impact of the combination of exercise training and a fiber-rich diet on the amelioration of MASLD. Male APOE*3-Leiden.CETP mice were fed a high-fat high-cholesterol diet with or without the addition of fiber (10% inulin) and exercise trained on a treadmill, or remained sedentary. Exercise training and fiber supplementation reduced fat mass gain and lowered plasma glucose levels. Only the combination treatment, however, induced fat loss and decreased plasma triglyceride and cholesterol levels compared with sedentary control mice. Exercise training with and without the addition of fiber had a similar ameliorating effect on the MASLD score. Only exercise without fiber decreased the hepatic expression of inflammatory markers. Fiber diet was mainly responsible for remodeling the gut microbial composition, with an increase in the relative abundance of the short-chain fatty acid (SCFA)-producing genera Anaerostipes and Muribaculaceae, whereas, surprisingly, exercise training alone and with fiber resulted in the highest increase of SCFA production. Overall, the combination of exercise training and dietary fiber decreases fat mass and improves glucose and lipid homeostasis but does not have an additional synergistic positive effect on liver health compared with exercise training alone.NEW & NOTEWORTHY The combination of dietary fiber intake and exercise training has a synergetic beneficial effect on the metabolic health, resulting in fat loss, lowered blood glucose, and lowered plasma lipid levels in mice with steatotic liver disease. However, fiber supplementation, despite a positive remodulation of the gut-liver axis, does not have an additional positive effect on liver health compared with exercise training alone.
Collapse
Affiliation(s)
- Artemiy Kovynev
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Mikołaj M Charchuta
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Amina Begtašević
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Quinten R Ducarmon
- Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Patrick C N Rensen
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Milena Schönke
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
4
|
Guo S, Chu H, Peng B, Niu J, Yang X, Guo Y, Wang Z, Zhang W. Effects of Lactic Acid and Glyceryl Lactate on Growth Performance, Antioxidant Capacity, and Intestinal Health of Piglets. Antioxidants (Basel) 2025; 14:391. [PMID: 40298648 PMCID: PMC12023924 DOI: 10.3390/antiox14040391] [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: 02/19/2025] [Revised: 03/16/2025] [Accepted: 03/24/2025] [Indexed: 04/30/2025] Open
Abstract
The aim of this study was to evaluate the effects of lactic acid and glyceryl lactate on growth performance, antioxidant capacity, and intestinal health in piglets. This study included 240 castrated male piglets (initial body weight: 7.50 ± 0.54 kg) assigned to four groups: CON (basal diet), LA (basal diet + 0.5% lactic acid), GL (basal diet + 0.5% glyceryl lactate), and LG (basal diet + 0.5% lactic acid + 0.5% glyceryl lactate). Each group had six replicates of 10 piglets. The trial lasted 28 days. Compared with the control group, the GL and LG groups showed enhanced growth performance and reduced diarrhea rate in piglets. The LA and LG groups showed decreased intestinal chyme pH and increased digestive enzyme activities. Moreover, the GL and LG groups displayed elevated jejunal mRNA levels of the tight junction protein occludin and mucin MUC2, enhanced expression levels of Nrf2 signaling pathway genes, increased activities of the antioxidant enzymes GPX and CAT, and reduced MDA content. Acidifier supplementation also modulated cecal bacterial abundance and short-chain fatty acid (SCFA) content. Genera such as Faecalibaculum, Nocardiopsis, Collinsella, CAG269, Allobaculum, and Enterococcus were affected. In conclusion, glyceryl lactate and its combination with lactic acid improved piglet growth performance by enhancing intestinal barrier function, antioxidant capacity, microbial community structure, and SCFA production.
Collapse
Affiliation(s)
- Shuaiju Guo
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; (S.G.); (B.P.); (J.N.); (X.Y.); (Y.G.)
| | - Huiling Chu
- College of Food and Information Engineering, Zhengzhou University of Light Industry, Zhengzhou 450000, China;
| | - Bangwang Peng
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; (S.G.); (B.P.); (J.N.); (X.Y.); (Y.G.)
| | - Junlong Niu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; (S.G.); (B.P.); (J.N.); (X.Y.); (Y.G.)
| | - Xiaopeng Yang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; (S.G.); (B.P.); (J.N.); (X.Y.); (Y.G.)
| | - Yongpeng Guo
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; (S.G.); (B.P.); (J.N.); (X.Y.); (Y.G.)
| | - Zhixiang Wang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; (S.G.); (B.P.); (J.N.); (X.Y.); (Y.G.)
| | - Wei Zhang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; (S.G.); (B.P.); (J.N.); (X.Y.); (Y.G.)
| |
Collapse
|
5
|
Xu T, Fang D, Xu T, Tao X, Wang Z, Liu Y. Exercise-driven gut microbiota alterations enhance colonization resistance against methicillin-resistant Staphylococcus aureus. Cell Rep 2025; 44:115424. [PMID: 40080501 DOI: 10.1016/j.celrep.2025.115424] [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: 09/05/2024] [Revised: 12/15/2024] [Accepted: 02/20/2025] [Indexed: 03/15/2025] Open
Abstract
Gut microbiota plays a crucial role in resisting the invasion of pathogens, particularly multidrug-resistant (MDR) bacteria, which pose a significant threat to public health. While exercise offers numerous health benefits, its impact on host colonization resistance remains largely unclear. In this study, we demonstrate that moderate exercise significantly reduces gut colonization by methicillin-resistant Staphylococcus aureus (MRSA), a clinically important MDR pathogen. Moreover, we identify an understudied strain of the intestinal probiotic Dubosiella newyorkensis (L8) as a critical factor in mediating exercise-induced colonization resistance against MRSA. Mechanistically, L8 enhances the deprivation of fucose, a crucial carbon source essential for MRSA growth and pathogenicity. This process relies on the high binding affinity of pyruvate to the ILE257 site of the lactate dehydrogenase in L8. Overall, our work highlights the importance of moderate exercise in maintaining host colonization resistance and demonstrates L8 as a probiotic in protecting against MRSA colonization.
Collapse
Affiliation(s)
- Tingting Xu
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Dan Fang
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Tianqi Xu
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Xiuying Tao
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Zhiqiang Wang
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; Institute of Comparative Medicine, Yangzhou University, Yangzhou 225009, China.
| | - Yuan Liu
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; Institute of Comparative Medicine, Yangzhou University, Yangzhou 225009, China.
| |
Collapse
|
6
|
Tan J, Yang R, Xiao L, Dong L, Zhong Z, Zhou L, Qin W. Risk Stratification in Immunoglobulin A Nephropathy Using Network Biomarkers: Development and Validation Study. J Med Internet Res 2025; 27:e65563. [PMID: 40063072 PMCID: PMC11933752 DOI: 10.2196/65563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/14/2024] [Accepted: 01/29/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND Traditional risk models for immunoglobulin A nephropathy (IgAN), which primarily rely on renal indicators, lack comprehensive assessment and therapeutic guidance, necessitating more refined and integrative approaches. OBJECTIVE This study integrated network biomarkers with unsupervised learning clustering (k-means clustering based on network biomarkers [KMN]) to refine risk stratification in IgAN and explore its clinical value. METHODS Involving a multicenter prospective cohort, we analyzed 1460 patients and validated the approach externally with 200 additional patients. Deeper metabolic and microbiomic insights were gained from 2 distinct cohorts: 63 patients underwent ultraperformance liquid chromatography-mass spectrometry, while another 45 underwent fecal 16S RNA sequencing. Our approach used hierarchical clustering and k-means methods, using 3 sets of indicators: demographic and renal indicators, renal and extrarenal indicators, and network biomarkers derived from all indicators. RESULTS Among 6 clustering methods tested, the KMN scheme was the most effective, accurately reflecting patient severity and prognosis with a prognostic accuracy area under the curve (AUC) of 0.77, achieved solely through cluster grouping without additional indicators. The KMN stratification significantly outperformed the existing International IgA Nephropathy Prediction Tool (AUC of 0.72) and renal function-renal histology grading schemes (AUC of 0.69). Clinically, this stratification facilitated personalized treatment, recommending angiotensin-converting enzyme inhibitors or angiotensin receptor blockers for lower-risk groups and considering immunosuppressive therapy for higher-risk groups. Preliminary findings also indicated a correlation between IgAN progression and alterations in serum metabolites and gut microbiota, although further research is needed to establish causality. CONCLUSIONS The effectiveness and applicability of the KMN scheme indicate its substantial potential for clinical application in IgAN management.
Collapse
Affiliation(s)
- Jiaxing Tan
- Division of Nephrology, Department of Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Rongxin Yang
- College of Computer Science, Sichuan University, Chengdu, China
| | - Liyin Xiao
- College of Computer Science, Sichuan University, Chengdu, China
| | - Lingqiu Dong
- Division of Nephrology, Department of Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Zhengxia Zhong
- Division of Nephrology, Department of Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Ling Zhou
- Division of Nephrology, Zigong Third People's Hospital, Zigong, China
| | - Wei Qin
- Division of Nephrology, Department of Medicine, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
7
|
Peng R, Wang W, Liang L, Han R, Li Y, Wang H, Wang Y, Li W, Feng S, Zhou J, Huang Y, Wu F, Wu K. The brain-gut microbiota network (BGMN) is correlated with symptom severity and neurocognition in patients with schizophrenia. Neuroimage 2025; 308:121052. [PMID: 39875038 DOI: 10.1016/j.neuroimage.2025.121052] [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: 12/14/2023] [Revised: 01/19/2025] [Accepted: 01/23/2025] [Indexed: 01/30/2025] Open
Abstract
The association between the human brain and gut microbiota, known as the "brain-gut-microbiota axis", is involved in the neuropathological mechanisms of schizophrenia (SZ); however, its association patterns and correlations with symptom severity and neurocognition are still largely unknown. In this study, 43 SZ patients and 55 normal controls (NCs) were included, and resting-state functional magnetic resonance imaging (rs-fMRI) and gut microbiota data were acquired for each participant. First, the brain features of brain images and functional brain networks were computed from rs-fMRI data; the gut features of gut microbiota abundance and the gut microbiota network were computed from gut microbiota data. Second, we propose a novel methodology to construct an individual brain-gut microbiota network (BGMN) for each participant by combining the brain and gut features via multiple strategies. Third, discriminative models between SZ patients and NCs were built using the connectivity matrices of the BGMN as input features. Moreover, the correlations between the most discriminative features and the scores of symptom severity and neurocognition were analyzed in SZ patients. The results showed that the best discriminative model between SZ patients and NCs was achieved using the connectivity matrices of the BGMN when all the brain and gut features were integrated, with an accuracy of 0.90 and an area under the curve value of 0.97. The most discriminative features were related primarily to the genera Faecalibacterium and Collinsella, in which the genus Faecalibacterium was linked to the visual system and subcortical cortices and the genus Collinsella was linked to the default network and subcortical cortices. Furthermore, parts of the most discriminative features were significantly correlated with the scores of neurocognition in the SZ patients. The methodology for constructing individual BGMNs proposed in this study can help us reveal the associations between the brain and gut microbiota and understand the neuropathology of SZ.
Collapse
Affiliation(s)
- Runlin Peng
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, China
| | - Wei Wang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, China
| | - Liqin Liang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, China
| | - Rui Han
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, China
| | - Yi Li
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, China
| | - Haiyuan Wang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, China
| | - Yuran Wang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, China
| | - Wenhao Li
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, China
| | - Shixuan Feng
- Department of Psychiatry, The Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou 510370, China
| | - Jing Zhou
- School of Material Science and Engineering, South China University of Technology, Guangzhou 510006, China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China
| | - Yuanyuan Huang
- Department of Psychiatry, The Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou 510370, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou 510370, China
| | - Fengchun Wu
- Department of Psychiatry, The Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou 510370, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou 510370, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou 510370, China.
| | - Kai Wu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China; Department of Aging Research and Geriatric Medicine, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan.
| |
Collapse
|
8
|
Houttu V, Boulund U, Troelstra M, Csader S, Stols-Gonçalves D, Mak AL, Dijk AMV, Bouts J, Winkelmeijer M, Verdoes X, van den Berg-Faay S, Lek D, Ronteltap T, de Haan F, Jorstad H, Männistö V, Savonen K, Pentikäinen H, Hanhineva K, Babu AF, Panagiotou G, van Delden O, Verheij J, Doukas M, Nederveen A, Schwab U, Grefhorst A, Nieuwdorp M, Holleboom AG. Deep phenotyping of patients with MASLD upon high-intensity interval training. JHEP Rep 2025; 7:101289. [PMID: 40051412 PMCID: PMC11883402 DOI: 10.1016/j.jhepr.2024.101289] [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: 02/26/2024] [Revised: 11/17/2024] [Accepted: 11/22/2024] [Indexed: 03/09/2025] Open
Abstract
Background & Aims Exercise is a key component of lifestyle management in patients with metabolic dysfunction-associated steatotic liver disease (MASLD), but neither its therapeutic effect on the active stage of the disease, that is metabolic dysfunction-associated steatohepatitis (MASH) nor the mediating mechanisms have been characterized. Therefore, we performed multi-omic phenotyping of patients with MASLD-MASH on an exercise program. Methods Fifteen patients with MASLD conducted high-intensity interval training (HIIT) combined with home-based training for 12 weeks. MASLD was evaluated using histology, transient elastography, and multiparametric magnetic resonance imaging (MRI) before and after the intervention. Change in maximal oxygen consumption (VO2max) and MRI-determined liver fat were compared with a control group of patients with MASLD (n = 22). RNA sequencing was performed on liver, muscle, and fat biopsies of patients in the exercise group. Stool was analyzed by shotgun metagenomics and untargeted metabolomics was performed on plasma, urine, adipose, and stool. Results HIIT increased VO2max by 10.1% and improved mitochondrial metabolism in skeletal muscle, indicating improved cardiorespiratory fitness and adherence. VO2max increased significantly in the exercise group compared with controls. Histologically, no reduction in steatosis, MASH, or liver fibrosis was observed; however, transient elastography tended to improve. MRI-determined liver fat did not change in the exercise group compared with controls. HIIT induced changes in mRNA expression of genes related to beiging of adipose tissue and fibrogenesis in liver. In addition, specific gut microbial taxa and metabolites changed. Conclusions HIIT increased cardiorespiratory fitness and induced beneficial gene expression changes in muscle, adipose tissue, and liver, but without translation into histological improvement of MASLD. Longer exercise intervention trials are warranted to validate or refute current recommendations for exercise as a cornerstone treatment for MASLD-MASH. Impact and implications Despite exercise being considered as a key component of lifestyle management for steatotic liver disease, neither the clinical effects nor the mechanisms involved are completely understood. We show that a high-intensity interval training (HIIT) program in 15 patients with metabolic dysfunction-associated steatotic liver disease (MASLD) improved cardiorespiratory fitness, compared with 22 control patients with MASLD who did not participate in an exercise program, however, it did not improve MASLD. HIIT induced a positive effect on fat tissue and muscle metabolism which was accompanied with changes in certain gut bacteria and metabolites in blood and urine. These findings improve our understanding of the effects of exercise on the whole-body metabolism in relation to steatotic liver disease. As such, this study provides a basis for future exercise interventions in patients with MASLD, required to thoroughly test current guideline advice for exercise as a cornerstone treatment for MASLD of all stages. Clinical trial registry Dutch Trial Register (registration number NL7932).
