1
|
Wang D, Miao J, Zhang L, Zhang L. Research advances in the diagnosis and treatment of MASLD/MASH. Ann Med 2025; 57. [DOI: 10.1080/07853890.2024.2445780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 12/01/2024] [Accepted: 12/02/2024] [Indexed: 01/06/2025] Open
Affiliation(s)
- Dekai Wang
- Department of General Practice, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jinxian Miao
- Department of General Practice, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Lihua Zhang
- Department of General Practice, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Lin Zhang
- Department of General Practice, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
2
|
Zhu T, Shen D, Cai X, Jin Y, Tu H, Wang S, Pan Q. The causal relationship between gut microbiota and preterm birth: a two-sample Mendelian randomization study. J Matern Fetal Neonatal Med 2025; 38:2432528. [PMID: 39721770 DOI: 10.1080/14767058.2024.2432528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 11/07/2024] [Accepted: 11/14/2024] [Indexed: 12/28/2024]
Abstract
BACKGROUND Preterm birth, a significant global health concern, has been associated with alterations in the gut microbiota. However, the causal nature of this relationship remains uncertain due to the limitations inherent in observational studies. PURPOSE To investigate the potential causal relationship between gut microbiota imbalances and preterm birth. METHODS We conducted a two-sample Mendelian randomization (MR) study using genome-wide association study (GWAS) data from the MiBioGen consortium focusing on microbiota and preterm birth. Single nucleotide polymorphisms (SNPs) associated with the microbiota were selected as instrumental variables. The inverse variance weighting (IVW) method was used to estimate causality. We confirmed pleiotropy and identified and excluded outlier SNPs using MR-PRESSO and MR-Egger regression. Cochran's Q test was applied to assess heterogeneity among SNPs, and a leave-one-out analysis was performed to evaluate the influence of individual SNPs on overall estimates. RESULTS Our findings provide evidence for a causal link between specific components of the gut microbiota and preterm birth, with the identification of relevant metabolites. CONCLUSION This study highlights the causal role of gut microbiota imbalances in preterm birth, offering novel insights into the development of preterm birth and potential targets for prevention strategies.
Collapse
Affiliation(s)
- Tao Zhu
- Department of Clinical Laboratory, Sir Run Run Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Dandan Shen
- Department of Laboratory Medicine, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiao Cai
- Department of Clinical Laboratory, Sir Run Run Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yuanling Jin
- Department of Clinical Laboratory, Sir Run Run Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Haixia Tu
- Department of Clinical Laboratory, Sir Run Run Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Shouxing Wang
- Department of Clinical Laboratory, Sir Run Run Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qianglong Pan
- Department of Clinical Laboratory, Sir Run Run Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
3
|
Afsar B, Afsar RE, Caliskan Y, Lentine KL. Use of Anti-interleukin-1 Agents in Kidney Transplant Recipients with Familial Mediterranean Fever and Amyloidosis: What have been learned so far? CURRENT TRANSPLANTATION REPORTS 2025; 12:4. [PMID: 40092658 PMCID: PMC11905541 DOI: 10.1007/s40472-025-00461-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/02/2025] [Indexed: 03/19/2025]
Abstract
Purpose of Review Familial Mediterranean fever (FMF) is the most common monogenic auto-inflammatory disease causing amyloidosis (AA type) which may result in development of end-stage kidney disease (ESKD). Colchicine is the initial treatment option for patients FMF/amyloidosis both before and after KT. Although, kidney transplantation (KT) can be offered to patients with ESKD due to FMF/amyloidosis, FMF attacks did not resolve in some of kidney transplant recipients (KTRs) and de-novo development of amyloidosis after KT may be observed despite colchicine treatment. For these patients, other treatment options are warranted including anti-interleukin-1 agents such as anakinra and canakinumab. The purpose of the review is to summarize the use of anti-interleukin-1 agents in KTRs with FMF and amyloidosis. Recent Findings Recent studies showed that these agents are effective in KTRs in terminating FMF attacks and decreasing inflammatory parameters. Furthermore, no significant interaction with immunosuppressive drugs were recorded and side effects were few. However, there are various knowledge gaps.
Collapse
Affiliation(s)
- Baris Afsar
- Saint Louis University, School of Medicine, SSM Health Saint Louis University Hospital, Department of Nephrology
| | - Rengin Elsurer Afsar
- Saint Louis University, School of Medicine, SSM Health Saint Louis University Hospital, Department of Nephrology
| | - Yasar Caliskan
- Saint Louis University Transplant Center, SSM Health Saint Louis University Hospital, St. Louis, MO, USA
| | - Krista L. Lentine
- Saint Louis University Transplant Center, SSM Health Saint Louis University Hospital, St. Louis, MO, USA
| |
Collapse
|
4
|
Gao D, Bing C, Griffiths HR. Disrupted adipokine secretion and inflammatory responses in human adipocyte hypertrophy. Adipocyte 2025; 14:2485927. [PMID: 40176539 PMCID: PMC11980453 DOI: 10.1080/21623945.2025.2485927] [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: 07/02/2024] [Revised: 12/27/2024] [Accepted: 12/30/2024] [Indexed: 04/04/2025] Open
Abstract
Adipocyte hypertrophy is a critical contributor to obesity-induced inflammation and insulin resistance. This study employed a human adipocyte hypertrophy model to investigate the adipokine release, inflammatory responses, and the intracellular singling pathways. Hypertrophic adipocytes exhibited increased lipid content and lipolysis, a decline of anti-inflammatory adipokine adiponectin release and an increase of pro-inflammatory adipokine leptin release compared to mature adipocytes. Moreover, TNFα and LPS exacerbated the decrease in adiponectin secretion by hypertrophic adipocytes while promoting the secretion of leptin, MCP-1 and IL-6, which is associated with impaired activation of p38 and JNK MAPK and persistent activation of ERK and IκBα in hypertrophic adipocytes. These altered adipokine secretions and inflammatory responses within hypertrophic adipocytes may contribute to adipocyte dysfunction in human obesity.