Collapse
Affiliation(s)
- Veera Houttu
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Experimental Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Ulrika Boulund
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Experimental Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Marian Troelstra
- Department of Radiology, and Nuclear Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Susanne Csader
- School of Medicine, Institute of Public Health, and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Daniela Stols-Gonçalves
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Anne Linde Mak
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Anne-Marieke van Dijk
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Julia Bouts
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Maaike Winkelmeijer
- Experimental Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Xanthe Verdoes
- Experimental Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Sandra van den Berg-Faay
- Department of Radiology, and Nuclear Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Donne Lek
- Polifysiek, Amsterdam University of Applied Science, Amsterdam, The Netherlands
| | - Ted Ronteltap
- Polifysiek, Amsterdam University of Applied Science, Amsterdam, The Netherlands
| | - Ferdinand de Haan
- Polifysiek, Amsterdam University of Applied Science, Amsterdam, The Netherlands
| | - Harald Jorstad
- Department of Cardiology, Amsterdam Movement Sciences, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Ville Männistö
- Department of Medicine, University of Eastern Finland, and Kuopio University Hospital, Kuopio, Finland
| | - Kai Savonen
- School of Medicine, Institute of Public Health, and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
- Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
| | | | - Kati Hanhineva
- School of Medicine, Institute of Public Health, and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
- Afekta Technologies Ltd., Kuopio, Finland
- Department of Life Technologies, Food Chemistry, and Food Development Unit, University of Turku, Turku, Finland
| | - Ambrin Farizah Babu
- School of Medicine, Institute of Public Health, and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
- Afekta Technologies Ltd., Kuopio, Finland
| | - Gianni Panagiotou
- Department of Microbiome Dynamics, Leibniz Institute for Natural Product Research, and Infection Biology, Hans Knöll Institute (HKI), Jena, Germany
- Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Faculty of Biological Sciences, Friedrich Schiller University, Jena, Germany
- Jena University Hospital, Friedrich Schiller University, Jena, Germany
| | - Otto van Delden
- Department of Interventional Radiology, Amsterdam UMC, Amsterdam, The Netherlands
| | - Joanne Verheij
- Department of Pathology, Amsterdam UMC, Amsterdam, The Netherlands
| | - Michial Doukas
- Department of Pathology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Aart Nederveen
- Department of Radiology, and Nuclear Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Ursula Schwab
- School of Medicine, Institute of Public Health, and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Aldo Grefhorst
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Experimental Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Max Nieuwdorp
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Experimental Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Adriaan Georgius Holleboom
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Experimental Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
9
|
Ng HY, Zhang L, Tan JT, Hui RWH, Yuen MF, Seto WK, Leung WK, Cheung KS. Gut Microbiota Predicts Treatment Response to Empagliflozin Among MASLD Patients Without Diabetes Mellitus. Liver Int 2025; 45:e70023. [PMID: 39950834 PMCID: PMC11827547 DOI: 10.1111/liv.70023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 01/11/2025] [Accepted: 01/30/2025] [Indexed: 02/17/2025]
Abstract
BACKGROUND AND AIM We aimed to investigate whether gut microbiota could predict the treatment response to pharmacological agents among metabolic dysfunction-associated steatotic liver disease (MASLD) patients without diabetes mellitus (DM), as data are lacking. METHODS We prospectively followed up non-diabetic MASLD patients who used empagliflozin. Clinical, anthropometric, laboratory assessments and magnetic resonance imaging-proton density fat fraction (MRI-PDFF) were performed from baseline to week 52 (EOT). Baseline stool samples were collected, and shotgun DNA metagenomic sequencing was performed to profile microbiome. The primary outcome was treatment response to empagliflozin at EOT, defined as MRI-PDFF decline ≥ 30% at EOT from baseline. Linear discriminant analysis [LDA] effect size was used to identify putative bacterial species. Multivariable logistic regression was used to derive adjusted odds ratio (aOR) of outcome with bacterial species by adjusting for clinical factors. RESULTS Twenty-two (48.9%) of 45 patients (median age: 56.9 years [IQR: 51.0-63.2]; male: 23 [51.1%]) achieved treatment response at EOT. There was difference in alpha diversity (Shannon index: p < 0.001; Simpson index: p = 0.001) and beta diversity (p = 0.048) in baseline microbiome between treatment response and non-response groups. Faecalibacterium prausnitzii (log10LDAscore = 4.27), Lachnospira pectinoschiza (log10LDAscore = 3.99), Anaerostipes hadrus (log10LDAscore = 3.98), Roseburia faecis (log10LDAscore = 3.97), Roseburia inulinivorans (log10LDAscore = 3.58) and Agathobaculum butyriciproducens (log10LDAscore = 2.77) were enriched in the treatment response group. L. pectinoschiza (aOR: 34.1; p = 0.015), A. hadrus (aOR:35.0; p = 0.032) and A. butyriciproducens (aOR:22.3; p = 0.023) independently predicted treatment response but not clinical factors. These three species collectively predicted treatment response with AUROC of 0.89 (95% CI: 0.80-0.99). CONCLUSIONS Certain gut bacterial species, particularly the combination of A. hadrus, L. pectinoschiza and A. butyriciproducens, may predict treatment response to empagliflozin in MAFLD patients without DM.
Collapse
Affiliation(s)
- Ho Yu Ng
- Li Ka Shing Faculty of MedicineThe University of Hong Kong, Queen Mary HospitalHong Kong
| | - Lina Zhang
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of MedicineThe University of Hong Kong, Queen Mary HospitalHong Kong
| | - Jing Tong Tan
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of MedicineThe University of Hong Kong, Queen Mary HospitalHong Kong
| | - Rex Wan Hin Hui
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of MedicineThe University of Hong Kong, Queen Mary HospitalHong Kong
| | - Man Fung Yuen
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of MedicineThe University of Hong Kong, Queen Mary HospitalHong Kong
- State Key Laboratory of Liver ResearchThe University of Hong KongHong Kong
| | - Wai Kay Seto
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of MedicineThe University of Hong Kong, Queen Mary HospitalHong Kong
- State Key Laboratory of Liver ResearchThe University of Hong KongHong Kong
| | - Wai K. Leung
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of MedicineThe University of Hong Kong, Queen Mary HospitalHong Kong
| | - Ka Shing Cheung
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of MedicineThe University of Hong Kong, Queen Mary HospitalHong Kong
| |
Collapse
|
10
|
Zhu W. Roles of olive oil and physical exercise in non-alcoholic fatty liver disease after ultrasound-based evaluation. World J Hepatol 2025; 17:100243. [PMID: 40027569 PMCID: PMC11866150 DOI: 10.4254/wjh.v17.i2.100243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 01/09/2025] [Accepted: 01/14/2025] [Indexed: 02/20/2025] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) without special drugs shows symptoms of liver fat accumulation and steatosis in patients without alcohol intake. Ultrasound evaluation is a critical method in the early diagnosis of NAFLD stages as well as image processing and should be encouraged. Olive oil is an important component of the Mediterranean diet and has a beneficial role in the prevention of NAFLD progression. Physical activity and exercise can have anti-oxidant and anti-inflammatory effects to reduce liver fat and body weight via regulation of mitochondrial capacity in the development of NAFLD. Both the Mediterranean diet and physical exercise should be combined to achieve the ideal fat content reduction and weight loss in patients with NAFLD.
Collapse
Affiliation(s)
- Wei Zhu
- Shanghai Xi Rong Information Science and Technology Company Limited, National Science and Technology Park, Tongji University, Shanghai 200092, China
- School of Life Sciences and Technology, Tongji University, Shanghai 200092, China.
| |
Collapse
|
11
|
Jarmakiewicz-Czaja S, Sokal-Dembowska A, Filip R. Effects of Selected Food Additives on the Gut Microbiome and Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD). MEDICINA (KAUNAS, LITHUANIA) 2025; 61:192. [PMID: 40005309 PMCID: PMC11857189 DOI: 10.3390/medicina61020192] [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: 12/30/2024] [Revised: 01/17/2025] [Accepted: 01/21/2025] [Indexed: 02/27/2025]
Abstract
The purpose of this article is to present selected food additives as disruptors of normal intestinal homeostasis with a potential impact on the development of metabolic dysfunction-associated steatotic liver disease (MASLD). A comprehensive literature search was conducted in three major electronic databases: PubMed, ScienceDirect, and Google Scholar. MASLD is a prevalent liver condition that is closely related to the global rise in obesity. Its pathogenesis is multifactorial, with genetic, environmental, and metabolic factors playing a key role. The "multiple-hit" hypothesis suggests that a Western-style diet, rich in ultra-processed foods, saturated fats, and food additives, combined with low physical activity, contributes to obesity, which promotes lipid accumulation in the liver. Recent studies underscore the role of impaired intestinal homeostasis in the development of MASLD. Food additives, including preservatives, emulsifiers, and sweeteners, affect gut health and liver function. Selected preservatives inhibit pathogenic microorganisms but disrupt the intestinal microbiota, leading to changes in intestinal permeability and liver dysfunction. Some emulsifiers and thickeners can cause inflammation and alter the gut microbiome, contributing to liver steatosis. Furthermore, the use of sweeteners such as sucralose and aspartame has been linked to changes in liver metabolism and intestinal microbial composition, which in turn promotes metabolic disorders.
Collapse
Affiliation(s)
- Sara Jarmakiewicz-Czaja
- Faculty of Health Sciences and Psychology, University of Rzeszow, 35-959 Rzeszow, Poland; (S.J.-C.); (A.S.-D.)
| | - Aneta Sokal-Dembowska
- Faculty of Health Sciences and Psychology, University of Rzeszow, 35-959 Rzeszow, Poland; (S.J.-C.); (A.S.-D.)
| | - Rafał Filip
- Gastroenterology Clinic, Center for Comprehensive Treatment of Inflammatory, Bowel Disease Regional Hospital No. 2 in Rzeszow, 35-301 Rzeszow, Poland
- Department of Internal Medicine, Faculty of Medicine, University of Rzeszow, 35-959 Rzeszow, Poland
| |
Collapse
|
12
|
Nychas E, Marfil-Sánchez A, Chen X, Mirhakkak M, Li H, Jia W, Xu A, Nielsen HB, Nieuwdorp M, Loomba R, Ni Y, Panagiotou G. Discovery of robust and highly specific microbiome signatures of non-alcoholic fatty liver disease. MICROBIOME 2025; 13:10. [PMID: 39810263 PMCID: PMC11730835 DOI: 10.1186/s40168-024-01990-y] [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: 11/03/2024] [Accepted: 11/26/2024] [Indexed: 01/16/2025]
Abstract
BACKGROUND The pathogenesis of non-alcoholic fatty liver disease (NAFLD) with a global prevalence of 30% is multifactorial and the involvement of gut bacteria has been recently proposed. However, finding robust bacterial signatures of NAFLD has been a great challenge, mainly due to its co-occurrence with other metabolic diseases. RESULTS Here, we collected public metagenomic data and integrated the taxonomy profiles with in silico generated community metabolic outputs, and detailed clinical data, of 1206 Chinese subjects w/wo metabolic diseases, including NAFLD (obese and lean), obesity, T2D, hypertension, and atherosclerosis. We identified highly specific microbiome signatures through building accurate machine learning models (accuracy = 0.845-0.917) for NAFLD with high portability (generalizable) and low prediction rate (specific) when applied to other metabolic diseases, as well as through a community approach involving differential co-abundance ecological networks. Moreover, using these signatures coupled with further mediation analysis and metabolic dependency modeling, we propose synergistic defined microbial consortia associated with NAFLD phenotype in overweight and lean individuals, respectively. CONCLUSION Our study reveals robust and highly specific NAFLD signatures and offers a more realistic microbiome-therapeutics approach over individual species for this complex disease. Video Abstract.
Collapse
Affiliation(s)
- Emmanouil Nychas
- Department of Microbiome Dynamics, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Beutenbergstraße 11A, Jena, 07745, Germany
| | - Andrea Marfil-Sánchez
- Department of Microbiome Dynamics, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Beutenbergstraße 11A, Jena, 07745, Germany
| | - Xiuqiang Chen
- Department of Microbiome Dynamics, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Beutenbergstraße 11A, Jena, 07745, Germany
| | - Mohammad Mirhakkak
- Department of Microbiome Dynamics, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Beutenbergstraße 11A, Jena, 07745, Germany
| | - Huating Li
- Department of Endocrinology and Metabolism, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Diabetes Institute, Shanghai, 200233, China
| | - Weiping Jia
- Department of Endocrinology and Metabolism, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Diabetes Institute, Shanghai, 200233, China
| | - Aimin Xu
- The State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China
- Department of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| | | | - Max Nieuwdorp
- Amsterdam UMC, Location AMC, Department of Vascular Medicine, University of Amsterdam, Amsterdam, The Netherlands
| | - Rohit Loomba
- Department of Medicine, MASLD Research Center, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Yueqiong Ni
- Department of Microbiome Dynamics, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Beutenbergstraße 11A, Jena, 07745, Germany.
- Department of Endocrinology and Metabolism, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Diabetes Institute, Shanghai, 200233, China.
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, Jena, Germany.
| | - Gianni Panagiotou
- Department of Microbiome Dynamics, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Beutenbergstraße 11A, Jena, 07745, Germany.
- Faculty of Biological Sciences, Friedrich Schiller University, Jena, 07745, Germany.
- Department of Medicine, The University of Hong Kong, Hong Kong SAR, China.
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, Jena, Germany.
| |
Collapse
|
13
|
Monti E, Vianello C, Leoni I, Galvani G, Lippolis A, D’Amico F, Roggiani S, Stefanelli C, Turroni S, Fornari F. Gut Microbiome Modulation in Hepatocellular Carcinoma: Preventive Role in NAFLD/NASH Progression and Potential Applications in Immunotherapy-Based Strategies. Cells 2025; 14:84. [PMID: 39851512 PMCID: PMC11764391 DOI: 10.3390/cells14020084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 01/07/2025] [Accepted: 01/08/2025] [Indexed: 01/26/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a heterogeneous tumor associated with several risk factors, with non-alcoholic fatty liver disease (NAFLD) emerging as an important cause of liver tumorigenesis. Due to the obesity epidemics, the occurrence of NAFLD has significantly increased with nearly 30% prevalence worldwide. HCC often arises in the background of chronic liver disease (CLD), such as nonalcoholic steatohepatitis (NASH) and cirrhosis. Gut microbiome (GM) alterations have been linked to NAFLD progression and HCC development, with several investigations reporting a crucial role for the gut-liver axis and microbial metabolites in promoting CLD. Moreover, the GM affects liver homeostasis, energy status, and the immune microenvironment, influencing the response to immunotherapy with interesting therapeutic implications. In this review, we summarize the main changes in the GM and derived metabolites (e.g., short-chain fatty acids and bile acids) occurring in HCC patients and influencing NAFLD progression, emphasizing their potential as early diagnostic biomarkers and prognostic tools. We discuss the weight loss effects of diet-based interventions and healthy lifestyles for the treatment of NAFLD patients, highlighting their impact on the restoration of the intestinal barrier and GM structure. We also describe encouraging preclinical findings on the modulation of GM to improve liver functions in CLD, boost the antitumor immune response (e.g., probiotic supplementations or anti-hypercholesterolemic drug treatment), and ultimately delay NAFLD progression to HCC. The development of safe and effective strategies that target the gut-liver axis holds promise for liver cancer prevention and treatment, especially if personalized options will be considered.
Collapse
Affiliation(s)
- Elisa Monti
- Department for Life Quality Studies, University of Bologna, Corso d’Augusto 237, 47921 Rimini, Italy; (E.M.); (C.V.); (I.L.); (G.G.); (A.L.); (C.S.)
- Centre for Applied Biomedical Research—CRBA, University of Bologna, 40138 Bologna, Italy
| | - Clara Vianello
- Department for Life Quality Studies, University of Bologna, Corso d’Augusto 237, 47921 Rimini, Italy; (E.M.); (C.V.); (I.L.); (G.G.); (A.L.); (C.S.)
- Centre for Applied Biomedical Research—CRBA, University of Bologna, 40138 Bologna, Italy
| | - Ilaria Leoni
- Department for Life Quality Studies, University of Bologna, Corso d’Augusto 237, 47921 Rimini, Italy; (E.M.); (C.V.); (I.L.); (G.G.); (A.L.); (C.S.)
- Centre for Applied Biomedical Research—CRBA, University of Bologna, 40138 Bologna, Italy
| | - Giuseppe Galvani
- Department for Life Quality Studies, University of Bologna, Corso d’Augusto 237, 47921 Rimini, Italy; (E.M.); (C.V.); (I.L.); (G.G.); (A.L.); (C.S.)
- Centre for Applied Biomedical Research—CRBA, University of Bologna, 40138 Bologna, Italy
| | - Annalisa Lippolis
- Department for Life Quality Studies, University of Bologna, Corso d’Augusto 237, 47921 Rimini, Italy; (E.M.); (C.V.); (I.L.); (G.G.); (A.L.); (C.S.)
| | - Federica D’Amico
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy; (F.D.); (S.R.); (S.T.)
| | - Sara Roggiani
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy; (F.D.); (S.R.); (S.T.)
- Human Microbiomics Unit, Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy
| | - Claudio Stefanelli
- Department for Life Quality Studies, University of Bologna, Corso d’Augusto 237, 47921 Rimini, Italy; (E.M.); (C.V.); (I.L.); (G.G.); (A.L.); (C.S.)
| | - Silvia Turroni
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy; (F.D.); (S.R.); (S.T.)
- IRCCS Azienda Ospedaliero, Universitaria di Bologna, 40138 Bologna, Italy
| | - Francesca Fornari
- Department for Life Quality Studies, University of Bologna, Corso d’Augusto 237, 47921 Rimini, Italy; (E.M.); (C.V.); (I.L.); (G.G.); (A.L.); (C.S.)
- IRCCS Azienda Ospedaliero, Universitaria di Bologna, 40138 Bologna, Italy
| |
Collapse
|
14
|
Fan YH, Zhang S, Wang Y, Wang H, Li H, Bai L. Inter-organ metabolic interaction networks in non-alcoholic fatty liver disease. Front Endocrinol (Lausanne) 2025; 15:1494560. [PMID: 39850476 PMCID: PMC11754069 DOI: 10.3389/fendo.2024.1494560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 12/16/2024] [Indexed: 01/25/2025] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a multisystem metabolic disorder, marked by abnormal lipid accumulation and intricate inter-organ interactions, which contribute to systemic metabolic imbalances. NAFLD may progress through several stages, including simple steatosis (NAFL), non-alcoholic steatohepatitis (NASH), cirrhosis, and potentially liver cancer. This disease is closely associated with metabolic disorders driven by overnutrition, with key pathological processes including lipid dysregulation, impaired lipid autophagy, mitochondrial dysfunction, endoplasmic reticulum (ER) stress, and local inflammation. While hepatic lipid metabolism in NAFLD is well-documented, further research into inter-organ communication mechanisms is crucial for a deeper understanding of NAFLD progression. This review delves into intrahepatic networks and tissue-specific signaling mediators involved in NAFLD pathogenesis, emphasizing their impact on distal organs.
Collapse
Affiliation(s)
- Yu-Hong Fan
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China
| | - Siyao Zhang
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China
| | - Ye Wang
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China
| | - Hongni Wang
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China
| | - Hongliang Li
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Ganzhou, China
- Department of Cardiology, Renmin Hospital, Wuhan University, Wuhan, China
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Lan Bai
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, China
| |
Collapse
|
15
|
Yu C, Sun R, Yang W, Gu T, Ying X, Ye L, Zheng Y, Fan S, Zeng X, Yao S. Exercise ameliorates osteopenia in mice via intestinal microbial-mediated bile acid metabolism pathway. Theranostics 2025; 15:1741-1759. [PMID: 39897551 PMCID: PMC11780523 DOI: 10.7150/thno.104186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 12/09/2024] [Indexed: 02/04/2025] Open
Abstract
Rationale: Physical exercise is essential for skeletal integrity and bone health. The gut microbiome, as a pivotal modulator of overall physiologic states, is closely associated with skeletal homeostasis and bone metabolism. However, the potential role of intestinal microbiota in the exercise-mediated bone gain remains unclear. Methods: We conducted microbiota depletion and fecal microbiota transplantation (FMT) in ovariectomy (OVX) mice and aged mice to investigate whether the transfer of gut ecological traits could confer the exercise-induced bone protective effects. The study analyzed the gut microbiota and metabolic profiles via 16S rRNA gene sequencing and LC-MS untargeted metabolomics to identify key microbial communities and metabolites responsible for bone protection. Transcriptome sequencing and RNA interference were employed to explore the molecular mechanisms. Results: We found that gut microbiota depletion hindered the osteogenic benefits of exercise, and FMT from exercised osteoporotic mice effectively mitigated osteopenia. Comprehensive profiling of the microbiome and metabolome revealed that the exercise-matched FMT reshaped intestinal microecology and metabolic landscape. Notably, alterations in bile acid metabolism, specifically the enrichment of taurine and ursodeoxycholic acid, mediated the protective effects on bone mass. Mechanistically, FMT from exercised mice activated the apelin signaling pathway and restored the bone-fat balance in recipient MSCs. Conclusion: Our study underscored the important role of the microbiota-metabolic axis in the exercise-mediated bone gain, heralding a potential breakthrough in the treatment of osteoporosis.
Collapse
Affiliation(s)
- Congcong Yu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration, Translational Research of Zhejiang Province Hangzhou, Zhejiang 310016, China
| | - Rongtai Sun
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration, Translational Research of Zhejiang Province Hangzhou, Zhejiang 310016, China
| | - Wentao Yang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration, Translational Research of Zhejiang Province Hangzhou, Zhejiang 310016, China
| | - Tianyuan Gu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration, Translational Research of Zhejiang Province Hangzhou, Zhejiang 310016, China
| | - Xiaozhang Ying
- Zhejiang Hospital of Integrated Traditional Chinese and Western Medicine, Hangzhou, Zhejiang 310016, China
| | - Lin Ye
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration, Translational Research of Zhejiang Province Hangzhou, Zhejiang 310016, China
| | - Yang Zheng
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration, Translational Research of Zhejiang Province Hangzhou, Zhejiang 310016, China
- Research Institute of Orthopedics, The Affiliated Jiangnan Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Shunwu Fan
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration, Translational Research of Zhejiang Province Hangzhou, Zhejiang 310016, China
| | - Xiangjun Zeng
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, Zhejiang 311100, China
| | - Shasha Yao
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration, Translational Research of Zhejiang Province Hangzhou, Zhejiang 310016, China
| |
Collapse
|
16
|
Feuerbacher JF, Cheng R, Sedliak M, Hu M, Finni TJ, Umlauff L, Schumann M, Cheng S. Serum Metabolome Signature Response to Different Types of Resistance Training. Int J Sports Med 2025; 46:22-31. [PMID: 39255827 DOI: 10.1055/a-2412-3410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Pneumatic resistance training (PRT) facilitates a longer time under tension that might lead to greater changes in body composition when compared to traditional resistance training (TRT), possibly enhancing serum metabolite concentrations indicative of healthy metabolic function. To assess the impact of PRT and TRT on muscular strength, body composition, and serum metabolome, 69 men (age: 31.8±7.2 years, height: 179.7±5.4 cm, weight: 81.1±9.9 kg) were randomized into two 10-week intervention groups (PRT:n=24 and TRT:n=24) and one control group (CON:n=21). Serum metabolite concentrations were assessed before and after the training intervention by high-throughput nuclear magnetic resonance. Fat mass and lean mass were obtained by bioimpedance analysis. The training intervention resulted in an increase in lean mass for both PRT (1.85±2.69%; p=0.003) and TRT (2.72±4.53%; p=0.004), while only PRT reduced statistically significantly in body fat percentage (PRT: -5.08±10.76%; p=0.019). Only in PRT and TRT significant increases in small high-density lipoproteins (S-HDL-L) and small HDL particles (S-HDL-P) were observed. When controlling for fat and lean mass, the effects on S-HDL-L/S-HDL-P diminished. Network analysis may suggest that PRT and TRT result in an increase in network connectivity and robustness. It appears that the observed improvements are associated with changes in body composition.
Collapse
Affiliation(s)
- Joshua Frederik Feuerbacher
- Department of Molecular and Cellular Sport Medicine, German Sport University Cologne, Cologne, Germany
- Department of Sports Medicine and Exercise Therapy, Chemnitz University of Technology, Chemnitz, Germany
| | - Runtan Cheng
- Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
- Faculty of Physical Education, Shanghai Jiao Tong University, Shanghai, China
| | - Milan Sedliak
- Department of Biological and Medical Sciences, Faculty of Physical Education, Comenius University in Bratislava, Bratislava, Slovakia
| | - Min Hu
- Guangzhou Sport University, Guangzhou Sport University, Guangzhou, China
| | - Taija Juutinen Finni
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyvaskyla, Finland
| | - Lisa Umlauff
- Department of Molecular and Cellular Sport Medicine, German Sport University Cologne, Cologne, Germany
| | - Moritz Schumann
- Department of Sports Medicine and Exercise Therapy, Chemnitz University of Technology, Chemnitz, Germany
- Faculty of Physical Education, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shulin Cheng
- Faculty of Physical Education, Shanghai Jiao Tong University, Shanghai, China
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyvaskyla, Finland
- Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
17
|
Li M, Yuan W, Kong X, Wu H, Cai Z, Zhu W, Lu X. Proton pump inhibitors reduce chemotherapeutic hepatotoxicity and enhance hepatic uptake and accumulation of drug-loaded extracellular vesicles. Sci Rep 2024; 14:28163. [PMID: 39548145 PMCID: PMC11568174 DOI: 10.1038/s41598-024-75775-6] [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: 04/23/2024] [Accepted: 10/08/2024] [Indexed: 11/17/2024] Open
Abstract
Extracellular vesicles (EVs) are involved in the progression of various diseases. Tumor cell-derived EVs (TEVs) are a particular concern, as they can induce fatty liver by promoting liver macrophages to secrete tumor necrosis factor (TNF), thus enhancing the toxicity of chemotherapy. Therefore, reducing pathogenic EV production is a potential strategy for treating EV-related diseases. However, there are currently no effective clinical reagents to obtain this purpose. In addition, EVs are also natural and ideal drug-delivery vehicles. Improving the delivery efficiency of EVs remains a challenge. Proton pump inhibitors (PPIs) have been demonstrated to promote cell uptake of EVs by inducing micropinocytosis. Here, we show that PPIs can accelerate TEV clearance, reduce TEV uptake by liver macrophages and decrease the mRNA expression of TNF in liver macrophages of tumor-bearing mice. Correspondingly, the fatty liver phenotypes are alleviated, and the tolerance to chemotherapy is improved in these mice. Furthermore, our findings indicate that PPIs facilitate the uptake of red blood cell-derived EVs (RBC-EVs) loaded with antisense oligonucleotides of Trim21 (Trim21-ASOs) by the liver macrophages of obesity. Consequently, the inhibition of macrophage inflammatory responses in obese mice mediated by RBC-EVs/Trim21-ASOs was further enhanced by PPIs, resulting in a more profound improvement in obesity and related metabolic disorders. In conclusion, our findings demonstrated that PPIs can effectively clear pathogenic EVs and enhance the delivery efficacy of EV vehicles, making them a highly promising clinical prospect.
Collapse
Affiliation(s)
- Mengyu Li
- Department of Internal Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310058, P.R. China
| | - Weiyi Yuan
- Institute of Immunology, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou, 310058, P.R. China
| | - Xianghui Kong
- Institute of Immunology, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou, 310058, P.R. China
| | - Hao Wu
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, P.R. China
| | - Zhijian Cai
- Institute of Immunology, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou, 310058, P.R. China
| | - Weiguo Zhu
- Department of Cardiology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, P.R. China.
| | - Xinliang Lu
- Institute of Immunology, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou, 310058, P.R. China.
| |
Collapse
|
18
|
Pérez-Prieto I, Plaza-Florido A, Ubago-Guisado E, Ortega FB, Altmäe S. Physical activity, sedentary behavior and microbiome: A systematic review and meta-analysis. J Sci Med Sport 2024; 27:793-804. [PMID: 39048485 DOI: 10.1016/j.jsams.2024.07.003] [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: 05/16/2023] [Revised: 05/18/2024] [Accepted: 07/02/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND The effects of physical activity and sedentary behavior on human health are well known, however, the molecular mechanisms are poorly understood. Growing evidence points to physical activity as an important modulator of the composition and function of microbial communities, while evidence of sedentary behavior is scarce. We aimed to synthesize and meta-analyze the current evidence about the effects of physical activity and sedentary behavior on microbiome across different body sites and in different populations. METHODS A systematic search in PubMed, Web of Science, Scopus and Cochrane databases was conducted until September 2022. Random-effects meta-analyses including cross-sectional studies (active vs. inactive/athletes vs. non-athletes) or trials reporting the chronic effect of physical activity interventions on gut microbiome alpha-diversity in healthy individuals were performed. RESULTS Ninety-one studies were included in this systematic review. Our meta-analyses of 2632 participants indicated no consistent effect of physical activity on microbial alpha-diversity, although there seems to be a trend toward a higher microbial richness in athletes compared to non-athletes. Most of studies reported an increase in short-chain fatty acid-producing bacteria such as Akkermansia, Faecalibacterium, Veillonella or Roseburia in active individuals and after physical activity interventions. CONCLUSIONS Physical activity levels were positively associated with the relative abundance of short-chain fatty acid-producing bacteria. Athletes seem to have a richer microbiome compared to non-athletes. However, high heterogeneity between studies avoids obtaining conclusive information on the role of physical activity in microbial composition. Future multi-omics studies would enhance our understanding of the molecular effects of physical activity and sedentary behavior on the microbiome.
Collapse
Affiliation(s)
- Inmaculada Pérez-Prieto
- Department of Biochemistry and Molecular Biology I, Faculty of Sciences, University of Granada, Spain; Instituto de Investigación Biosanitaria ibs.GRANADA, Spain.
| | - Abel Plaza-Florido
- Department of Physical Education and Sports, Faculty of Sport Sciences, Sport and Health University Research Institute (iMUDS), University of Granada, Spain; Pediatric Exercise and Genomics Research Center, UC Irvine School of Medicine, United States.
| | - Esther Ubago-Guisado
- Instituto de Investigación Biosanitaria ibs.GRANADA, Spain; Department of Physical Education and Sports, Faculty of Sport Sciences, Sport and Health University Research Institute (iMUDS), University of Granada, Spain
| | - Francisco B Ortega
- Department of Physical Education and Sports, Faculty of Sport Sciences, Sport and Health University Research Institute (iMUDS), University of Granada, Spain; CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Granada, Spain; Faculty of Sport and Health Sciences, University of Jyväskylä, Finland.
| | - Signe Altmäe
- Department of Biochemistry and Molecular Biology I, Faculty of Sciences, University of Granada, Spain; Instituto de Investigación Biosanitaria ibs.GRANADA, Spain; Division of Obstetrics and Gynaecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Huddinge, Stockholm, Sweden; Department of Gynaecology and Reproductive Medicine, Karolinska University Hospital, Huddinge, Stockholm, Sweden.
| |
Collapse
|
19
|
Varghese S, Rao S, Khattak A, Zamir F, Chaari A. Physical Exercise and the Gut Microbiome: A Bidirectional Relationship Influencing Health and Performance. Nutrients 2024; 16:3663. [PMID: 39519496 PMCID: PMC11547208 DOI: 10.3390/nu16213663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 10/18/2024] [Accepted: 10/19/2024] [Indexed: 11/16/2024] Open
Abstract
Background/Objectives: The human gut microbiome is a complex ecosystem of microorganisms that can influence our health and exercise habits. On the other hand, physical exercise can also impact our microbiome, affecting our health. Our narrative review examines the bidirectional relationship between physical activity and the gut microbiome, as well as the potential for targeted probiotic regimens to enhance sports performance. Methods: We conducted a comprehensive literature review to select articles published up till January 2024 on the topics of physical exercise, sports, probiotics, and gut microbiota from major scientific databases, incorporating over 100 studies. Results: We found that the impact of physical activity on the gut microbiome varies with the type and intensity of exercise. Moderate exercise promotes a healthy immune system, while high-intensity exercise for a long duration can cause a leaky gut and consequent systemic inflammation, which may disrupt the microbial balance. Combining aerobic and resistance training significantly affects bacterial diversity, linked to a lower prevalence of chronic metabolic disorders. Furthermore, exercise enhances gut microbiome diversity, increases SCFA production, improves nutrient utilization, and modulates neural and hormonal pathways, improving gut barrier integrity. Our findings also showed probiotic supplementation is associated with decreased inflammation, enhanced sports performance, and fewer gastrointestinal disturbances, suggesting that the relationship between the gut microbiome and physical activity is mutually influential. Conclusions: The bidirectional relationship between physical activity and the gut microbiome is exemplified by how exercise can promote beneficial bacteria while a healthy gut microbiome can potentially enhance exercise ability through various mechanisms. These findings underscore the importance of adding potential tailored exercise regimens and probiotic supplementation that consider individual microbiome profiles into exercise programs.