Collapse
Affiliation(s)
- Dan Gao
- Institute of Molecular and Translational Medicine, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
- Ministry of Education, Key Laboratory of Environment and Genes Related to Diseases Xi’an Jiaotong University, Xi’an, China
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Center, Xi’an, China
| | - Chen Bing
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | | |
Collapse
|
5
|
Sall I, Foxall R, Felth L, Maret S, Rosa Z, Gaur A, Calawa J, Pavlik N, Whistler JL, Whistler CA. Gut dysbiosis was inevitable, but tolerance was not: temporal responses of the murine microbiota that maintain its capacity for butyrate production correlate with sustained antinociception to chronic morphine. Gut Microbes 2025; 17:2446423. [PMID: 39800714 PMCID: PMC11730370 DOI: 10.1080/19490976.2024.2446423] [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: 04/16/2024] [Revised: 11/24/2024] [Accepted: 12/18/2024] [Indexed: 01/16/2025] Open
Abstract
The therapeutic benefits of opioids are compromised by the development of analgesic tolerance, which necessitates higher dosing for pain management thereby increasing the liability for drug dependence and addiction. Rodent models indicate opposing roles of the gut microbiota in tolerance: morphine-induced gut dysbiosis exacerbates tolerance, whereas probiotics ameliorate tolerance. Not all individuals develop tolerance, which could be influenced by differences in microbiota, and yet no study design has capitalized upon this natural variation. We leveraged natural behavioral variation in a murine model of voluntary oral morphine self-administration to elucidate the mechanisms by which microbiota influences tolerance. Although all mice shared similar morphine-driven microbiota changes that largely masked informative associations with variability in tolerance, our high-resolution temporal analyses revealed a divergence in the progression of dysbiosis that best explained sustained antinociception. Mice that did not develop tolerance maintained a higher capacity for production of the short-chain fatty acid (SCFA) butyrate known to bolster intestinal barriers and promote neuronal homeostasis. Both fecal microbial transplantation (FMT) from donor mice that did not develop tolerance and dietary butyrate supplementation significantly reduced the development of tolerance independently of suppression of systemic inflammation. These findings could inform immediate therapies to extend the analgesic efficacy of opioids.
Collapse
Affiliation(s)
- Izabella Sall
- Department of Molecular, Cellular, & Biomedical Sciences, University of New Hampshire, Durham, NH, USA
- Graduate program in Molecular and Evolutionary Systems Biology, University of New Hampshire, Durham, NH, USA
| | - Randi Foxall
- Department of Molecular, Cellular, & Biomedical Sciences, University of New Hampshire, Durham, NH, USA
| | - Lindsey Felth
- Center for Neuroscience, University of California–Davis, Davis, CA, USA
| | - Soren Maret
- Department of Molecular, Cellular, & Biomedical Sciences, University of New Hampshire, Durham, NH, USA
| | - Zachary Rosa
- Center for Neuroscience, University of California–Davis, Davis, CA, USA
| | - Anirudh Gaur
- Center for Neuroscience, University of California–Davis, Davis, CA, USA
| | - Jennifer Calawa
- Department of Molecular, Cellular, & Biomedical Sciences, University of New Hampshire, Durham, NH, USA
- Microbiology Graduate Program, University of New Hampshire, Durham, NH, USA
| | - Nadia Pavlik
- Department of Molecular, Cellular, & Biomedical Sciences, University of New Hampshire, Durham, NH, USA
| | - Jennifer L. Whistler
- Center for Neuroscience, University of California–Davis, Davis, CA, USA
- Department of Physiology and Membrane Biology, UC Davis School of Medicine, Davis, CA, USA
| | - Cheryl A. Whistler
- Department of Molecular, Cellular, & Biomedical Sciences, University of New Hampshire, Durham, NH, USA
| |
Collapse
|
6
|
Gonzalez M, Robinson S, Mills NL, Eriksson M, Sandström T, Newby DE, Olsson T, Blomberg A, Söderberg S. Vasomotor and fibrinolytic effects of leptin in man. SCAND CARDIOVASC J 2025; 59:2478867. [PMID: 40066842 DOI: 10.1080/14017431.2025.2478867] [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: 10/25/2024] [Revised: 02/05/2025] [Accepted: 03/07/2025] [Indexed: 03/20/2025]
Abstract
OBJECTIVES The adipocyte-derived hormone leptin has been associated with the pathogenesis of cardiovascular disease. The mechanisms underlying this association are unclear but may relate to effects on the vascular endothelium. Our aim was to explore the effects of leptin on endothelial vasomotor and fibrinolytic function in healthy volunteers and patients with coronary artery disease. DESIGN The vascular effects of leptin were assessed infusing recombinant human leptin in healthy volunteers during measuring vasomotor response by venous occlusion plethysmography. Additionally, circulating levels of leptin were analysed in relation to endothelial dysfunction in patients with established coronary artery disease. RESULTS In healthy male volunteers, intra-arterial infusion of recombinant human leptin (80, 800 and 8,000 ng/min; n = 10) did not affect basal forearm blood flow, plasma tissue plasminogen activator (tPA) or plasminogen activator inhibitor type 1 concentrations (all p > 0.05). However, during concomitant co-infusion with leptin (800 ng/min; n = 10), drug-induced vasodilatation was reduced (p = 0.001), and tPA activity increased (p = 0.002). In patients with coronary artery disease, those with the high plasma leptin levels had reduced drug-induced vasodilatation (p < 0.001), and increased net release of tPA antigen and activity (p < 0.001 and p = 0.03, respectively) compared to those with low levels. The study has been registered retrospectively at Clinical Trials with number NCT04374500. CONCLUSION Intrabrachial leptin infusion did not affect the basal vascular tone, whereas acute and chronic hyperleptinemia was associated with blunted vasoreactivity in healthy volunteers, and in patients with coronary artery disease.
Collapse
Affiliation(s)
- Manuel Gonzalez
- Department of Public Health and Clinical Medicine, Section of Medicine, Umeå University, Umeå, Sweden
| | - Simon Robinson
- Royal Jubilee Hospital, Victoria Heart Institute Foundation, Victoria, BC, Canada
| | - Nicholas L Mills
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | | | - Thomas Sandström
- Department of Public Health and Clinical Medicine, Section of Medicine, Umeå University, Umeå, Sweden
| | - David E Newby
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Tommy Olsson
- Department of Public Health and Clinical Medicine, Section of Medicine, Umeå University, Umeå, Sweden
| | - Anders Blomberg
- Department of Public Health and Clinical Medicine, Section of Medicine, Umeå University, Umeå, Sweden
| | - Stefan Söderberg
- Department of Public Health and Clinical Medicine, Section of Medicine, Umeå University, Umeå, Sweden
| |
Collapse
|
7
|
Barnett D, Thijs C, Mommers M, Endika M, Klostermann C, Schols H, Smidt H, Nauta A, Arts I, Penders J. Why do babies cry? Exploring the role of the gut microbiota in infantile colic, constipation, and cramps in the KOALA birth cohort study. Gut Microbes 2025; 17:2485326. [PMID: 40159147 PMCID: PMC11959906 DOI: 10.1080/19490976.2025.2485326] [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: 10/23/2024] [Revised: 02/17/2025] [Accepted: 03/18/2025] [Indexed: 04/02/2025] Open
Abstract
Gastrointestinal symptoms are common during infancy, including infantile colic. Colic can be loosely defined as prolonged and recurrent crying without obvious cause. The cause indeed remains unclear despite much research. Results on infant nutrition are inconclusive, but prior work has linked maternal mental health to infant crying. Recently, several small studies have described associations between gut microbiota and colic. We used a larger cohort to examine the role of the microbiota in infant gastrointestinal health, while also accounting for other biopsychosocial factors. Using fecal 16S rRNA gene amplicon sequencing data from 1,012 infants in the KOALA birth cohort, we examined associations between the 1-month gut microbiota and parent-reported functional gastrointestinal symptoms throughout infancy, including colic, constipation, and cramps. These analyses were adjusted for biopsychosocial factors that were associated with symptoms in a broader analysis involving 2,665 participants. In 257 infants, we also explored associations between breastmilk human milk oligosaccharides (HMOs) and gastrointestinal symptoms. Higher relative abundance of Staphylococcus at one month was associated with less constipation in the first three months of life. Conversely, Ruminococcus gnavus group abundance was associated with more colicky symptoms, particularly between four and seven months. Breastmilk concentrations of the HMOs lacto-N-hexaose (LNH) and lacto-N-neohexaose (LNnH) were associated with less constipation in the first three months. Our results support the conclusion that gut microbiota are relevant in infantile colic and constipation. However more work is needed to elucidate the underlying mechanisms, and explore their interplay with other relevant biopsychosocial factors such as maternal mental health.