Collapse
Affiliation(s)
| | | | | | | | - Ali Chaari
- Department of Biochemistry, Premedical Division, Weill Cornell Medicine–Qatar, Qatar Foundation, Education City, Doha P.O. Box 24144, Qatar; (S.V.); (S.R.); (A.K.); (F.Z.)
| |
Collapse
|
20
|
Keating SE, Chawla Y, De A, George ES. Lifestyle intervention for metabolic dysfunction-associated fatty liver disease: a 24-h integrated behavior perspective. Hepatol Int 2024; 18:959-976. [PMID: 38717691 PMCID: PMC11450077 DOI: 10.1007/s12072-024-10663-9] [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: 12/29/2023] [Accepted: 02/13/2024] [Indexed: 10/05/2024]
Abstract
INTRODUCTION The prevalence, health and socioeconomic burden of metabolic dysfunction-associated fatty liver disease (MAFLD) is growing, increasing the need for novel evidence-based lifestyle approaches. Lifestyle is the cornerstone for MAFLD management and co-existing cardiometabolic dysfunction. The aim of this review was to evaluate the evidence for lifestyle management of MAFLD, with a specific lens on 24-hour integrated behaviour and provide practical recommendations for implementation of the evidence. RESULTS Weight loss ≥ 7-10% is central to lifestyle management; however, liver and cardiometabolic benefits are attainable with improved diet quality and exercise even without weight loss. Lifestyle intervention for MAFLD should consider an integrated '24-h' approach that is cognisant of diet, physical activity/exercise, sedentary behavior, smoking, alcohol intake and sleep. Dietary management emphasises energy deficit and improved diet quality, especially the Mediterranean diet, although sociocultural adaptations to meet preferences should be considered. Increasing physical activity and reducing sedentary behavior can prevent MAFLD, with strongest evidence in MAFLD supporting regular structured moderate-vigorous aerobic exercise for 150-240 min/week. Resistance training in addition to aerobic exercise should be considered and prioritised for those who are losing body mass via diet and/or pharmacological approaches and those with sarcopenia, to minimise bone and lean mass loss. Limited evidence suggests that sleep is important for MAFLD prevention. Emerging novel approaches to diet and exercise may address some of the key barriers to behaviour change (e.g. lack of time, access to resources and social support). FUTURE DIRECTIONS Large-scale multidisciplinary trials in people with MAFLD with long-term follow-up, that can be scaled up into mainstream healthcare, are required. Future management guidelines should consider the heterogeneity of MAFLD and specialised models of care that coordinate the health workforce to manage the increased and growing MAFLD population.
Collapse
Affiliation(s)
- Shelley E Keating
- School of Human Movement and Nutrition Sciences, The University of Queensland, Brisbane, QLD, Australia.
| | - Yogesh Chawla
- Kalinga Institute of Medical Sciences (KIMS), Bhubaneshwar, India
| | - Arka De
- Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Elena S George
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Australia
| |
Collapse
|
21
|
Guan H, Zhao S, Li J, Wang Y, Niu P, Zhang Y, Zhang Y, Fang X, Miao R, Tian J. Exploring the design of clinical research studies on the efficacy mechanisms in type 2 diabetes mellitus. Front Endocrinol (Lausanne) 2024; 15:1363877. [PMID: 39371930 PMCID: PMC11449758 DOI: 10.3389/fendo.2024.1363877] [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] [Received: 01/08/2024] [Accepted: 08/23/2024] [Indexed: 10/08/2024] Open
Abstract
This review examines the complexities of Type 2 Diabetes Mellitus (T2DM), focusing on the critical role of integrating omics technologies with traditional experimental methods. It underscores the advancements in understanding the genetic diversity of T2DM and emphasizes the evolution towards personalized treatment modalities. The paper analyzes a variety of omics approaches, including genomics, methylation, transcriptomics, proteomics, metabolomics, and intestinal microbiomics, delineating their substantial contributions to deciphering the multifaceted mechanisms underlying T2DM. Furthermore, the review highlights the indispensable role of non-omics experimental techniques in comprehending and managing T2DM, advocating for their integration in the development of tailored medicine and precision treatment strategies. By identifying existing research gaps and suggesting future research trajectories, the review underscores the necessity for a comprehensive, multidisciplinary approach. This approach synergistically combines clinical insights with cutting-edge biotechnologies, aiming to refine the management and therapeutic interventions of T2DM, and ultimately enhancing patient outcomes. This synthesis of knowledge and methodologies paves the way for innovative advancements in T2DM research, fostering a deeper understanding and more effective treatment of this complex condition.
Collapse
Affiliation(s)
- Huifang Guan
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Shuang Zhao
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Jiarui Li
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Ying Wang
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Ping Niu
- Department of Encephalopathy, The Affiliated Hospital of Changchun university of Chinese Medicine, Jilin, China
| | - Yuxin Zhang
- Institute of Metabolic Diseases, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yanjiao Zhang
- Institute of Metabolic Diseases, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xinyi Fang
- Institute of Metabolic Diseases, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate College, Beijing University of Chinese Medicine, Beijing, China
| | - Runyu Miao
- Institute of Metabolic Diseases, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate College, Beijing University of Chinese Medicine, Beijing, China
| | - Jiaxing Tian
- Institute of Metabolic Diseases, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
22
|
Garcia-Mateo S, Rondinella D, Ponziani FR, Miele L, Gasbarrini A, Cammarota G, Lanas Á, Gomollón F. Gut microbiome and metabolic dysfunction-associated steatotic liver disease: Pathogenic role and potential for therapeutics. Best Pract Res Clin Gastroenterol 2024; 72:101924. [PMID: 39645278 DOI: 10.1016/j.bpg.2024.101924] [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] [Received: 04/06/2024] [Accepted: 05/02/2024] [Indexed: 12/09/2024]
Abstract
Gut microbiota plays key functions in the human body, and its alteration is associated with several human disorders. Moreover, its manipulation is being investigated as a potential therapeutic strategy. In this narrative review we will dissect the involvement of the gut microbiota and of the gut-liver axis on metabolic dysfunction-associated steatotic liver disease (MASLD). Additionally, we will review the effects of lifestyle interventions commonly used for MASLD (i.e. Mediterranean diet and physical exercise) on gut microbiome, to understand if their beneficial effect can be microbially mediated. Finally, we will discuss the role and the available evidence of therapeutic microbiome modulators, including prebiotics, probiotics, symbiotics, and fecal microbiota transplantation (FMT), in the management of MASLD.
Collapse
Affiliation(s)
- Sandra Garcia-Mateo
- Department of Gastroenterology, "Lozano Blesa" Clinical Hospital, 50009, Zaragoza, Spain; Aragón Health Research Institute (IIS Aragón), 50009, Zaragoza, Spain; School of Medicine, University of Zaragoza, 50009, Zaragoza, Spain.
| | - Debora Rondinella
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
| | - Francesca Romana Ponziani
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
| | - Luca Miele
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
| | - Antonio Gasbarrini
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
| | - Giovanni Cammarota
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
| | - Ángel Lanas
- Department of Gastroenterology, "Lozano Blesa" Clinical Hospital, 50009, Zaragoza, Spain; Aragón Health Research Institute (IIS Aragón), 50009, Zaragoza, Spain; School of Medicine, University of Zaragoza, 50009, Zaragoza, Spain
| | - Fernando Gomollón
- Department of Gastroenterology, "Lozano Blesa" Clinical Hospital, 50009, Zaragoza, Spain; Aragón Health Research Institute (IIS Aragón), 50009, Zaragoza, Spain; School of Medicine, University of Zaragoza, 50009, Zaragoza, Spain
| |
Collapse
|
23
|
Li W, Tang H, Xue K, Ying T, Wu M, Qu Z, Dong C, Jin T, Brunius C, Hallmans G, Åman P, Johansson A, Landberg R, Liu Y, He G. Personalized Microbial Fingerprint Associated with Differential Glycemic Effects of a Whole Grain Rye Intervention on Chinese Adults. Mol Nutr Food Res 2024; 68:e2400274. [PMID: 39091068 DOI: 10.1002/mnfr.202400274] [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: 04/14/2024] [Revised: 07/13/2024] [Indexed: 08/04/2024]
Abstract
SCOPE This study aims to identify the gut enterotypes that explain differential responses to intervention with whole grain rye by proposing an "enterotype - metabolic" model. METHODS AND RESULTS A 12-week randomized controlled trial is conducted in Chinese adults, with 79 subjects consuming whole grain products with fermented rye bran (FRB) and 77 consuming refined wheat products in this exploratory post-hoc analysis. Responders or non-responders are identified according to whether blood glucose decreased by more than 10% after rye intervention. Compared to non-responders, responders in FRB have higher baseline Bacteroides (p < 0.001), associated with reduced blood glucose (p < 0.001), increased Faecalibacterium (p = 0.020) and Erysipelotrichaceae_UCG.003 (p = 0.022), as well as deceased 7β-hydroxysteroid dehydrogenase (p = 0.033) after intervention. The differentiated gut microbiota and metabolites between responders and non-responders after intervention are enriched in aminoacyl-tRNA biosynthesis. CONCLUSION The work confirms the previously suggested importance of microbial enterotypes in differential responses to whole grain interventions and supports taking enterotypes into consideration for improved efficacy of whole grain intervention for preventing type 2 diabetes. Altered short-chain fatty acids and bile acid metabolism might be a potential mediator for the beneficial effects of whole grain rye on glucose metabolism.
Collapse
Affiliation(s)
- Wenyun Li
- School of Public Health, Fudan University/Key Laboratory of Public Health Safety, Ministry of Education, Shanghai, 200032, China
| | - Huiru Tang
- State Key Laboratory of Genetic Engineering, Zhongshan Hospital and School of Life Sciences, Laboratory of Metabonomics and Systems Biology, Human Phenome Institute, Fudan University, Shanghai, 200032, China
| | - Kun Xue
- School of Public Health, Fudan University/Key Laboratory of Public Health Safety, Ministry of Education, Shanghai, 200032, China
| | - Tao Ying
- School of Public Health, Fudan University/Key Laboratory of Public Health Safety, Ministry of Education, Shanghai, 200032, China
| | - Min Wu
- School of Public Health, Fudan University/Key Laboratory of Public Health Safety, Ministry of Education, Shanghai, 200032, China
| | - Zheng Qu
- Department of Gastroenterology, Zhongye Hospital, Shanghai, 200003, China
| | - Chenglin Dong
- Department of Clinical Laboratory, Zhongye Hospital, Shanghai, 200003, China
| | - Taiyi Jin
- School of Public Health, Fudan University/Key Laboratory of Public Health Safety, Ministry of Education, Shanghai, 200032, China
| | - Carl Brunius
- Department of Life Sciences, Chalmers University of Technology, Gothenburg, 412 96, Sweden
| | - Göran Hallmans
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, 901 87, Sweden
| | - Per Åman
- Department of Molecular Sciences, Swedish University of Agricultural Sciences, Uppsala, 750 07, Sweden
| | - Anders Johansson
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, 901 87, Sweden
- Department of Odontology, Section of Molecular Periodontology, Umeå University, Umeå, 901 87, Sweden
| | - Rikard Landberg
- Department of Life Sciences, Chalmers University of Technology, Gothenburg, 412 96, Sweden
| | - Yuwei Liu
- School of Public Health, Fudan University/Key Laboratory of Public Health Safety, Ministry of Education, Shanghai, 200032, China
| | - Gengsheng He
- School of Public Health, Fudan University/Key Laboratory of Public Health Safety, Ministry of Education, Shanghai, 200032, China
| |
Collapse
|
24
|
Li M, Wang L, Lin D, Liu Z, Wang H, Yang Y, Sun C, Ye J, Liu Y. Advanced Bioinspired Multifunctional Platforms Focusing on Gut Microbiota Regulation. ACS NANO 2024; 18:20886-20933. [PMID: 39080827 DOI: 10.1021/acsnano.4c05013] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2025]
Abstract
Gut microbiota plays a crucial role in maintaining host homeostasis, impacting the progression and therapeutic outcomes of diseases, including inflammatory bowel disease, cancer, hepatic conditions, obesity, cardiovascular pathologies, and neurologic disorders, via immune, neural, and metabolic mechanisms. Hence, the gut microbiota is a promising target for disease therapy. The safety and precision of traditional microbiota regulation methods remain a challenge, which limits their widespread clinical application. This limitation has catalyzed a shift toward the development of multifunctional delivery systems that are predicated on microbiota modulation. Guided by bioinspired strategies, an extensive variety of naturally occurring materials and mechanisms have been emulated and harnessed for the construction of platforms aimed at the monitoring and modulation of gut microbiota. This review outlines the strategies and advantages of utilizing bioinspired principles in the design of gut microbiota intervention systems based on traditional regulation methods. Representative studies on the development of bioinspired therapeutic platforms are summarized, which are based on gut microbiota modulation to confer multiple pharmacological benefits for the synergistic management of diseases. The prospective avenues and inherent challenges associated with the adoption of bioinspired strategies in the refinement of gut microbiota modulation platforms are proposed to augment the efficacy of disease treatment.
Collapse
Affiliation(s)
- Muqing Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, P.R. China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, P.R. China
| | - LuLu Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, P.R. China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, P.R. China
| | - Demin Lin
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, P.R. China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, P.R. China
| | - Zihan Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, P.R. China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, P.R. China
| | - Hongliang Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, P.R. China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, P.R. China
| | - Yanfang Yang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, P.R. China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, P.R. China
| | - Chunmeng Sun
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, P.R. China
| | - Jun Ye
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, P.R. China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, P.R. China
| | - Yuling Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, P.R. China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, P.R. China
| |
Collapse
|
25
|
Xue J, Liu Y, Liu B, Jia X, Fang X, Qin S, Zhang Y. Celastrus orbiculatus Thunb. extracts and celastrol alleviate NAFLD by preserving mitochondrial function through activating the FGF21/AMPK/PGC-1α pathway. Front Pharmacol 2024; 15:1444117. [PMID: 39161898 PMCID: PMC11330833 DOI: 10.3389/fphar.2024.1444117] [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: 06/06/2024] [Accepted: 07/22/2024] [Indexed: 08/21/2024] Open
Abstract
Objective Non-alcoholic fatty liver disease (NAFLD) is a prevalent chronic liver disease globally, characterized by the accumulation of lipids, oxidative stress, and mitochondrial dysfunction in the liver. Celastrus orbiculatus Thunb. (COT) and its active compound celastrol (CEL) have demonstrated antioxidant and anti-inflammatory properties. Our prior research has shown the beneficial effects of COT in mitigating NAFLD induced by a high-fat diet (HFD) in guinea pigs by reducing hepatic lipid levels and inhibiting oxidative stress. This study further assessed the effects of COT on NAFLD and explored its underlying mitochondria-related mechanisms. Methods COT extract or CEL was administered as an intervention in C57BL/6J mice fed a HFD or in HepG2 cells treated with sodium oleate. Oral glucose tolerance test, biochemical parameters including liver enzymes, blood lipid, and pro-inflammatory factors, and steatosis were evaluated. Meanwhile, mitochondrial ultrastructure and indicators related to oxidative stress were tested. Furthermore, regulators of mitochondrial function were measured using RT-qPCR and Western blot. Results The findings demonstrated significant reductions in hepatic steatosis, oxidative stress, and inflammation associated with NAFLD in both experimental models following treatment with COT extract or CEL. Additionally, improvements were observed in mitochondrial structure, ATP content, and ATPase activity. This improvement can be attributed to the significant upregulation of mRNA and protein expression levels of key regulators including FGF21, AMPK, PGC-1α, PPARγ, and SIRT3. Conclusion These findings suggest that COT may enhance mitochondrial function by activating the FGF21/AMPK/PGC-1α signaling pathway to mitigate NAFLD, which indicated that COT has the potential to target mitochondria and serve as a novel therapeutic option for NAFLD.
Collapse
Affiliation(s)
- Junli Xue
- Taishan Institute for Hydrogen Biomedicine, The Second Affiliated Hospital of Shandong First Medical University and Shandong Academy of Medical Sciences, Tai’an, Shandong, China
| | - Yunchao Liu
- School of Pharmaceutical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Ji’nan, Shandong, China
| | - Boyan Liu
- Taishan Institute for Hydrogen Biomedicine, The Second Affiliated Hospital of Shandong First Medical University and Shandong Academy of Medical Sciences, Tai’an, Shandong, China
| | - Xiubin Jia
- School of Pharmaceutical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Ji’nan, Shandong, China
| | - Xinsheng Fang
- College of Agronomy, Shandong Agricultural University, Tai’an, Shandong, China
| | - Shucun Qin
- Taishan Institute for Hydrogen Biomedicine, The Second Affiliated Hospital of Shandong First Medical University and Shandong Academy of Medical Sciences, Tai’an, Shandong, China
| | - Ying Zhang
- School of Pharmaceutical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Ji’nan, Shandong, China
| |
Collapse
|
26
|
Gao W, Chen X, Wu S, Jin L, Chen X, Mao G, Wan X, Xing W. Monascus red pigments alleviate high-fat and high-sugar diet-induced NAFLD in mice by modulating the gut microbiota and metabolites. Food Sci Nutr 2024; 12:5762-5775. [PMID: 39139961 PMCID: PMC11317676 DOI: 10.1002/fsn3.4208] [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: 12/18/2023] [Revised: 03/21/2024] [Accepted: 04/27/2024] [Indexed: 08/15/2024] Open
Abstract
Monascus red pigments (MRP) may have benefits against NAFLD with an unclear mechanism. This study aimed to explore the protective effect of MRP supplementation against NAFLD through regulation of gut microbiota and metabolites. The C57BL/6 mice animals were randomly allocated into the normal diet (NC), HFHS diet-induced NAFLD model, and MRP intervention group fed with HFHS diet. Serum lipid profiles and liver function parameters were measured. Liver and colon histopathology analysis was conducted to determine the injury in the liver and colon. 16S rRNA gene sequencing was employed to analyze gut microbial composition from fecal samples. Untargeted metabonomics was performed to analyze changes in metabolites in serum and fecal samples. MRP supplementation significantly improved the HFHS-induced alterations in body weight, lipid profiles, and liver function (p < .01). MRP supplementation decreased the abundance of Akkermansia, Candidatus saccharimonas, Dubosiella, and Oscillibacter, while increasing Lactobacillus, Lachnospiraceae NK4A136 group, and Rikenella in mice fed the HFHS diet. Furthermore, MRP supplementation improved the serum and fecal metabolic profiles induced by the HFHS diet, primarily involving the arachidonic acid metabolism, unsaturated fatty acid biosynthesis, and adipocyte lipolysis pathways. Liver function and lipid profiles were closely associated with the abundance of Lactobacillus, Streptococcus, Oscillibacter, Akkemansia, and Desulfovibrio (p < .01). These findings revealed that MRP supplementation may help restore gut microbiota composition and balance its metabolites, thereby improving NAFLD. This study presents a novel outlook on the potential benefits of MRP supplementation in ameliorating NAFLD and supports the application of MRP as a new functional food.