Collapse
Affiliation(s)
- David Barnett
- Maastricht Centre for Systems Biology, Maastricht University, Maastricht, Netherlands
- NUTRIM School for Nutrition and Translational Research in Metabolism, Department of Medical Microbiology, Infectious Diseases and Infection Prevention, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Carel Thijs
- CAPHRI Care and Public Health Research Institute, Department of Epidemiology, Maastricht University, Maastricht, Netherlands
| | - Monique Mommers
- CAPHRI Care and Public Health Research Institute, Department of Epidemiology, Maastricht University, Maastricht, Netherlands
| | - Martha Endika
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, Netherlands
| | - Cynthia Klostermann
- Department of Food Chemistry, Wageningen University & Research, Wageningen, Netherlands
| | - Henk Schols
- Department of Food Chemistry, Wageningen University & Research, Wageningen, Netherlands
| | - Hauke Smidt
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, Netherlands
| | - Arjen Nauta
- FrieslandCampina Ingredients, FrieslandCampina, Amersfoort, Netherlands
| | - Ilja Arts
- Maastricht Centre for Systems Biology, Maastricht University, Maastricht, Netherlands
| | - John Penders
- NUTRIM School for Nutrition and Translational Research in Metabolism, Department of Medical Microbiology, Infectious Diseases and Infection Prevention, Maastricht University Medical Center+, Maastricht, Netherlands
| |
Collapse
|
8
|
Qiao S, Wang T, Sun J, Han J, Dai H, Du M, Yang L, Guo CJ, Liu C, Liu SJ, Liu H. Cross-feeding-based rational design of a probiotic combination of Bacterides xylanisolvens and Clostridium butyricum therapy for metabolic diseases. Gut Microbes 2025; 17:2489765. [PMID: 40190016 PMCID: PMC11980479 DOI: 10.1080/19490976.2025.2489765] [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: 04/29/2024] [Revised: 12/13/2024] [Accepted: 03/18/2025] [Indexed: 04/11/2025] Open
Abstract
The human gut microbiota has gained interest as an environmental factor that contributes to health or disease. The development of next-generation live biotherapeutic products (LBPs) is a promising strategy to modulate the gut microbiota and improve human health. In this study, we identified a novel cross-feeding interaction between Bacteroides xylanisolvens and Clostridium butyricum and developed their combination into a novel LBP for treating metabolic syndrome. Using in-silico analysis and in vitro experiments, we demonstrated that B. xylanisolvens supported the growth and butyrate production of C. butyricum by supplying folate, while C. butyricum reciprocated by providing pABA for folate biosynthesis. Animal gavage experiments showed that the two-strain combination LBP exhibited superior therapeutic efficacy against metabolic disorders in high-fat diet-induced obese (DIO) mice compared to either single-strain treatment. Further omics-based analyses revealed that the single-strain treatments exhibited distinct taxonomic preferences in modulating the gut microbiota, whereas the combination LBP achieved more balanced modulation to preserve taxonomic diversity to a greater extent, thereby enhancing the stability and resilience of the gut microbiome. Moreover, the two-strain combinations more effectively restored gut microbial functions by reducing disease-associated pathways and opportunistic pathogen abundance. This work demonstrates the development of new LBP therapy for metabolic diseases from cross-feeding microbial pairs which exerted better self-stability and robust efficacy in complex intestinal environments compared to conventional single-strain LBPs.
Collapse
Affiliation(s)
- Shanshan Qiao
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, P. R. China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Tao Wang
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, P. R. China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Jingzu Sun
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, P. R. China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Junjie Han
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, P. R. China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Huanqin Dai
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, P. R. China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Mengxuan Du
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, P. R. China
| | - Lan Yang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, P. R. China
| | - Chun-Jun Guo
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Chang Liu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, P. R. China
| | - Shuang-Jiang Liu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, P. R. China
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, P. R. China
| | - Hongwei Liu
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, P. R. China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, P. R. China
| |
Collapse
|
9
|
Ast HK, Hammer M, Zhang S, Bruton A, Hatsu IE, Leung B, McClure R, Srikanth P, Farris Y, Norby-Adams L, Robinette LM, Arnold LE, Swann JR, Zhu J, Karstens L, Johnstone JM. Gut microbiome changes with micronutrient supplementation in children with attention-deficit/hyperactivity disorder: the MADDY study. Gut Microbes 2025; 17:2463570. [PMID: 39963956 PMCID: PMC11845018 DOI: 10.1080/19490976.2025.2463570] [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: 07/03/2024] [Revised: 01/17/2025] [Accepted: 02/02/2025] [Indexed: 02/23/2025] Open
Abstract
Micronutrients have demonstrated promise in managing inattention and emotional dysregulation in children with attention-deficit/hyperactivity disorder (ADHD). One plausible pathway by which micronutrients improve symptoms is the gut microbiome. This study examines changes in fecal microbial composition and diversity after micronutrient supplementation in children with ADHD (N = 44) and highlights potential mechanisms responsible for the behavioral improvement, as determined by blinded clinician-rated global improvement response to micronutrients. Participants represent a sub-group of the Micronutrients for ADHD in Youth (MADDY) study, a double blind randomized controlled trial in which participants received micronutrients or placebo for 8 weeks, followed by an 8-week open extension. Stool samples collected at baseline, week 8, and week 16 were analyzed using 16S rRNA amplicon sequencing targeting the V4 hypervariable region. Pairwise compositional analyses investigated changes in fecal microbial composition between micronutrients versus placebo and responders versus non-responders. A significant change in microbial evenness, as measured by alpha diversity, and beta-diversity, as measured by Bray-Curtis, was observed following micronutrients supplementation. The phylum Actinobacteriota decreased in the micronutrients group compared to placebo. Two butyrate-producing bacterial families: Rikenellaceae and Oscillospiraceae, exhibited a significant increase in change following micronutrients between responders versus non-responders. These findings suggest that micronutrients modulated the composition of the fecal microbiota and identified specific bacterial changes associated with micronutrient responders.