Collapse
Affiliation(s)
- Wenyan Gao
- School of PharmacyHangzhou Medical CollegeHangzhouChina
| | - Xinghao Chen
- School of PharmacyHangzhou Medical CollegeHangzhouChina
| | - Shaokang Wu
- Department of PharmacyQingdao Sixth People's HospitalQingdaoChina
| | - Lu Jin
- School of PharmacyHangzhou Medical CollegeHangzhouChina
| | - Xu Chen
- School of PharmacyHangzhou Medical CollegeHangzhouChina
| | - Genxiang Mao
- Zhejiang Provincial Key Lab of GeriatricsZhejiang HospitalHangzhouChina
| | - Xiaoqing Wan
- Zhejiang Provincial Key Lab of GeriatricsZhejiang HospitalHangzhouChina
| | - Wenmin Xing
- Zhejiang Provincial Key Lab of GeriatricsZhejiang HospitalHangzhouChina
| |
Collapse
|
27
|
Liu Q, Li X, Pan Y, Liu Q, Li Y, He C, Zheng N, Wang Y, Wang H, Wang Y, Sheng L, Zhang B, Shen T, Wu G, Li H, Wang X, Zhang W, Hu Y, Zhao Y. Efficacy and safety of Qushi Huayu, a traditional Chinese medicine, in patients with nonalcoholic fatty liver disease in a randomized controlled trial. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 130:155398. [PMID: 38788390 DOI: 10.1016/j.phymed.2024.155398] [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: 12/04/2023] [Revised: 01/19/2024] [Accepted: 01/28/2024] [Indexed: 05/26/2024]
Abstract
BACKGROUND The effective treatment of non-alcoholic fatty liver disease (NAFLD) is an unmet medical need. Qushi Huayu (QSHY) is an empirical herbal formula with promising effects in NAFLD rodent models and a connection to gut microbiota regulation. HYPOTHESIS/PURPOSE This study aimed to evaluate the effects of QSHY in patients with NAFLD through a multicenter, randomized, double-blind, double-dummy clinical trial. STUDY DESIGN A total of 246 eligible patients with NAFLD and liver dysfunction were evenly divided to receive either QSHY and Dangfei Liganning capsule (DFLG) simulant or QSHY simulant and DFLG (an approved proprietary Chinese medicine for NAFLD in China) for 24 weeks. The primary outcomes were changes in liver fat content, assessed using vibration-controlled transient elastography, and serum alanine aminotransferase (ALT) levels from baseline to Week 24. RESULTS Both QSHY and DFLG led to reductions in liver fat content and liver enzyme levels post-intervention (p < 0.05). Compared to DFLG, QSHY treatment improved ALT (β, -0.128 [95 % CI, -0.25, -0.005], p = 0.041), aspartate transaminase (β, -0.134 [95 % CI, -0.256 to -0.012], p = 0.032), and fibrosis-4 score (β, -0.129 [95 % CI, -0.254 to -0.003], p = 0.044) levels. QSHY markedly improved gut dysbiosis compared to DFLG, with changes in Escherichia-Shigella and Bacteroides abundance linked to its therapeutic effect on reducing ALT. Patients with a high ALT response after QSHY treatment showed superior reductions in peripheral levels of phenylalanine and tyrosine, along with an elevation in the related microbial metabolite p-Hydroxyphenylacetic acid. CONCLUSION Our results demonstrate favorable clinical potential for QSHY in the treatment of NAFLD.
Collapse
Affiliation(s)
- Qiaohong Liu
- Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Institute of Liver Diseases, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiaojing Li
- Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Institute of Liver Diseases, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yuqing Pan
- Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Institute of Liver Diseases, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Qian Liu
- Department of gastroenterology, Baoshan District Hospital of Integrated Traditional Chinese Medicine of Shanghai, Shanghai 201900, China
| | - Ying Li
- Department of Infectious disease, Longhua Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Cong He
- Department of gastroenterology, Yueyang Hospital of Integrative Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Ningning Zheng
- School of Pharmacy, Shanghai University of Traditional ChineseMedicine, Shanghai 201203, China
| | - Yan Wang
- Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Institute of Liver Diseases, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Huichao Wang
- Department of gastroenterology, Yueyang Hospital of Integrative Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Yan Wang
- Department of Infectious disease, Longhua Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Lili Sheng
- School of Pharmacy, Shanghai University of Traditional ChineseMedicine, Shanghai 201203, China
| | - Binbin Zhang
- Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Institute of Liver Diseases, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Tianbai Shen
- Department of Infectious disease, Longhua Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Gaosong Wu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Houkai Li
- School of Pharmacy, Shanghai University of Traditional ChineseMedicine, Shanghai 201203, China
| | - Xiaosu Wang
- Department of gastroenterology, Yueyang Hospital of Integrative Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China.
| | - Wei Zhang
- Department of Infectious disease, Longhua Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| | - Yiyang Hu
- Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Institute of Liver Diseases, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Yu Zhao
- Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Institute of Liver Diseases, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
28
|
Mac Aogáin M, Dicker AJ, Mertsch P, Chotirmall SH. Infection and the microbiome in bronchiectasis. Eur Respir Rev 2024; 33:240038. [PMID: 38960615 PMCID: PMC11220623 DOI: 10.1183/16000617.0038-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 05/02/2024] [Indexed: 07/05/2024] Open
Abstract
Bronchiectasis is marked by bronchial dilatation, recurrent infections and significant morbidity, underpinned by a complex interplay between microbial dysbiosis and immune dysregulation. The identification of distinct endophenotypes have refined our understanding of its pathogenesis, including its heterogeneous disease mechanisms that influence treatment and prognosis responses. Next-generation sequencing (NGS) has revolutionised the way we view airway microbiology, allowing insights into the "unculturable". Understanding the bronchiectasis microbiome through targeted amplicon sequencing and/or shotgun metagenomics has provided key information on the interplay of the microbiome and host immunity, a central feature of disease progression. The rapid increase in translational and clinical studies in bronchiectasis now provides scope for the application of precision medicine and a better understanding of the efficacy of interventions aimed at restoring microbial balance and/or modulating immune responses. Holistic integration of these insights is driving an evolving paradigm shift in our understanding of bronchiectasis, which includes the critical role of the microbiome and its unique interplay with clinical, inflammatory, immunological and metabolic factors. Here, we review the current state of infection and the microbiome in bronchiectasis and provide views on the future directions in this field.
Collapse
Affiliation(s)
- Micheál Mac Aogáin
- Biochemical Genetics Laboratory, Department of Biochemistry, St. James's Hospital, Dublin, Ireland
- Clinical Biochemistry Unit, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Alison J Dicker
- Respiratory Research Group, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Pontus Mertsch
- Department of Medicine V, LMU University Hospital, LMU Munich, Comprehensive Pneumology Center (CPC), Member of the German Center of Lung Research (DZL), Munich, Germany
| | - Sanjay H Chotirmall
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Department of Respiratory and Critical Care Medicine, Tan Tock Seng Hospital, Singapore, Singapore
| |
Collapse
|
29
|
Zhang Y, Cao C, Li C, Witt RG, Huang H, Tsung A, Zhang H. Physical exercise in liver diseases. Hepatology 2024:01515467-990000000-00900. [PMID: 38836646 DOI: 10.1097/hep.0000000000000941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 05/14/2024] [Indexed: 06/06/2024]
Abstract
Liver diseases contribute to ~2 million deaths each year and account for 4% of all deaths globally. Despite various treatment options, the management of liver diseases remains challenging. Physical exercise is a promising nonpharmacological approach to maintain and restore homeostasis and effectively prevent and mitigate liver diseases. In this review, we delve into the mechanisms of physical exercise in preventing and treating liver diseases, highlighting its effects on improving insulin sensitivity, regulating lipid homeostasis, and modulating immune function. In addition, we evaluate the impact of physical exercise on various liver diseases, including liver ischemia/reperfusion injury, cardiogenic liver disease, metabolic dysfunction-associated steatotic liver disease, portal hypertension, cirrhosis, and liver cancer. In conclusion, the review underscores the effectiveness of physical exercise as a beneficial intervention in combating liver diseases.
Collapse
Affiliation(s)
- Yunwei Zhang
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA
| | - Chunyan Cao
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA
| | - Chaofan Li
- Department of Medicine, Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, Virginia, USA
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Russell G Witt
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA
| | - Hai Huang
- Division of Hepatology, Center for Immunology and Inflammation, Departments of Molecular Medicine, Medicine, and Surgery at the School of Medicine, Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Allan Tsung
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA
| | - Hongji Zhang
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
30
|
Tan J, Yang R, Xiao L, Xia Y, Qin W. Personalized decision support system for tailoring IgA nephropathy treatment strategies. Eur J Intern Med 2024; 124:69-77. [PMID: 38443263 DOI: 10.1016/j.ejim.2024.02.014] [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] [Received: 11/29/2023] [Revised: 01/06/2024] [Accepted: 02/04/2024] [Indexed: 03/07/2024]
Abstract
BACKGROUND The ongoing debate surrounding the use of immunosuppressive treatments for IgA nephropathy (IgAN) underscores the demand for personalized and effective strategies. METHODS Analyzed data from 807 IgAN patients over 5+ years using three methods: Random Forest with molecular biomarkers, network biomarkers with graph engineering, and an auto-encoder model. All models were trained using identical demographic, clinical, and pathological data, employing an 80-20 split for training and testing purposes. RESULTS In the comprehensive assessment of IgAN prognosis, the Random Forest model, employing molecular biomarkers, demonstrated strong performance metrics (AUC = 0.83, sensitivity = 0.51, specificity = 0.96). However, traditional graph feature engineering on patient-specific networks outperformed these results with an AUC of 0.90, sensitivity of 0.64, and specificity of 0.94. The Auto-encoder model showed the best accuracy (AUC = 0.91, sensitivity = 0.46, specificity = 0.96). The findings highlighted the superior predictive capabilities of network biomarkers over molecular biomarkers for adverse renal outcome prediction in IgAN. Consequently, we integrated Auto-encoder-derived Network Biomarkers with Random Forest Models to enhance prognostic precision in diverse IgAN treatment scenarios. The prediction for the prognosis of patients receiving supportive care, glucocorticoid therapy, and immunosuppressant treatment yielded AUC values of 0.95, 0.96, and 1, respectively, indicating high specificity. Drawing from these insights, we pioneered the development of an innovative decision support model for IgAN treatment. This model demonstrated the ability to make medical decisions comparable to those by experienced nephrologists, enabling the customization of personalized disease management strategies. CONCLUSION Our system accurately predicted IgAN prognosis and evaluated various treatment efficacies, aiding physicians in devising optimal therapeutic strategies for patients.
Collapse
Affiliation(s)
- Jiaxing Tan
- Division of Nephrology, Department of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Rongxin Yang
- College of Computer Science, Sichuan University, Chengdu, Sichuan, China
| | - Liyin Xiao
- College of Computer Science, Sichuan University, Chengdu, Sichuan, China
| | - Yuanlin Xia
- School of Mechanical Engineering, Sichuan University College of Computer Science, Sichuan University, Chengdu, China
| | - Wei Qin
- Division of Nephrology, Department of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
31
|
Qi Z, LE S, Cheng R, DU X, Zhao C, Zhang Z, Zhang X, Feng L, Schumann M, Mao L, Cheng S. Responses of the Serum Lipid Profile to Exercise and Diet Interventions in Nonalcoholic Fatty Liver Disease. Med Sci Sports Exerc 2024; 56:1036-1045. [PMID: 38247038 DOI: 10.1249/mss.0000000000003388] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2024]
Abstract
BACKGROUND This study aimed to assess the response patterns of circulating lipids to exercise and diet interventions in nonalcoholic fatty liver disease (NAFLD). METHODS The 8.6-month four-arm randomized controlled study comprised 115 NAFLD patients with prediabetes who were assigned to aerobic exercise (AEx; n = 29), low-carbohydrate diet (Diet; n = 28), AEx plus low-carbohydrate diet (AED; n = 29), and nonintervention (NI, n = 29) groups. Hepatic fat content (HFC) was quantified by proton magnetic resonance spectroscopy. Serum lipidomic analytes were measured using liquid chromatography-mass spectrometry. RESULTS After intervention, the total level of phosphatidylcholine (PC) increased significantly in the AEx group ( P = 0.043), whereas phosphatidylethanolamine (PE) and triacylglycerol decreased significantly in the AED group ( P = 0.046 and P = 0.036, respectively), and phosphatidylserine decreased in the NI group ( P = 0.002). Changes of 21 lipid metabolites were significantly associated with changes of HFC, among which half belonged to PC. Most of the molecules related to insulin sensitivity belonged to sphingomyelin (40 of 79). Controlling for the change of visceral fat, the significant associations between lipid metabolites and HFC remained. In addition, baseline serum lipids could predict the response of HFC to exercise and/or diet interventions (PE15:0/18:0 for AED, area under the curve (AUC) = 0.97; PE22:6(4Z,7Z,10Z,13Z,16Z,19Z)/0:0 for AEx, AUC = 0.90; and PC14:1(9Z)/19:1(9Z) for Diet, AUC = 0.92). CONCLUSIONS Changes of lipidome after exercise and/or diet interventions were associated with HFC reductions, which are independent of visceral fat reduction, particularly in metabolites belonging to PC. Importantly, baseline PE could predict the HFC response to exercise, and PC predicted the response to diet. These results indicate that a circulating metabolomics panel can be used to facilitate clinical implementation of lifestyle interventions for NAFLD management.
Collapse
Affiliation(s)
- Zhen Qi
- Physical Education Department, Shanghai Jiao Tong University, Shanghai, CHINA
| | | | - Runtan Cheng
- Exercise Translational Medicine Center, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, CHINA
| | - Xiaming DU
- Department of Orthopedic, Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, CHINA
| | - Can Zhao
- School of Athletic Performance, Shanghai University of Sport, Shanghai, CHINA
| | - Zhengyun Zhang
- Department of Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, CHINA
| | - Xiaobo Zhang
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital of Wenzhou Medical University, Wenzhou, CHINA
| | - Lei Feng
- Instrumental Analysis Center, Shanghai Jiao Tong University, Shanghai, CHINA
| | | | - Lijuan Mao
- School of Physical Education and Sport Training, Shanghai University of Sport, Shanghai 200438, CHINA
| | | |
Collapse
|
32
|
Dallio M, Sangineto M, Romeo M, Cipullo M, Coppola A, Mammone S, Di Gioia G, Masarone M, Persico M, Serviddio G, Federico A. The influence of acute lifestyle changes on NAFLD evolution in a multicentre cohort: a matter of body composition. Nutr Diabetes 2024; 14:33. [PMID: 38802382 PMCID: PMC11130147 DOI: 10.1038/s41387-024-00294-2] [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] [Received: 11/13/2023] [Revised: 05/14/2024] [Accepted: 05/15/2024] [Indexed: 05/29/2024] Open
Abstract
BACKGROUND Unhealthy lifestyles represent a key element fueling Non-alcoholic fatty liver disease (NAFLD) onset and worsening. We aimed to evaluate the effects of forced acute lifestyle changes on NAFLD evolution. METHODS 187 NAFLD patients were followed two years pre- and two years during the lockdown social restrictions in three Italian medical centers. For each patient, biochemical, clinical, non-invasive liver fibrosis, nutritional, and body composition data were collected. RESULTS An increase in fats and carbohydrate intake associated with impaired weekly physical activity during the lockdown was demonstrated as well as an increase in body mass index and waist-hip-ratio (p < 0.0001 for all). Total cholesterol, low-density lipoprotein, high-density lipoprotein, triglycerides, glucose, insulin, homeostatic model assessment for insulin resistance, and transaminases worsened during the lockdown (glucose: p = 0.0007; p < 0.0001 for the others). Moreover, NAFLD fibrosis score, liver stiffness, and controlled attenuation parameter were also impaired during the same period (p < 0.0001 for all). The bioelectrical impedance analysis (BIA) evidenced an increase of fat mass (FM), and a reduction of free fat mass (FFM) and body cell mass (BCM) (p < 0.0001 for all). The lockdown overall hepatocellular carcinoma (HCC) and Milan-out HCC occurrence revealed Hazard Ratio (HR): 2.398, 95% Confidence Interval (CI):1.16-5, p = 0.02, and HR:5.931, CI:2-17.6, p = 0.008 respectively. A liver disease stage and comorbidities independent association between both the assessed outcomes and body composition analysis in terms of mean values and variation (T1-T2 Δ) was demonstrated. CONCLUSIONS The acute lifestyle changes impacted NAFLD evolution via body composition modifications negatively influencing the HCC occurrence.
Collapse
Affiliation(s)
- Marcello Dallio
- Hepatogastroenterology Division, Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Moris Sangineto
- University Center for Research and Treatment of Liver Diseases (C.U.R.E.), Liver Unit, University of Foggia, Foggia, Italy
| | - Mario Romeo
- Hepatogastroenterology Division, Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy.
| | - Marina Cipullo
- Hepatogastroenterology Division, Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Annachiara Coppola
- Hepatogastroenterology Division, Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Simone Mammone
- Hepatogastroenterology Division, Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Giuseppe Di Gioia
- University Center for Research and Treatment of Liver Diseases (C.U.R.E.), Liver Unit, University of Foggia, Foggia, Italy
| | - Mario Masarone
- Department of Medicine and Surgery, "Scuola Medica Salernitana", Internal Medicine and Hepatology Unit, University of Salerno, Salerno, Italy
| | - Marcello Persico
- Department of Medicine and Surgery, "Scuola Medica Salernitana", Internal Medicine and Hepatology Unit, University of Salerno, Salerno, Italy
| | - Gaetano Serviddio
- University Center for Research and Treatment of Liver Diseases (C.U.R.E.), Liver Unit, University of Foggia, Foggia, Italy
| | - Alessandro Federico
- Hepatogastroenterology Division, Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| |
Collapse
|
33
|
Min L, Ablitip A, Wang R, Luciana T, Wei M, Ma X. Effects of Exercise on Gut Microbiota of Adults: A Systematic Review and Meta-Analysis. Nutrients 2024; 16:1070. [PMID: 38613103 PMCID: PMC11013040 DOI: 10.3390/nu16071070] [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: 02/23/2024] [Revised: 03/25/2024] [Accepted: 04/01/2024] [Indexed: 04/14/2024] Open
Abstract
BACKGROUND The equilibrium between gut microbiota (GM) and the host plays a pivotal role in maintaining overall health, influencing various physiological and metabolic functions. Emerging research suggests that exercise modulates the abundance and functionality of gut bacteria, yet the comprehensive effects on GM diversity remain to be synthesized. OBJECTIVES AND DESIGN The study aims to quantitatively examine the effect of exercise on the diversity of gut microbiota of adults using a systemic review and meta-analysis approach. METHODS PubMed, Ebsco, Embase, Web of Science, Cochrane Central Register of Controlled Trials, the China National Knowledge Infrastructure, and Wanfang Data were searched from their inception to September 2023. Exercise intervention studies with a control group that describe and compare the composition of GM in adults, using 16S rRNA gene sequencing, were included in this meta-analysis. RESULTS A total of 25 studies were included in this meta-analysis with a total of 1044 participants. Based on a fixed-effects model [Chi2 = 29.40, df = 20 (p = 0.08); I2 = 32%], the pooled analysis showed that compared with the control group, exercise intervention can significantly increase the alpha diversity of adult GM, using the Shannon index as an example [WMD = 0.05, 95% CI (0.00, 0.09); Z = 1.99 (p = 0.05)]. In addition, exercise interventions were found to significantly alter GM, notably decreasing Bacteroidetes and increasing Firmicutes, indicating a shift in the Firmicutes/Bacteroidetes ratio. The subgroup analysis indicates that females and older adults appear to exhibit more significant changes in the Shannon Index and observed OTUs. CONCLUSIONS Exercise may be a promising way to improve GM in adults. In particular, the Shannon index was significantly increased after exercise. Distinct responses in GM diversity to exercise interventions based on gender and age implicated that more research was needed.
Collapse
Affiliation(s)
- Leizi Min
- Division of Sports Science and Physical Education, Tsinghua University, Beijing 100084, China; (L.M.); (A.A.)
| | - Alimjan Ablitip
- Division of Sports Science and Physical Education, Tsinghua University, Beijing 100084, China; (L.M.); (A.A.)
| | - Rui Wang
- Division of Sports Science and Physical Education, Tsinghua University, Beijing 100084, China; (L.M.); (A.A.)
| | - Torquati Luciana
- Department of Public Health and Sport Sciences, Medical School, University of Exeter, Exeter EX1 2HZ, UK;
| | - Mengxian Wei
- Division of Sports Science and Physical Education, Tsinghua University, Beijing 100084, China; (L.M.); (A.A.)
| | - Xindong Ma
- Division of Sports Science and Physical Education, Tsinghua University, Beijing 100084, China; (L.M.); (A.A.)
| |
Collapse
|
34
|
Wu X, Kong L, Feng Y, Zheng R, Zhou J, Sun J, Liu S. Communication mediated interaction between bacteria and microalgae advances photogranulation. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 914:169975. [PMID: 38218496 DOI: 10.1016/j.scitotenv.2024.169975] [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: 10/21/2023] [Revised: 12/16/2023] [Accepted: 01/04/2024] [Indexed: 01/15/2024]
Abstract
Recently, photogranules composed of bacteria and microalgae for carbon-negative nitrogen removal receive extensive attention worldwide, yet which type of bacteria is helpful for rapid formation of photogranules and whether they depend on signaling communication remain elusive. Varied signaling communication was analyzed using metagenomic method among bacteria and microalgae in via of two types of experimentally verified signaling molecule from bacteria to microalgae, which include indole-3-acetic acid (IAA) and N-acyl homoserine lactones (AHLs) during the operation of photo-bioreactors. Signaling communication is helpful for the adaptability of bacteria to survive with algae. Compared with non-signaling bacteria, signaling bacteria more easily adapt to the varied conditions, evidenced by the increased abundance in the operated reactors. Signaling bacteria are easier to enter the phycosphere, and they dominate the interactions between bacteria and algae rather than non-signaling bacteria. The co-abundance groups (CAGs) with signaling bacteria possess higher abundance than that without signaling bacteria (22.27 % and 6.67 %). Importantly, signaling bacteria accessibly interact with microalgae, which possess higher degree centralities and 32.50 % of them are keystone nodes in the network, in contrast to only 18.66 % of non-signaling bacteria. Thauera carrying both IAA and AHLs synthase genes are highly enriched and positively correlated with nitrogen removal rate. Our work not only highlights the essential roles of signaling communication between microalgae and bacteria in the development of photogranules, but also enriches our understanding of microbial sociobiology.
Collapse
Affiliation(s)
- Xiaogang Wu
- College of Environmental Sciences and Engineering, Peking University, Beijing 100871, China; Key Laboratory of Water and Sediment Sciences, Ministry of Education, Peking University, Beijing 100871, China
| | - Lingrui Kong
- College of Environmental Sciences and Engineering, Peking University, Beijing 100871, China; Key Laboratory of Water and Sediment Sciences, Ministry of Education, Peking University, Beijing 100871, China
| | - Yiming Feng
- College of Environmental Sciences and Engineering, Peking University, Beijing 100871, China; Key Laboratory of Water and Sediment Sciences, Ministry of Education, Peking University, Beijing 100871, China
| | - Ru Zheng
- College of Environmental Sciences and Engineering, Peking University, Beijing 100871, China; Key Laboratory of Water and Sediment Sciences, Ministry of Education, Peking University, Beijing 100871, China
| | - Jianhang Zhou
- College of Environmental Sciences and Engineering, Peking University, Beijing 100871, China; Key Laboratory of Water and Sediment Sciences, Ministry of Education, Peking University, Beijing 100871, China
| | - Jingqi Sun
- College of Environmental Sciences and Engineering, Peking University, Beijing 100871, China; Key Laboratory of Water and Sediment Sciences, Ministry of Education, Peking University, Beijing 100871, China
| | - Sitong Liu
- College of Environmental Sciences and Engineering, Peking University, Beijing 100871, China; Key Laboratory of Water and Sediment Sciences, Ministry of Education, Peking University, Beijing 100871, China.
| |
Collapse
|
35
|
Jin L, Diaz-Canestro C, Wang Y, Tse MA, Xu A. Exerkines and cardiometabolic benefits of exercise: from bench to clinic. EMBO Mol Med 2024; 16:432-444. [PMID: 38321233 PMCID: PMC10940599 DOI: 10.1038/s44321-024-00027-z] [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: 10/13/2023] [Revised: 01/19/2024] [Accepted: 01/22/2024] [Indexed: 02/08/2024] Open
Abstract
Regular exercise has both immediate and long-lasting benefits on cardiometabolic health, and has been recommended as a cornerstone of treatment in the management of diabetes and cardiovascular conditions. Exerkines, which are defined as humoral factors responsive to acute or chronic exercise, have emerged as important players conferring some of the multiple cardiometabolic benefits of exercise. Over the past decades, hundreds of exerkines released from skeletal muscle, heart, liver, adipose tissue, brain, and gut have been identified, and several exerkines (such as FGF21, IL-6, and adiponectin) have been exploited therapeutically as exercise mimetics for the treatment of various metabolic and cardiovascular diseases. Recent advances in metagenomics have led to the identification of gut microbiota, a so-called "hidden" metabolic organ, as an additional class of exerkines determining the efficacy of exercise in diabetes prevention, cardiac protection, and exercise performance. Furthermore, multiomics-based studies have shown the feasibility of using baseline exerkine signatures to predict individual responses to exercise with respect to metabolic and cardiorespiratory health. This review aims to explore the molecular pathways whereby exerkine networks mediate the cardiometabolic adaptations to exercise by fine-tuning inter-organ crosstalk, and discuss the roadmaps for translating exerkine-based discovery into the therapeutic application and personalized medicine in the management of the cardiometabolic disease.
Collapse
Affiliation(s)
- Leigang Jin
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
- Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Candela Diaz-Canestro
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
- Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yu Wang
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China
| | - Michael Andrew Tse
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
- Centre for Sports and Exercise, The University of Hong Kong, Hong Kong, China
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China.
- Department of Medicine, The University of Hong Kong, Hong Kong, China.
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
36
|
Cullen JMA, Shahzad S, Kanaley JA, Ericsson AC, Dhillon J. The effects of 6 wk of resistance training on the gut microbiome and cardiometabolic health in young adults with overweight and obesity. J Appl Physiol (1985) 2024; 136:349-361. [PMID: 38059291 DOI: 10.1152/japplphysiol.00350.2023] [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: 06/08/2023] [Revised: 11/21/2023] [Accepted: 11/21/2023] [Indexed: 12/08/2023] Open
Abstract
Obesity is a known risk factor for the development of insulin resistance and other cardiometabolic disorders. Recently, the gut microbiome has been associated with obesity and subsequent health complications. Exercise has been regularly utilized as a therapeutic intervention to treat obesity and its associated comorbidities. This study examined the effects of a 6-wk resistance training exercise program (RT) on the diversity, composition, and metabolic pathways of the gut microbiome. Sedentary young adults (age 18-35 yr) with overweight and obesity (BMI 25-45 kg/m2) were recruited to participate in this randomized controlled trial. Participants were randomized to RT (n = 16), a 6-wk resistance training program (3 days/wk), or control (CT) (n = 16), a nonexercising control. Main outcomes of the study included gut microbiome measures (taxa abundances, diversity, and predicted function) and cardiometabolic outcomes [blood pressure (BP) and glucoregulation]. Increased abundances of Roseburia, a short-chain fatty acid (SCFA) producer were observed over 6 wk (W6) with RT compared with CT (group × week, P < 0.05, q < 0.25). RT also induced marginal alterations in predicted microbial metabolic and cell motility pathways compared with CT (group × week, P < 0.05, q < 0.25). However, RT did not significantly impact overall microbial diversity. Furthermore, RT resulted in higher quantitative insulin-sensitivity check index (QUICKI) and lower diastolic BP at W6 compared with CT [baseline (BL)-adjusted P < 0.05]. RT had mixed effects on the gut microbiome. Although RT increased abundances of Roseburia and induced minor changes in microbial pathways, it is important to consider these changes in the context of the overall stability observed in the microbiome composition.NEW & NOTEWORTHY Resistance training induces mixed changes in the gut microbiome, including an increase in the abundances of the Roseburia genus and minor alterations in microbial pathways. However, it is vital to interpret these changes in light of the broader context, where we observe stability in the overall microbiome composition. This stability may be attributed to the microbiome's resilience, demonstrating its capacity to withstand short-term physiological stressors.
Collapse
Affiliation(s)
- John M A Cullen
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri, United States
| | - Shahim Shahzad
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri, United States
| | - Jill A Kanaley
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri, United States
| | - Aaron C Ericsson
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri, United States
| | - Jaapna Dhillon
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri, United States
| |
Collapse
|
37
|
Theys C, Vanderhaeghen T, Van Dijck E, Peleman C, Scheepers A, Ibrahim J, Mateiu L, Timmermans S, Vanden Berghe T, Francque SM, Van Hul W, Libert C, Vanden Berghe W. Loss of PPARα function promotes epigenetic dysregulation of lipid homeostasis driving ferroptosis and pyroptosis lipotoxicity in metabolic dysfunction associated Steatotic liver disease (MASLD). FRONTIERS IN MOLECULAR MEDICINE 2024; 3:1283170. [PMID: 39086681 PMCID: PMC11285560 DOI: 10.3389/fmmed.2023.1283170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 12/14/2023] [Indexed: 08/02/2024]
Abstract
Metabolic Dysfunction Associated Steatotic Liver Disease (MASLD) is a growing epidemic with an estimated prevalence of 20%-30% in Europe and the most common cause of chronic liver disease worldwide. The onset and progression of MASLD are orchestrated by an interplay of the metabolic environment with genetic and epigenetic factors. Emerging evidence suggests altered DNA methylation pattern as a major determinant of MASLD pathogenesis coinciding with progressive DNA hypermethylation and gene silencing of the liver-specific nuclear receptor PPARα, a key regulator of lipid metabolism. To investigate how PPARα loss of function contributes to epigenetic dysregulation in MASLD pathology, we studied DNA methylation changes in liver biopsies of WT and hepatocyte-specific PPARα KO mice, following a 6-week CDAHFD (choline-deficient, L-amino acid-defined, high-fat diet) or chow diet. Interestingly, genetic loss of PPARα function in hepatocyte-specific KO mice could be phenocopied by a 6-week CDAHFD diet in WT mice which promotes epigenetic silencing of PPARα function via DNA hypermethylation, similar to MASLD pathology. Remarkably, genetic and lipid diet-induced loss of PPARα function triggers compensatory activation of multiple lipid sensing transcription factors and epigenetic writer-eraser-reader proteins, which promotes the epigenetic transition from lipid metabolic stress towards ferroptosis and pyroptosis lipid hepatoxicity pathways associated with advanced MASLD. In conclusion, we show that PPARα function is essential to support lipid homeostasis and to suppress the epigenetic progression of ferroptosis-pyroptosis lipid damage associated pathways towards MASLD fibrosis.