Collapse
Affiliation(s)
- Hayleigh K. Ast
- Department of Psychiatry, Center for Mental Health Innovation, Oregon Health & Science University, Portland, OR, USA
| | - Matthew Hammer
- Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, OR, USA
| | - Shiqi Zhang
- Department of Human Sciences, The Ohio State University, Columbus, OH, USA
| | - Alisha Bruton
- Department of Psychiatry, Center for Mental Health Innovation, Oregon Health & Science University, Portland, OR, USA
| | - Irene E. Hatsu
- Department of Human Sciences, The Ohio State University, Columbus, OH, USA
| | - Brenda Leung
- Faculty of Health Sciences, University of Lethbridge, Lethbridge, AB, Canada
| | - Ryan McClure
- Pacific Northwest National Laboratory, Richland, WA, USA
| | - Priya Srikanth
- Oregon Health and Science University-Portland State University School of Public Health, Portland, OR, USA
| | - Yuliya Farris
- Pacific Northwest National Laboratory, Richland, WA, USA
| | - Lydia Norby-Adams
- Department of Psychiatry, Center for Mental Health Innovation, Oregon Health & Science University, Portland, OR, USA
- Helfgott Research Institute, National University of Natural Medicine, Portland, OR, USA
| | - Lisa M. Robinette
- Department of Human Sciences, The Ohio State University, Columbus, OH, USA
| | - L. Eugene Arnold
- Department of Psychiatry & Behavioral Health, The Ohio State University, Columbus, OH, USA
| | - Jonathan R. Swann
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Jiangjiang Zhu
- Department of Human Sciences, The Ohio State University, Columbus, OH, USA
| | - Lisa Karstens
- Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, OR, USA
| | - Jeanette M. Johnstone
- Department of Psychiatry, Center for Mental Health Innovation, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
10
|
Singh S, Abu Y, Antoine D, Gomez D, Tao J, Truitt B, Roy S. Probiotic supplementation mitigates sex-dependent nociceptive changes and gut dysbiosis induced by prenatal opioid exposure. Gut Microbes 2025; 17:2464942. [PMID: 39950489 PMCID: PMC11834462 DOI: 10.1080/19490976.2025.2464942] [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: 06/20/2024] [Revised: 11/21/2024] [Accepted: 02/04/2025] [Indexed: 02/20/2025] Open
Abstract
The gut microbiome has emerged as a promising target for modulating adverse effects of opioid exposure due to its significant role in health and disease. Opioid use disorder (OUD) has become increasingly prevalent, specifically in women of reproductive age, contributing to an increased incidence of offspring exposed to opioids in utero. Recent studies have shown that prenatal opioid exposure (POE) is associated with notable changes to the maternal gut microbiome, with subsequent implications for the offspring's microbiome and other adverse outcomes. However, the role of the gut microbiome in mediating sex-based differences in pain sensitivity has not yet been investigated. In this study, both male and female C57BL/6 offspring were used to determine sex-based differences in nociception and gut microbial composition as a result of POE. Our data reveals significant sex-based differences in offspring prenatally exposed to opioids. The gut microbiome of opioid-exposed females showed an enrichment of commensal bacteria including Lactobacillus compared to opioid-exposed males. Additionally, POE females demonstrated decreased nociceptive sensitivity, while males demonstrated increased nociceptive sensitivity. RNA sequencing of the prefrontal cortex showed sex-based differences in several canonical pathways, including an increase in the opioid signaling pathway of opioid-exposed females, which was not observed in males. Microbiome modification via maternal probiotic supplementation attenuated sex-based differences throughout the early stages of life. Together, our study provides further insight on sex-based differences arising from POE and highlights the pivotal role of the gut microbiome as a modifiable target for mitigating its negative effects.
Collapse
Affiliation(s)
- Salma Singh
- Department of Surgery, School of Medicine, University of Miami Miller, Miami, USA
| | - Yaa Abu
- Department of Surgery, School of Medicine, University of Miami Miller, Miami, USA
| | - Danielle Antoine
- Department of Surgery, School of Medicine, University of Miami Miller, Miami, USA
- Department of Neuroscience, School of Medicine, University of Miami Miller, Miami, USA
| | - Daniel Gomez
- Department of Surgery, School of Medicine, University of Miami Miller, Miami, USA
| | - Junyi Tao
- Department of Surgery, School of Medicine, University of Miami Miller, Miami, USA
| | - Bridget Truitt
- Department of Surgery, School of Medicine, University of Miami Miller, Miami, USA
- Department of Neuroscience, School of Medicine, University of Miami Miller, Miami, USA
| | - Sabita Roy
- Department of Surgery, School of Medicine, University of Miami Miller, Miami, USA
| |
Collapse
|
11
|
Petersen C, Satheesh Babu AK, Della Lucia CM, Paz HA, Iglesias-Carres L, Zhong Y, Jalili T, Symons JD, Shankar K, Neilson AP, Wankhade UD, Anandh Babu PV. Gut microbes metabolize strawberry phytochemicals and mediate their beneficial effects on vascular inflammation. Gut Microbes 2025; 17:2446375. [PMID: 39760464 DOI: 10.1080/19490976.2024.2446375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 11/02/2024] [Accepted: 12/18/2024] [Indexed: 01/07/2025] Open
Abstract
Evidence suggests that a healthy gut microbiome is essential for metabolizing dietary phytochemicals. However, the microbiome's role in metabolite production and the influence of gut dysbiosis on this process remain unclear. Further, studies on the relationship among gut microbes, metabolites, and biological activities of phytochemicals are limited. We addressed this knowledge gap using strawberry phytochemicals as a model. C57BL/6J mice were fed a standard diet [C]; strawberry-supplemented diet (~2 human servings) [CS]; strawberry-supplemented diet and treated with antibiotics (to deplete gut microbes) [CSA]; high-fat diet (HFD) [HF]; strawberry-supplemented HFD [HS]; and strawberry-supplemented HFD and treated with antibiotics [HSA] for 12 weeks. First, antibiotic treatment suppressed the production of selected metabolites (CSA vs. CS), and p-coumaric acid was identified as a strawberry-derived microbial metabolite. Second, HFD-induced dysbiosis negatively affected metabolite production (HS vs. HF), and hippuric acid was identified as a microbial metabolite in HFD conditions. Third, dietary strawberries improved HFD-induced vascular inflammation (HS vs. HF). However, antibiotic treatment reduced metabolite production and abolished the vascular effects of strawberries (HSA vs. HS), indicating the importance of gut microbes in mediating the vascular benefits of strawberries via metabolites. Fourth, strawberry supplementation decreased Coprobacillus that was positively associated with vascular inflammation, whereas it increased Lachnospiraceae that was negatively associated with vascular inflammation and positively associated with hippuric acid. Fifth, hippuric acid was negatively associated with vascular inflammation. Our study fills in some pieces of the giant puzzle regarding the influence of gut microbes on the biological activities of phytochemicals. HFD-induced gut dysbiosis negatively impacts metabolite production and a strong association exists among gut microbes, strawberry-derived microbial metabolites, and the vascular benefits of dietary strawberries. Further, our study provides significant proof of concept to warrant future research on the use of strawberries as a nutritional strategy to prevent vascular complications.
Collapse
Affiliation(s)
- Chrissa Petersen
- Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, UT, USA
| | | | - Ceres Mattos Della Lucia
- Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, UT, USA
| | - Henry A Paz
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Lisard Iglesias-Carres
- Plants for Human Health Institute, Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University, Kannapolis, NC, USA
| | - Ying Zhong
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Thunder Jalili
- Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, UT, USA
| | - J David Symons
- Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, UT, USA
| | - Kartik Shankar
- Department of Pediatrics, Section of Nutrition, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Andrew P Neilson
- Plants for Human Health Institute, Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University, Kannapolis, NC, USA
| | - Umesh D Wankhade
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Pon Velayutham Anandh Babu
- Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
12
|
Hu L, Li J, Hu L, Zhang M, Wang Y, Wang M, Xu Y. Relationship between gastrointestinal disturbances, blood lipid levels, inflammatory markers, and preterm birth. J OBSTET GYNAECOL 2025; 45:2475065. [PMID: 40254961 DOI: 10.1080/01443615.2025.2475065] [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: 12/11/2024] [Accepted: 02/27/2025] [Indexed: 04/22/2025]
Abstract
BACKGROUND The challenging incidence of preterm birth, the underlying causes of preterm birth remain unclear. This study determined the relationship between disturbed gastrointestinal symptoms, inflammatory markers, blood lipid levels, and preterm birth. METHOD One hundred and twenty pregnant women with preterm labour were compared to 120 pregnant women with full-term deliveries. All subjects underwent lactose breath and serologic testing. The correlation between small intestinal bacterial overgrowth (SIBO)-positivity, gastrointestinal symptoms, inflammatory factors, and blood lipid metabolism and preterm birth was analysed using the Spearman method. RESULTS SIBO, hydrogen, and methane levels were significantly higher in the preterm birth (PTB) group than the full-term birth (FTB) group at different time points (P < 0.05); Levels of high-sensitivity C-reactive protein (hs-CRP) (3.95[2.70-5.77] vs. 2.47[1.45-3.83]), Interleukin (IL)-10 (3.05[2.27-4.33] vs. 2.09[1.04-3.47]), IL-6 (5.23[3.95-8.50] vs. 2.98[2.22-4.44]), tumour necrosis factor -alpha (TNF-α) (3.23[1.55-4.90] vs. 1.76[0.98-3.10]), total cholesterol (TC) (5.52[4.97-5.95] vs. 5.24[4.73-5.85]), and triglycerides (TG) (2.58[2.04-3.53] vs. 2.24[1.59-3.05]) were significantly higher in the PTB group than the FTB group (P < 0.05). Abdominal distension (2.67[1.67-3.00] vs. 2.33[1.67-2.67]) and constipation (2.00[1.33-2.00] vs. 1.67[1.33-2.00]) scores were also markedly higher in the PTB group than the FTB group (P < 0.05). Preterm birth was positively correlated with SIBO, TC, and TG levels. Additionally, SIBO was positively correlated with hs-CRP, IL-10, IL-6, and TNF-α levels, abdominal distension, and constipation (P < 0.05). Logistic regression analysis found the close association between positive SIBO, biochemistry indicators and preterm birth. CONCLUSION Gastrointestinal disturbances, hyperlipidaemia and SIBO-positivity are more likely to occur among pregnant women with preterm labour. Further research with a large sample size in multi-centers is needed to validate the results.
Collapse
Affiliation(s)
- Lulu Hu
- Department of Obstetrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jingjing Li
- Department of Obstetrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Lin Hu
- Department of Obstetrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Miao Zhang
- Department of Obstetrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yixin Wang
- Department of Obstetrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Mengqi Wang
- Department of Obstetrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yajuan Xu
- Department of Obstetrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
13
|
Shui W, Niu Y, Zhang C, Pan Q. Investigating the correlation between blood manganese concentrations and anemia in U.S. adults: a nationally representative study. Hematology 2025; 30:2460895. [PMID: 39977843 DOI: 10.1080/16078454.2025.2460895] [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: 11/16/2024] [Accepted: 01/27/2025] [Indexed: 02/22/2025] Open
Abstract
BACKGROUND The health implications of trace elements have become increasingly concerning, yet the connection between blood manganese levels and anemia remains insufficiently examined. This research endeavors to explore the potential linkage between blood manganese concentrations and anemia. METHODS Utilizing data from the National Health and Nutrition Examination Survey (NHANES) from 2011 to 2018, this study examines the correlation between blood manganese levels and anemia among U.S. adults, offering a comprehensive national perspective. The study included 11,300 adults aged 20 and above, with both blood manganese and hemoglobin levels measured. Generalized Additive Model (GAM) was applied to delineate smooth curves, and threshold effect analysis was performed to identify the inflection points of these curves. Subsequently, unconditional logistic regression was employed to assess the risk. RESULTS Our research involved a total of 11,300 individuals, among which 1,143 (10.1%) were identified with anemia. The curve fitting analysis indicated a U-shaped relationship between blood manganese levels and the risk of anemia. Specifically, when blood manganese levels were below 8.69 µg/L, increasing concentrations were linked to a decreased risk of anemia, with an adjusted OR of 0.838 (95% CI: 0.735-0.954), indicating a protective effect of this level of blood manganese against anemia. Conversely, when blood manganese levels were at or above 8.69 µg/L, further elevations were strongly associated with an increased risk of anemia, with the adjusted OR rising to 1.160 (95% CI: 1.124-1.196), suggesting that excessively high blood manganese levels significantly raised the risk of developing anemia. CONCLUSION This study provides novel insights into the association between blood manganese levels and anemia. Further extensive, population-based cohort studies are necessary to validate the causality and to uncover the intrinsic toxicological mechanisms.