Collapse
Affiliation(s)
- Claudia Theys
- Protein Chemistry, Proteomics and Epigenetic Signaling (PPES), Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Tineke Vanderhaeghen
- Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | | | - Cedric Peleman
- Laboratory of Experimental Medicine and Pediatrics, Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
- Pathophysiology Lab, Infla-Med Centre of Excellence, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Anne Scheepers
- Center of Medical Genetics, University of Antwerp, Antwerp, Belgium
| | - Joe Ibrahim
- Center of Medical Genetics, University of Antwerp, Antwerp, Belgium
| | - Ligia Mateiu
- Center of Medical Genetics, University of Antwerp, Antwerp, Belgium
| | - Steven Timmermans
- Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Tom Vanden Berghe
- Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Pathophysiology Lab, Infla-Med Centre of Excellence, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Sven M. Francque
- Laboratory of Experimental Medicine and Pediatrics, Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
- Department of Gastroenterology and Hepatology, Antwerp University Hospital, Edegem, Belgium
| | - Wim Van Hul
- Center of Medical Genetics, University of Antwerp, Antwerp, Belgium
| | - Claude Libert
- Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Wim Vanden Berghe
- Protein Chemistry, Proteomics and Epigenetic Signaling (PPES), Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
38
|
Qian H, Zuo Y, Wen S, Wang X, Liu Y, Li T. Impact of exercise training on gut microbiome imbalance in obese individuals: a study based on Mendelian randomization analysis. Front Physiol 2024; 14:1264931. [PMID: 38235382 PMCID: PMC10792044 DOI: 10.3389/fphys.2023.1264931] [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: 07/21/2023] [Accepted: 12/14/2023] [Indexed: 01/19/2024] Open
Abstract
Objective: The aim of this study was to investigate the relationship between exercise and gut Microbiome and to assess its possible causality. Methods: Using Mendelian randomization (MR) research methods, we collected genetic data from different populations, including genetic variants associated with relative abundance or presence of microbial taxa as instrumental variables. At the same time, we extracted results related to obesity and gut Microbiome from existing relevant studies and used inverse variance weighting (IVW), weighted median, and MR-Egger regression to assess the causal relationship between obesity and gut Microbiome. We plotted forest plots and scatter plots of the association between obesity and gut Microbiome. Results: Gut Microbiome was positively associated with obesity, and four bacterial genera (Akkermansia, RuminococcaceaeUCG011, Holdemania, and Intestinimonas) were associated with obesity according to inverse variance-weighted estimation in at least one MR method. Inverse variance weighted estimation showed that obesity was associated with obesity in Akkermansia (OR = 0.810, 95% CI 0.608-1.079, p = 0.04), RuminococcaceaeUCG011 (OR = 1.238, 95% CI 0. 511-2.999, p = 0.04), Holdemania Intestinimonas (OR = 1.214, 95% CI 1.002-1.470, p = 0.03), and Intestinimonas (OR = 0.747, 95% CI 0.514-1.086, p = 0.01) had a relevant effect. Obesity decreased the abundance of Akkermansia, Intestinimonas microbiome and increased the abundance of RuminococcaceaeUCG011, Holdemania microbiome. Conclusion: The results of this study, conducted using a two-sample Mendelian randomization method, suggest a causal relationship between obesity and intestinal microbiome. Obesity decreased the abundance of Akkermansia, Intestinimonas microbiome and increased the abundance of RuminococcaceaeUCG011, Holdemania microbiome. More randomized controlled trials are necessary to elucidate the protective effects of exercise on gut Microbiome and its unique protective mechanisms.
Collapse
Affiliation(s)
- Haonan Qian
- Department of Physical Education, Hanyang University, Seoul, Republic of Korea
| | - Yuxin Zuo
- Department of Health and Physical Education, The Education University of Hong Kong, Tai Po, Hong Kong SAR, China
| | - Shixiong Wen
- Department of Physical Education, Hanyang University, Seoul, Republic of Korea
| | - Xilong Wang
- Department of Physical Education, Hanyang University, Seoul, Republic of Korea
| | - Yaowen Liu
- Department of Physical Education, Hanyang University, Seoul, Republic of Korea
| | - Tianwei Li
- The University of Edinburgh, Physical Activity for Health Research Center, Edinburgh, United Kingdom
| |
Collapse
|
39
|
Gao C, Wei J, Lu C, Wang L, Dong D, Sun M. A new perspective in intestinal microecology: lifting the veil of exercise regulation of cardiometabolic diseases. Gut Microbes 2024; 16:2404141. [PMID: 39305272 PMCID: PMC11418258 DOI: 10.1080/19490976.2024.2404141] [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] [Received: 03/21/2024] [Revised: 09/02/2024] [Accepted: 09/09/2024] [Indexed: 09/25/2024] Open
Abstract
Cardiometabolic diseases (CMDs), encompassing cardiovascular and metabolic dysfunctions, characterized by insulin resistance, dyslipidemia, hepatic steatosis, and inflammation, have been identified with boosting morbidity and mortality due to the dearth of efficacious therapeutic interventions. In recent years, studies have shown that variations in gut microbiota and its own metabolites can influence the occurrence of CMDs. Intriguingly, the composition and function of the gut microbiota are susceptible to exercise patterns, thus affecting inflammatory, immune, and metabolic responses within the host. In this review, we introduce the key mechanisms of intestinal microecology involved in the onset and development of CMDs, discuss the relationship between exercise and intestinal microecology, and then analyze the role of intestinal microecology in the beneficial effects of exercise on CMDs, aiming at elucidating the gut-heart axis mechanisms of exercise mediated protective effect on CMDs, building avenues for the application of exercise in the management of CMDs.
Collapse
Affiliation(s)
- Can Gao
- College of Exercise and Health, Shenyang Sport University, Shenyang, Liaoning, P. R. China
| | - Jinwen Wei
- College of Exercise and Health, Shenyang Sport University, Shenyang, Liaoning, P. R. China
| | - Changxu Lu
- College of Exercise and Health, Shenyang Sport University, Shenyang, Liaoning, P. R. China
| | - Lijie Wang
- Department of Cardiology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, P. R. China
| | - Dan Dong
- College of Basic Medical Science, China Medical University, Shenyang, Liaoning, P. R. China
| | - Mingli Sun
- College of Exercise and Health, Shenyang Sport University, Shenyang, Liaoning, P. R. China
| |
Collapse
|
40
|
Effenberger M, Grander C, Grabherr F, Tilg H. Nonalcoholic Fatty Liver Disease and the Intestinal Microbiome: An Inseparable Link. J Clin Transl Hepatol 2023; 11:1498-1507. [PMID: 38161503 PMCID: PMC10752805 DOI: 10.14218/jcth.2023.00069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 04/21/2023] [Accepted: 07/18/2023] [Indexed: 01/03/2024] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) particularly affects patients with type 2 diabetes and obesity. The incidence of NAFLD has increased significantly over the last decades and is now pandemically across the globe. It is a complex systemic disease comprising hepatic lipid accumulation, inflammation, lipotoxicity, gut dysbiosis, and insulin resistance as main features and with the potential to progress to cirrhosis and hepatocellular carcinoma (HCC). In numerous animal and human studies the gut microbiota plays a key role in the pathogenesis of NAFLD, NAFLD-cirrhosis and NAFLD-associated HCC. Lipotoxicity is the driver of inflammation, insulin resistance, and liver injury. Likewise, western diet, obesity, and metabolic disorders may alter the gut microbiota, which activates innate and adaptive immune responses and fuels hereby hepatic and systemic inflammation. Indigestible carbohydrates are fermented by the gut microbiota to produce important metabolites, such as short-chain fatty acids and succinate. Numerous animal and human studies suggested a pivotal role of these metabolites in the progression of NAFLD and its comorbidities. Though, modification of the gut microbiota and/or the metabolites could even be beneficial in patients with NAFLD, NAFLD-cirrhosis, and NAFLD-associated HCC. In this review we collect the evidence that exogenous and endogenous hits drive liver injury in NAFLD and propel liver fibrosis and the progressing to advanced disease stages. NAFLD can be seen as the product of a complex interplay between gut microbiota, the immune response and metabolism. Thus, the challenge will be to understand its pathogenesis and to develop new therapeutic strategies.
Collapse
Affiliation(s)
- Maria Effenberger
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Christoph Grander
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Felix Grabherr
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
41
|
Cullen JMA, Shahzad S, Dhillon J. A systematic review on the effects of exercise on gut microbial diversity, taxonomic composition, and microbial metabolites: identifying research gaps and future directions. Front Physiol 2023; 14:1292673. [PMID: 38187136 PMCID: PMC10770260 DOI: 10.3389/fphys.2023.1292673] [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: 09/11/2023] [Accepted: 11/07/2023] [Indexed: 01/09/2024] Open
Abstract
The gut microbiome, hosting a diverse microbial community, plays a pivotal role in metabolism, immunity, and digestion. While the potential of exercise to influence this microbiome has been increasingly recognized, findings remain incongruous. This systematic review examined the effects of exercise on the gut microbiome of human and animal models. Databases (i.e., PubMed, Cochrane Library, Scopus, and Web of Science) were searched up to June 2022. Thirty-two exercise studies, i.e., 19 human studies, and 13 animal studies with a minimum of two groups that discussed microbiome outcomes, such as diversity, taxonomic composition, or microbial metabolites, over the intervention period, were included in the systematic review (PROSPERO registration numbers for human review: CRD42023394223). Results indicated that over 50% of studies found no significant exercise effect on human microbial diversity. When evident, exercise often augmented the Shannon index, reflecting enhanced microbial richness and evenness, irrespective of disease status. Changes in beta-diversity metrics were also documented with exercise but without clear directionality. A larger percentage of animal studies demonstrated shifts in diversity compared to human studies, but without any distinct patterns, mainly due to the varied effects of predominantly aerobic exercise on diversity metrics. In terms of taxonomic composition, in humans, exercise usually led to a decrease in the Firmicutes/Bacteroidetes ratio, and consistent increases with Bacteroides and Roseburia genera. In animal models, Coprococcus, another short chain fatty acid (SCFA) producer, consistently rose with exercise. Generally, SCFA producers were found to increase with exercise in animal models. With regard to metabolites, SCFAs emerged as the most frequently measured metabolite. However, due to limited human and animal studies examining exercise effects on microbial-produced metabolites, including SCFAs, clear patterns did not emerge. The overall risk of bias was deemed neutral. In conclusion, this comprehensive systematic review underscores that exercise can potentially impact the gut microbiome with indications of changes in taxonomic composition. The significant variability in study designs and intervention protocols demands more standardized methodologies and robust statistical models. A nuanced understanding of the exercise-microbiome relationship could guide individualized exercise programs to optimize health. Systematic Review Registration: https://www.crd.york.ac.uk/prospero/display_record.php?RecordID=394223, identifier CRD42023394223.
Collapse
Affiliation(s)
- John M A Cullen
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
| | - Shahim Shahzad
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
| | - Jaapna Dhillon
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
| |
Collapse
|
42
|
Jia X, Chen Q, Wu H, Liu H, Jing C, Gong A, Zhang Y. Exploring a novel therapeutic strategy: the interplay between gut microbiota and high-fat diet in the pathogenesis of metabolic disorders. Front Nutr 2023; 10:1291853. [PMID: 38192650 PMCID: PMC10773723 DOI: 10.3389/fnut.2023.1291853] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 11/27/2023] [Indexed: 01/10/2024] Open
Abstract
In the past two decades, the rapid increase in the incidence of metabolic diseases, including obesity, diabetes, dyslipidemia, non-alcoholic fatty liver disease, hypertension, and hyperuricemia, has been attributed to high-fat diets (HFD) and decreased physical activity levels. Although the phenotypes and pathologies of these metabolic diseases vary, patients with these diseases exhibit disease-specific alterations in the composition and function of their gut microbiota. Studies in germ-free mice have shown that both HFD and gut microbiota can promote the development of metabolic diseases, and HFD can disrupt the balance of gut microbiota. Therefore, investigating the interaction between gut microbiota and HFD in the pathogenesis of metabolic diseases is crucial for identifying novel therapeutic strategies for these diseases. This review takes HFD as the starting point, providing a detailed analysis of the pivotal role of HFD in the development of metabolic disorders. It comprehensively elucidates the impact of HFD on the balance of intestinal microbiota, analyzes the mechanisms underlying gut microbiota dysbiosis leading to metabolic disruptions, and explores the associated genetic factors. Finally, the potential of targeting the gut microbiota as a means to address metabolic disturbances induced by HFD is discussed. In summary, this review offers theoretical support and proposes new research avenues for investigating the role of nutrition-related factors in the pathogenesis of metabolic disorders in the organism.
Collapse
Affiliation(s)
- Xiaokang Jia
- School of Traditional Chinese Medicine, Hainan Medical University, Haikou, Hainan, China
| | - Qiliang Chen
- School of Basic Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Huiwen Wu
- School of Traditional Chinese Medicine, Hainan Medical University, Haikou, Hainan, China
| | - Hongbo Liu
- School of Traditional Chinese Medicine, Hainan Medical University, Haikou, Hainan, China
| | - Chunying Jing
- School of Traditional Chinese Medicine, Hainan Medical University, Haikou, Hainan, China
| | - Aimin Gong
- School of Traditional Chinese Medicine, Hainan Medical University, Haikou, Hainan, China
| | - Yuanyuan Zhang
- The Affiliated TCM Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
43
|
Ye X, Sun P, Lao S, Wen M, Zheng R, Lin Y, Gan L, Fan X, Wang P, Li Z, Yan X, Zhao L. Fgf21-Dubosiella axis mediates the protective effects of exercise against NAFLD development. Life Sci 2023; 334:122231. [PMID: 37935276 DOI: 10.1016/j.lfs.2023.122231] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 10/18/2023] [Accepted: 10/29/2023] [Indexed: 11/09/2023]
Abstract
AIM To explore the mechanism of gut microbiota mediates protective effects of exercise against non-alcoholic fatty liver disease (NAFLD) development. MAIN METHODS The male C57BL/6 mice were fed with high fat food (HFD) or normal diet (CON) respectively, and the obese mice were randomly divided into sedentariness (HFD) and exercise groups (HFD + Exe). The total intervention period was 18 weeks. Antibiotic treatment and fecal microbiota transplantation were applied to evaluate gut microbiota mediates the protective effects of exercise against NAFLD development. 16S rDNA profiling of gut microbiota and extracorporeal rehydration of Dubosiella newyorkensis were performed to identify the crucial role of Dubosiella in NAFLD improvement during exercise training. FGF21 knock-out mice were used to reveal the potential mechanism of exercise increased the abundance of Dubosiella. RT-PCR, Western blot, Histopathological examinations and Biochemical testing were performed to evaluate the lipid deposition and function in the liver. KEY FINDINGS Treadmill exercise significantly ameliorated hepatic function and mitigated lipid accumulation in NAFLD mice, and these hepatoprotective benefits were mostly mediated by the Dubosiella. In addition, the increased abundance of Dubosiella during exercise training was modulated by FGF21 specifically. SIGNIFICANCE In short, Dubosiella, chiefly regulated by FGF21 signaling during exercise training, has been discovered to govern the protective impacts of exercising counter to the development of NAFLD and exhibits a promising treatment target for NAFLD.
Collapse
Affiliation(s)
- Xiaochun Ye
- Department of Pharmacology, School of Basic Medical Sciences, Wenzhou Medical University, 325035 Wenzhou, Zhejiang, China; Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, 325035 Wenzhou, Zhejiang, China
| | - Peng Sun
- Department of Pharmacology, School of Basic Medical Sciences, Wenzhou Medical University, 325035 Wenzhou, Zhejiang, China; Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China
| | - Shuaiwei Lao
- Department of Pharmacology, School of Basic Medical Sciences, Wenzhou Medical University, 325035 Wenzhou, Zhejiang, China
| | - Meiyun Wen
- Department of Pharmacology, School of Basic Medical Sciences, Wenzhou Medical University, 325035 Wenzhou, Zhejiang, China
| | - Ruofang Zheng
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, 325035 Wenzhou, Zhejiang, China
| | - Yuanyuan Lin
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, 325035 Wenzhou, Zhejiang, China
| | - Lipeng Gan
- Department of Pharmacology, School of Basic Medical Sciences, Wenzhou Medical University, 325035 Wenzhou, Zhejiang, China; Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China
| | - Xia Fan
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Ping Wang
- Department of Pharmacology, School of Basic Medical Sciences, Wenzhou Medical University, 325035 Wenzhou, Zhejiang, China
| | - Zhiyong Li
- Department of Pharmacology, School of Basic Medical Sciences, Wenzhou Medical University, 325035 Wenzhou, Zhejiang, China; Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China.
| | - Xiaoqing Yan
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China.
| | - Longwei Zhao
- Department of Pharmacology, School of Basic Medical Sciences, Wenzhou Medical University, 325035 Wenzhou, Zhejiang, China; Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, 325035 Wenzhou, Zhejiang, China; School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China.
| |
Collapse
|
44
|
Fa P, Ke BG, Dupre A, Tsung A, Zhang H. The implication of neutrophil extracellular traps in nonalcoholic fatty liver disease. Front Immunol 2023; 14:1292679. [PMID: 38022519 PMCID: PMC10652891 DOI: 10.3389/fimmu.2023.1292679] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 10/20/2023] [Indexed: 12/01/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is an expanding worldwide health concern, and the underlying mechanisms contributing to its progression still need further exploration. Neutrophil extracellular traps (NETs) are intricate formations comprised of nuclear constituents and diverse antimicrobial granules that are released into the extracellular milieu by activated neutrophils upon various triggers, which play a pivotal part in the onset and advancement of NAFLD. NETs actively participate in the genesis of NAFLD by fostering oxidative stress and inflammation, ultimately resulting in hepatic fat accumulation and the escalation of liver injury. Recent insights into the interaction with other hepatic immune populations and mediators, such as macrophages and T regulatory cells, have revealed several important mechanisms that can trigger further liver injury. In conclusion, the formation of NETs emerged as an important factor in the development of NAFLD, offering a promising target for innovative therapeutic approaches against this debilitating condition. This comprehensive review seeks to compile existing studies exploring the involvement of NETs in the genesis of NAFLD and their influence on the immune response throughout the progression of NAFLD.