Collapse
Affiliation(s)
- Wei Shui
- Department of General Practice, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yuanyuan Niu
- Department of General Practice, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Changran Zhang
- Department of General Practice, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Qianying Pan
- Department of Hematology, the Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| |
Collapse
|
14
|
Tanaka M, Akiyama Y, Mori K, Hosaka I, Endo K, Ogawa T, Sato T, Suzuki T, Yano T, Ohnishi H, Hanawa N, Furuhashi M. Machine learning-based analyses of contributing factors for the development of hypertension: a comparative study. Clin Exp Hypertens 2025; 47:2449613. [PMID: 39773295 DOI: 10.1080/10641963.2025.2449613] [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: 02/22/2024] [Revised: 11/25/2024] [Accepted: 12/30/2024] [Indexed: 01/11/2025]
Abstract
OBJECTIVES Sufficient attention has not been given to machine learning (ML) models using longitudinal data for investigating important predictors of new onset of hypertension. We investigated the predictive ability of several ML models for the development of hypertension. METHODS A total of 15 965 Japanese participants (men/women: 9,466/6,499, mean age: 45 years) who received annual health examinations were randomly divided into a training group (70%, n = 11,175) and a test group (30%, n = 4,790). The predictive abilities of 58 candidates including fatty liver index (FLI), which is calculated by using body mass index, waist circumference and levels of γ-glutamyl transferase and triglycerides, were investigated by statistics analogous to the area under the curve (AUC) in receiver operating characteristic curve analyses using ML models including logistic regression, random forest, naïve Bayes, extreme gradient boosting and artificial neural network. RESULTS During a 10-year period (mean period: 6.1 years), 2,132 subjects (19.1%) in the training group and 917 subjects (19.1%) in the test group had new onset of hypertension. Among the 58 parameters, systolic blood pressure, age and FLI were identified as important candidates by random forest feature selection with 10-fold cross-validation. The AUCs of ML models were 0.765-0.825, and discriminatory capacity was significantly improved in the artificial neural network model compared to that in the logistic regression model. CONCLUSIONS The development of hypertension can be simply and accurately predicted by each ML model using systolic blood pressure, age and FLI as selected features. By building multiple ML models, more practical prediction might be possible.
Collapse
Affiliation(s)
- Marenao Tanaka
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
- Tanaka Medical Clinic, Yoichi, Japan
| | - Yukinori Akiyama
- Department of Neurosurgery, Sapporo Medical University, Sapporo, Japan
| | - Kazuma Mori
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Immunology and Microbiology, National Defense Medical College, Tokorozawa, Japan
| | - Itaru Hosaka
- Department of Cardiovascular Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Keisuke Endo
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Toshifumi Ogawa
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Cellular Physiology and Signal Transduction, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Tatsuya Sato
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Cellular Physiology and Signal Transduction, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Toru Suzuki
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
- Natori Toru Internal Medicine and Diabetes Clinic, Natori, Japan
| | - Toshiyuki Yano
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Hirofumi Ohnishi
- Department of Public Health, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Nagisa Hanawa
- Department of Health Checkup and Promotion, Keijinkai Maruyama Clinic, Sapporo, Japan
| | - Masato Furuhashi
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| |
Collapse
|
15
|
Thulasinathan B, Suvilesh KN, Maram S, Grossmann E, Ghouri Y, Teixeiro EP, Chan J, Kaif JT, Rachagani S. The impact of gut microbial short-chain fatty acids on colorectal cancer development and prevention. Gut Microbes 2025; 17:2483780. [PMID: 40189834 PMCID: PMC11980463 DOI: 10.1080/19490976.2025.2483780] [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: 12/18/2024] [Revised: 02/18/2025] [Accepted: 03/18/2025] [Indexed: 04/11/2025] Open
Abstract
Cancer is a long-term illness that involves an imbalance in cellular and immune functions. It can be caused by a range of factors, including exposure to environmental carcinogens, poor diet, infections, and genetic alterations. Maintaining a healthy gut microbiome is crucial for overall health, and short-chain fatty acids (SCFAs) produced by gut microbiota play a vital role in this process. Recent research has established that alterations in the gut microbiome led to decreased production of SCFA's in lumen of the colon, which associated with changes in the intestinal epithelial barrier function, and immunity, are closely linked to colorectal cancer (CRC) development and its progression. SCFAs influence cancer progression by modifying epigenetic mechanisms such as DNA methylation, histone modifications, and non-coding RNA functions thereby affecting tumor initiation and metastasis. This suggests that restoring SCFA levels in colon through microbiota modulation could serve as an innovative strategy for CRC prevention and treatment. This review highlights the critical relationship between gut microbiota and CRC, emphasizing the potential of targeting SCFAs to enhance gut health and reduce CRC risk.
Collapse
Affiliation(s)
- Boobalan Thulasinathan
- Department of Veterinary Medicine and Surgery, University of Missouri, Columbia, MO, USA
- Roy Blunt NextGen Precision Health Institute, University of Missouri, Columbia, MO, USA
| | - Kanve N. Suvilesh
- Roy Blunt NextGen Precision Health Institute, University of Missouri, Columbia, MO, USA
- Department of Surgery, Ellis Fischel Cancer Centre, University of Missouri, Columbia, MO, USA
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO, USA
| | - Sumanas Maram
- Roy Blunt NextGen Precision Health Institute, University of Missouri, Columbia, MO, USA
| | - Erik Grossmann
- Department of Surgery, Ellis Fischel Cancer Centre, University of Missouri, Columbia, MO, USA
- Department of Medicine, Digestive Centre, Ellis Fischel Cancer Centre, University of Missouri, Columbia, MO, USA
| | - Yezaz Ghouri
- Department of Medicine, Digestive Centre, Ellis Fischel Cancer Centre, University of Missouri, Columbia, MO, USA
| | - Emma Pernas Teixeiro
- Roy Blunt NextGen Precision Health Institute, University of Missouri, Columbia, MO, USA
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, MO, USA
| | - Joshua Chan
- Chemical and Biological Engineering, Colorado State University, Fort Collins, CO, USA
| | - Jussuf T. Kaif
- Department of Surgery, Ellis Fischel Cancer Centre, University of Missouri, Columbia, MO, USA
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO, USA
- Siteman Cancer Centre, Washington University, St. Louis, MO, USA
| | - Satyanarayana Rachagani
- Department of Veterinary Medicine and Surgery, University of Missouri, Columbia, MO, USA
- Roy Blunt NextGen Precision Health Institute, University of Missouri, Columbia, MO, USA
- Department of Surgery, Ellis Fischel Cancer Centre, University of Missouri, Columbia, MO, USA
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO, USA
- Siteman Cancer Centre, Washington University, St. Louis, MO, USA
| |
Collapse
|
16
|
Zhu H, Tian X, Tan Z, Yang X, Zhao Y. Soybean protein isolate versus sheep whey protein: A detailed comparison of their chemical composition, gastrointestinal digestion and fermentation properties. Food Chem 2025; 483:144239. [PMID: 40215742 DOI: 10.1016/j.foodchem.2025.144239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 03/24/2025] [Accepted: 04/05/2025] [Indexed: 05/08/2025]
Abstract
Plant proteins are emerging as alternatives to animal proteins. This study compared the physicochemical properties and nutritional characteristics of soybean protein isolate (SPI) and sheep whey protein (SWP), which is gaining recognition for its nutritional benefits. SWP exhibited higher amino acid content, larger particle size, increased turbidity, and superior foaming and emulsification capacities compared to SPI (p < 0.05). In contrast, SPI demonstrated better emulsification stability. In vitro, gastrointestinal digestion showed that SPI achieved a higher degree of hydrolysis in both gastric and intestinal phases, although SWP had a faster initial hydrolysis rate. During gut microbiota fermentation, SWP produced significantly higher levels of short-chain fatty acids (SCFAs) than SPI (p < 0.05). Non-targeted metabolomics revealed distinct metabolic differences, particularly in amino acid metabolism, bile acid synthesis, and hormone biosynthesis. These findings suggest SWP is suitable for nutritional supplementation, while SPI is better for dairy-based alternatives.
Collapse
Affiliation(s)
- Haoyan Zhu
- Key Laboratory of the Ministry of Education for Medicinal Resource and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China
| | - Xinwei Tian
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China.
| | - Zhengwei Tan
- Key Laboratory of the Ministry of Education for Medicinal Resource and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China
| | - Xingbin Yang
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Yan Zhao
- Key Laboratory of the Ministry of Education for Medicinal Resource and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China.
| |
Collapse
|
17
|
Zhang KR, Nair RM, Chen Y, Jin F, Dunaief J, VanderBeek BL. Association of Age-Related Macular Degeneration with Cholelithiasis. OPHTHALMOLOGY SCIENCE 2025; 5:100771. [PMID: 40291395 PMCID: PMC12022688 DOI: 10.1016/j.xops.2025.100771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 02/27/2025] [Accepted: 03/12/2025] [Indexed: 04/30/2025]
Abstract
Purpose Dysregulated lipid metabolism likely contributes to the pathogenesis of age-related macular degeneration (AMD). There is an overlap in risk factors between AMD and diseases of lipid metabolism, such as cholelithiasis, suggesting that an association between these diseases could provide insight into AMD pathogenesis. This study sought to determine if there is an association between cholelithiasis and AMD. Design A cohort study was conducted using patients in the Optum deidentified Clinformatics Data Mart database from January 1, 2000, to June 30, 2022. Participants Patients over the age of 55 with ≥2 years of data and no prior history of AMD were included. The exposed cohort included patients who had a history of cholelithiasis, cholecystitis, or cholecystectomy. The control cohort included patients with gastroesophageal reflux disease (GERD), matched for age ±3 years, sex, race, and year of index date. Methods Propensity scores were created using multivariable logistic regression and applied to inverse probability of treatment weighting (IPTW). Cox proportional hazard regression modeling with IPTW was used to compare progression to AMD in each cohort. Main Outcome Measures Progression to AMD for patients with cholelithiasis, cholecystitis, or a history of cholecystectomy. Results A total of 332 536 patients with cholelithiasis and 776 591 matched GERD controls were analyzed. After IPTW, the mean age (±standard deviation) was 66.6 ± 9.4 years in the cholelithiasis cohort and 67.5 (±10.3) years in the GERD cohort. Women comprised 58% of the cholelithiasis cohort and 57% of the GERD cohort. In the cholelithiasis cohort, 3511.7 (1.14%) were diagnosed with AMD, compared with 23 367.1 (2.92%) in the GERD cohort and corresponding to a significantly decreased hazard of AMD (adjusted hazard ratio [aHR] = 0.72, 95% confidence interval [CI]: 0.69-0.75, P < 0.0001). In the subanalysis, before IPTW weighting, AMD developed in 3809 of 275 897 (1.4%) patients with only cholelithiasis (aHR = 0.76, 95% CI: 0.73-0.80, P < 0.0001), 335 of 47 166 (0.71%) patients with cholecystitis (aHR = 0.54, 95% CI: 0.47-0.61, P < 0.0001), and 114 of 9473 (1.20%) patients who underwent cholecystectomy (aHR = 0.50, 95% CI: 0.41-0.63, P < 0.0001). Conclusions Cholelithiasis was associated with a 28% hazard reduction in AMD. More severe gallbladder disease conferred greater protection. Financial Disclosures Proprietary or commercial disclosure may be found in the Footnotes and Disclosures at the end of this article.
Collapse
Affiliation(s)
- Kevin R. Zhang
- Department of Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Rohini M. Nair
- Department of Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Yineng Chen
- Center for Preventative Ophthalmology and Biostatistics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Fangming Jin
- Center for Preventative Ophthalmology and Biostatistics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Joshua Dunaief
- Department of Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Brian L. VanderBeek
- Department of Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
- Center for Pharmacoepidemiology Research and Training, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
- Leonard Davis Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
18
|
Zhong L, Yue Y, Liu X, Chen H, Qian Y, Tang W, Chen B, Yuan Y, Shao H. The effects of six antidepressants on electrolytes, hepatic and renal functions, and glycolipid metabolism in patients with major depressive disorder. J Affect Disord 2025; 380:734-741. [PMID: 40169121 DOI: 10.1016/j.jad.2025.03.172] [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: 01/02/2025] [Revised: 03/26/2025] [Accepted: 03/29/2025] [Indexed: 04/03/2025]
Abstract
BACKGROUND Most antidepressants with similar pharmacological characteristics exhibit comparable therapeutic efficacy but differ in side effects. Therefore, we used a retrospective design to compare biochemical changes induced by six antidepressants and identify differences among them. METHODS Case records from 1706 hospitalized patients with major depressive disorder (MDD) receiving antidepressant monotherapy were divided into six groups based on the specific antidepressants used: paroxetine, sertraline, fluoxetine, escitalopram, venlafaxine, and duloxetine. Electrolytes, hepatic and renal functions, body weight, and glycolipid metabolism were assessed at baseline and 2 weeks post-antidepressant initiation. Paired analysis was used for comparing the changes prior to and after administration within each group, and analysis of covariance was used for evaluating the distinctions among the six groups. RESULTS After 2 weeks of treatment, significant decreases in serum sodium and chloride levels were observed with venlafaxine, duloxetine, and fluoxetine, while potassium, phosphorus, and carbon dioxide concentrations tended to increase across all six antidepressants. In terms of hepatic indicators, these antidepressants significantly elevated alanine aminotransferase (ALT), aspartate aminotransferase (AST), and γ-glutamyl transpeptidase (GGT) levels, with duloxetine showing the most pronounced changes from baseline, while decreasing total and direct bilirubin. Sertraline effectively reduced uric acid, although changes in renal indicators were mild with other antidepressants. Notably, these antidepressants were associated with an unfavorable lipid profile, particularly elevated triglycerides and cholesterol, but they lowered blood glucose during the acute phase. LIMITATION Residual confounding may indirectly influence the retrospective outcomes. CONCLUSION Early biochemical changes can distinguish differences among antidepressants and guide individualized medication.