Collapse
Affiliation(s)
- Pengyan Fa
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, United States
| | - Benjamin G. Ke
- School of Medicine, University of Virginia, Charlottesville, VA, United States
| | - Abigail Dupre
- School of Medicine, University of Virginia, Charlottesville, VA, United States
| | - Allan Tsung
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, United States
| | - Hongji Zhang
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
45
|
Brunner JD, Gallegos-Graves LA, Kroeger ME. Inferring microbial interactions with their environment from genomic and metagenomic data. PLoS Comput Biol 2023; 19:e1011661. [PMID: 37956203 PMCID: PMC10681327 DOI: 10.1371/journal.pcbi.1011661] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 11/27/2023] [Accepted: 11/04/2023] [Indexed: 11/15/2023] Open
Abstract
Microbial communities assemble through a complex set of interactions between microbes and their environment, and the resulting metabolic impact on the host ecosystem can be profound. Microbial activity is known to impact human health, plant growth, water quality, and soil carbon storage which has lead to the development of many approaches and products meant to manipulate the microbiome. In order to understand, predict, and improve microbial community engineering, genome-scale modeling techniques have been developed to translate genomic data into inferred microbial dynamics. However, these techniques rely heavily on simulation to draw conclusions which may vary with unknown parameters or initial conditions, rather than more robust qualitative analysis. To better understand microbial community dynamics using genome-scale modeling, we provide a tool to investigate the network of interactions between microbes and environmental metabolites over time. Using our previously developed algorithm for simulating microbial communities from genome-scale metabolic models (GSMs), we infer the set of microbe-metabolite interactions within a microbial community in a particular environment. Because these interactions depend on the available environmental metabolites, we refer to the networks that we infer as metabolically contextualized, and so name our tool MetConSIN: Metabolically Contextualized Species Interaction Networks.
Collapse
Affiliation(s)
- James D. Brunner
- Biosciences Division, Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
- Center for Nonlinear Studies, Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
| | | | - Marie E. Kroeger
- Biosciences Division, Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
| |
Collapse
|
46
|
Donati Zeppa S, Natalucci V, Agostini D, Vallorani L, Amatori S, Sisti D, Rocchi MBL, Pazienza V, Perri F, Villani A, Binda E, Panebianco C, Mencarelli G, Ciuffreda L, Ferri Marini C, Annibalini G, Lucertini F, Bartolacci A, Imperio M, Virgili E, Catalano V, Piccoli G, Stocchi V, Emili R, Barbieri E. Changes in gut microbiota composition after 12 weeks of a home-based lifestyle intervention in breast cancer survivors during the COVID-19 lockdown. Front Oncol 2023; 13:1225645. [PMID: 37727203 PMCID: PMC10505708 DOI: 10.3389/fonc.2023.1225645] [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: 05/19/2023] [Accepted: 08/18/2023] [Indexed: 09/21/2023] Open
Abstract
Background Breast cancer (BC) is the second-leading cause of cancer-related death worldwide. This study aimed to investigate the effects of a 12-week home-based lifestyle intervention (based on nutrition and exercise) on gut microbial composition in twenty BC survivors of the MoviS clinical trial (protocol: NCT04818359). Methods Gut microbiota analysis through 16S rRNA gene sequencing, anthropometrics, Mediterranean Diet (MD) adherence, and cardiometabolic parameters were evaluated before (Pre) and after (Post) the lifestyle intervention (LI). Results Beneficial effects of the LI were observed on MD adherence, and cardiometabolic parameters (pre vs post). A robust reduction of Proteobacteria was observed after LI, which is able to reshape the gut microbiota by modulating microorganisms capable of decreasing inflammation and others involved in improving the lipid and glycemic assets of the host. A significant negative correlation between fasting glucose and Clostridia_vadinBB60 (r = -0.62), insulin and homeostatic model assessment (HOMA) index and Butyricicoccus genera (r = -0.72 and -0.66, respectively), and HDL cholesterol and Escherichia/Shigella (r = -0.59) have been reported. Moreover, positive correlations were found between MD adherence and Lachnospiraceae_ND3007 (r = 0.50), Faecalibacterium (r = 0.38) and Butyricimonas (r = 0.39). Conclusion These data suggest that adopting a healthy lifestyle, may contribute to ameliorate several biological parameters that could be involved in the prevention of cancer relapses through the modulation of gut microbiota.
Collapse
Affiliation(s)
- Sabrina Donati Zeppa
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Valentina Natalucci
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Deborah Agostini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Luciana Vallorani
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Stefano Amatori
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Davide Sisti
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Marco B. L. Rocchi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Valerio Pazienza
- Division of Gastroenterology, Fondazione IRCCS “Casa Sollievo della Sofferenza” Hospital, San Giovanni Rotondo, Italy
| | - Francesco Perri
- Division of Gastroenterology, Fondazione IRCCS “Casa Sollievo della Sofferenza” Hospital, San Giovanni Rotondo, Italy
| | - Annacandida Villani
- Division of Gastroenterology, Fondazione IRCCS “Casa Sollievo della Sofferenza” Hospital, San Giovanni Rotondo, Italy
| | - Elena Binda
- Cancer Stem Cells Unit, Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapeutics (ISBReMIT), IRCSS Casa Sollievo della Sofferenza, Opera di San Pio da Pietrelcina, San Giovanni Rotondo, Italy
| | - Concetta Panebianco
- Division of Gastroenterology, Fondazione IRCCS “Casa Sollievo della Sofferenza” Hospital, San Giovanni Rotondo, Italy
| | - Gandino Mencarelli
- Cancer Stem Cells Unit, Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapeutics (ISBReMIT), IRCSS Casa Sollievo della Sofferenza, Opera di San Pio da Pietrelcina, San Giovanni Rotondo, Italy
| | - Luigi Ciuffreda
- Breast Surgery Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Carlo Ferri Marini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Giosué Annibalini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Francesco Lucertini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Alessia Bartolacci
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Marta Imperio
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Edy Virgili
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Vincenzo Catalano
- U.O.C. Oncologia Medica, ASUR Area Vasta 1, Ospedale Santa Maria della Misericordia di Urbino, Urbino, Italy
| | - Giovanni Piccoli
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | | | - Rita Emili
- U.O.C. Oncologia Medica, ASUR Area Vasta 1, Ospedale Santa Maria della Misericordia di Urbino, Urbino, Italy
| | - Elena Barbieri
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| |
Collapse
|
47
|
Stine JG, Long MT, Corey KE, Sallis RE, Allen AM, Armstrong MJ, Conroy DE, Cuthbertson DJ, Duarte-Rojo A, Hallsworth K, Hickman IJ, Kappus MR, Keating SE, Pugh CJA, Rotman Y, Simon TL, Vilar-Gomez E, Wai-Sun Wong V, Schmitz KH. Physical Activity and Nonalcoholic Fatty Liver Disease: A Roundtable Statement from the American College of Sports Medicine. Med Sci Sports Exerc 2023; 55:1717-1726. [PMID: 37126039 PMCID: PMC10524517 DOI: 10.1249/mss.0000000000003199] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
ABSTRACT Although physical activity (PA) is crucial in the prevention and clinical management of nonalcoholic fatty liver disease, most individuals with this chronic disease are inactive and do not achieve recommended amounts of PA. There is a robust and consistent body of evidence highlighting the benefit of participating in regular PA, including a reduction in liver fat and improvement in body composition, cardiorespiratory fitness, vascular biology, and health-related quality of life. Importantly, the benefits of regular PA can be seen without clinically significant weight loss. At least 150 min of moderate or 75 min of vigorous intensity PA are recommended weekly for all patients with nonalcoholic fatty liver disease, including those with compensated cirrhosis. If a formal exercise training program is prescribed, aerobic exercise with the addition of resistance training is preferred. In this roundtable document, the benefits of PA are discussed, along with recommendations for 1) PA assessment and screening; 2) how best to advise, counsel, and prescribe regular PA; and 3) when to refer to an exercise specialist.
Collapse
Affiliation(s)
- Jonathan G. Stine
- Division of Gastroenterology and Hepatology, Department of Medicine, The Pennsylvania State University- Milton S. Hershey Medical Center, Hershey PA
- Department of Public Health Sciences, The Pennsylvania State University- College of Medicine, Hershey PA
| | - Michelle T. Long
- Section of Gastroenterology, Evans Department of Medicine, Boston University School of Medicine, Boston, MA
| | - Kathleen E. Corey
- Division of Gastroenterology and Hepatology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Robert E. Sallis
- Department of Family Medicine and Sports Medicine, Kaiser Permanente Medical Center, Fontana, CA
| | - Alina M. Allen
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN
| | - Matthew J. Armstrong
- Liver Transplant Unit, Queen Elizabeth University Hospitals Birmingham, and NIHR Birmingham Biomedical Research Centre, Birmingham, UNITED KINGDOM
| | - David E. Conroy
- Department of Kinesiology, The Pennsylvania State University, University Park, PA
| | - Daniel J. Cuthbertson
- Department of Cardiovascular and Metabolic Medicine, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UNITED KINGDOM
| | - Andres Duarte-Rojo
- Division of Gastroenterology and Hepatology, Department of Medicine, Northwestern University, Chicago, IL
| | - Kate Hallsworth
- Newcastle NIHR Biomedical Research Centre and the Liver Unit, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UNITED KINGDOM
| | - Ingrid J. Hickman
- Department of Nutrition and Dietetics, Princess Alexandra Hospital, Brisbane, Queensland, AUSTRALIA
| | - Matthew R. Kappus
- Division of Gastroenterology and Hepatology, Duke University, Durham, NC
| | - Shelley E. Keating
- School of Human Movement and Nutrition Sciences, The University of Queensland, St Lucia, Queensland, AUSTRALIA
| | - Christopher J. A. Pugh
- Cardiff School of Sport & Health Sciences, Cardiff Metropolitan University, Cardiff, UNITED KINGDOM
| | - Yaron Rotman
- Liver & Energy Metabolism Section, Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD
| | - Tracey L. Simon
- Division of Gastroenterology and Hepatology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Eduardo Vilar-Gomez
- Division of Gastroenterology and Hepatology. Indiana University School of Medicine. Indianapolis
| | - Vincent Wai-Sun Wong
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, CHINA
| | | |
Collapse
|
48
|
Guan L, Liu R. The Role of Diet and Gut Microbiota Interactions in Metabolic Homeostasis. Adv Biol (Weinh) 2023; 7:e2300100. [PMID: 37142556 DOI: 10.1002/adbi.202300100] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 04/10/2023] [Indexed: 05/06/2023]
Abstract
Diet is a pivotal determinant in shaping the structure and function of resident microorganisms in the gut through different food components, nutritive proportion, and calories. The effects of diet on host metabolism and physiology can be mediated through the gut microbiota. Gut microbiota-derived metabolites have been shown to regulate glucose and lipid metabolism, energy consumption, and the immune system. On the other hand, emerging evidence indicates that baseline gut microbiota could predict the efficacy of diet intervention, highlighting gut microbiota can be harnessed as a biomarker in personalized nutrition. In this review, the alterations of gut microbiota in different dietary components and dietary patterns, and the potential mechanisms in the diet-microbiota crosstalk are summarized to understand the interactions of diet and gut microbiota on the impact of metabolic homeostasis.
Collapse
Affiliation(s)
- Lizhi Guan
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Disease, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the P. R. China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ruixin Liu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Disease, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the P. R. China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| |
Collapse
|
49
|
Vitulo M, Gnodi E, Rosini G, Meneveri R, Giovannoni R, Barisani D. Current Therapeutical Approaches Targeting Lipid Metabolism in NAFLD. Int J Mol Sci 2023; 24:12748. [PMID: 37628929 PMCID: PMC10454602 DOI: 10.3390/ijms241612748] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/07/2023] [Accepted: 08/10/2023] [Indexed: 08/27/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD, including nonalcoholic fatty liver (NAFL) and nonalcoholic steatohepatitis (NASH)) is a high-prevalence disorder, affecting about 1 billion people, which can evolve to more severe conditions like cirrhosis or hepatocellular carcinoma. NAFLD is often concomitant with conditions of the metabolic syndrome, such as central obesity and insulin-resistance, but a specific drug able to revert NAFL and prevent its evolution towards NASH is still lacking. With the liver being a key organ in metabolic processes, the potential therapeutic strategies are many, and range from directly targeting the lipid metabolism to the prevention of tissue inflammation. However, side effects have been reported for the drugs tested up to now. In this review, different approaches to the treatment of NAFLD are presented, including newer therapies and ongoing clinical trials. Particular focus is placed on the reverse cholesterol transport system and on the agonists for nuclear factors like PPAR and FXR, but also drugs initially developed for other conditions such as incretins and thyromimetics along with validated natural compounds that have anti-inflammatory potential. This work provides an overview of the different therapeutic strategies currently being tested for NAFLD, other than, or along with, the recommendation of weight loss.
Collapse
Affiliation(s)
- Manuela Vitulo
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (M.V.); (E.G.); (R.M.)
| | - Elisa Gnodi
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (M.V.); (E.G.); (R.M.)
| | - Giulia Rosini
- Department of Biology, University of Pisa, 56021 Pisa, Italy; (G.R.); (R.G.)
| | - Raffaella Meneveri
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (M.V.); (E.G.); (R.M.)
| | - Roberto Giovannoni
- Department of Biology, University of Pisa, 56021 Pisa, Italy; (G.R.); (R.G.)
| | - Donatella Barisani
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (M.V.); (E.G.); (R.M.)
| |
Collapse
|
50
|
Frankowski R, Kobierecki M, Wittczak A, Różycka-Kosmalska M, Pietras T, Sipowicz K, Kosmalski M. Type 2 Diabetes Mellitus, Non-Alcoholic Fatty Liver Disease, and Metabolic Repercussions: The Vicious Cycle and Its Interplay with Inflammation. Int J Mol Sci 2023; 24:ijms24119677. [PMID: 37298632 DOI: 10.3390/ijms24119677] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/26/2023] [Accepted: 05/31/2023] [Indexed: 06/12/2023] Open
Abstract
The prevalence of metabolic-related disorders, such as non-alcoholic fatty liver disease (NAFLD) and type 2 diabetes mellitus (DM2), has been increasing. Therefore, developing improved methods for the prevention, treatment, and detection of these two conditions is also necessary. In this study, our primary focus was on examining the role of chronic inflammation as a potential link in the pathogenesis of these diseases and their interconnections. A comprehensive search of the PubMed database using keywords such as "non-alcoholic fatty liver disease", "type 2 diabetes mellitus", "chronic inflammation", "pathogenesis", and "progression" yielded 177 relevant papers for our analysis. The findings of our study revealed intricate relationships between the pathogenesis of NAFLD and DM2, emphasizing the crucial role of inflammatory processes. These connections involve various molecular functions, including altered signaling pathways, patterns of gene methylation, the expression of related peptides, and up- and downregulation of several genes. Our study is a foundational platform for future research into the intricate relationship between NAFLD and DM2, allowing for a better understanding of the underlying mechanisms and the potential for introducing new treatment standards.
Collapse
Affiliation(s)
- Rafał Frankowski
- Students' Research Club, Department of Clinical Pharmacology, Medical University of Lodz, 90-153 Lodz, Poland
| | - Mateusz Kobierecki
- Students' Research Club, Department of Clinical Pharmacology, Medical University of Lodz, 90-153 Lodz, Poland
| | - Andrzej Wittczak
- Students' Research Club, Department of Clinical Pharmacology, Medical University of Lodz, 90-153 Lodz, Poland
| | | | - Tadeusz Pietras
- Department of Clinical Pharmacology, Medical University of Lodz, 90-153 Lodz, Poland
| | - Kasper Sipowicz
- Department of Interdisciplinary Disability Studies, The Maria Grzegorzewska University in Warsaw, 02-353 Warsaw, Poland
| | - Marcin Kosmalski
- Department of Clinical Pharmacology, Medical University of Lodz, 90-153 Lodz, Poland
| |
Collapse
|