Collapse
Affiliation(s)
- Lingjun Zhong
- Department of Pharmacy, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Yingying Yue
- Department of Psychiatry and Psychosomatics, Zhongda Hospital, School of Medicine, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, Southeast University, Nanjing, China
| | - Xiaoyun Liu
- Department of Psychiatry and Psychosomatics, Zhongda Hospital, School of Medicine, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, Southeast University, Nanjing, China
| | - Hualing Chen
- Department of Biostatistics, School of Public Health, Southeast University, Nanjing, China
| | - Yongkang Qian
- Department of Biostatistics, School of Public Health, Southeast University, Nanjing, China
| | - Wei Tang
- Department of Pharmacy, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Bingwei Chen
- Department of Biostatistics, School of Public Health, Southeast University, Nanjing, China.
| | - Yonggui Yuan
- Department of Psychiatry and Psychosomatics, Zhongda Hospital, School of Medicine, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, Southeast University, Nanjing, China.
| | - Hua Shao
- Department of Pharmacy, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China.
| |
Collapse
|
19
|
Sui X, Zhao J, Yang Y, Yang Y, Li K, Wang Z, Liu Z, Lu R, Zhang G. Epidemiological Dynamics of Burden and Health Inequalities in Metabolic Dysfunction-associated Steatotic Liver Disease in Adolescents at Global, Regional, and National Levels, 1990-2021. J Clin Exp Hepatol 2025; 15:102537. [PMID: 40226388 PMCID: PMC11987614 DOI: 10.1016/j.jceh.2025.102537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 02/24/2025] [Indexed: 04/15/2025] Open
Abstract
Background Metabolic dysfunction-associated steatotic liver disease (MASLD) has become one of the major causes of chronic liver disease among adolescents. However, epidemiological studies on MASLD in adolescents are still insufficient. In this study, we aim to investigate the global burden and the trend of MASLD in adolescents from 1990 to 2021. Methods The age-standardized incidence, prevalence, mortality, and disability-adjusted life years (DALYs) of MASLD were calculated based on the Global Burden of Disease (GBD) 2021 study and stratified by sex, socio-demographic index (SDI), GBD regions, and countries. The temporal trends were examined using the average annual percentage change (AAPC) and joinpoint regression. Results From 1990 to 2021, the global trends of age-standardized incidence rate (ASIR) and age-standardized prevalence rate (ASPR) of MASLD show notable increase, and the male is significantly higher than the female in adolescents. According to the incidence and prevalence, nations with low SDI confront a higher burden of MASLD. Besides, the inequality of incidence and prevalence between different SDI regions have shrunk in 2021, but the inequality of DALYs and mortality are still exacerbated. Decomposition analysis revealed that population growth and epidemiological changes were the main reasons for the increase in the incidence of MASLD. Conclusion From 1990 to 2021, there is a significant upward trend in the incidence of MASLD among adolescents worldwide. Of particular note are male adolescents, East Asian regions, and groups living in high SDI countries.
Collapse
Affiliation(s)
- Xiaohui Sui
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250014, China
| | - Junde Zhao
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250014, China
| | - Yuxin Yang
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250014, China
| | - Yikun Yang
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250014, China
| | - Kaifeng Li
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250011, China
| | - Zuocheng Wang
- Australian National University Research School of Biology, Canberra, 2601, Australia
| | - Ziqi Liu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250011, China
| | - Ruining Lu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250011, China
| | - Guiju Zhang
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250011, China
| |
Collapse
|
20
|
Zeng L, Zhang F. A positive association between high dietary medium-chain fatty acids intake and depression: Mediation of inflammation. J Affect Disord 2025; 380:767-776. [PMID: 40180053 DOI: 10.1016/j.jad.2025.03.186] [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: 09/24/2024] [Revised: 03/27/2025] [Accepted: 03/30/2025] [Indexed: 04/05/2025]
Abstract
AIMS Our aims were to investigate the association between dietary MCFAs and depression, with a focus on potential mediating factors. METHODS A total of 11,085 participants from NHANES were included in this study. The total intake of MCFAs, including octanoic acid (C8:0), decanoic acid (C10:0), and lauric acid (C12:0), was derived, and their respective ratios to total fatty acids were calculated as exposure. Depression was defined as a score of ≥10 on the Patient Health Questionnaire-9 (PHQ-9). Weighted logistic regression was used to analyze the relationship between MCFAs and depression. The contribution of MCFAs was assessed using weighted quartile sum (WQS). C-reactive protein (CRP) was included as a potential mediator in the analysis of underlying mechanisms. RESULTS The highest quartile (Q4) of dietary MCFAs was associated with a significantly greater likelihood of depression compared to the lowest quartile (Q1) (ORtotal MCFAs: 1.36, 95 % CI: 1.07-1.65; ORC8:0: 1.46, 95 % CI: 1.15-1.77; ORC10:0: 1.38, 95 % CI: 1.06-1.60; and ORC12:0: 1.31, 95 % CI: 1.03-1.68), but no significant associations were observed for Q2 and Q3. The WQS results indicated that C12:0 contributed the most to the association between total MCFAs and depression (weight percentage: 49.1 %). CRP partially mediated the association between the Q4 of MCFAs and depression, with a mediation proportion of 7.6 % to 9.1 %. CONCLUSIONS Excessive intake of MCFAs is associated with a higher risk of depression, particularly for C12:0, with CRP partially mediating this association.
Collapse
Affiliation(s)
- Lisha Zeng
- Department of Neurology, Pingxiang People's Hospital, Pingxiang, China
| | - Fengping Zhang
- Department of Nephrology, Jiujiang City Key Laboratory of Cell Therapy, JiuJiang NO.1 People's Hospital, Jiujiang, China.
| |
Collapse
|
21
|
He M, Zhang H, Luo Z, Duan X, Zhao F, Su P, Zeng Z, Zhou L, Chen C, Qiu J. Causal link between gut microbiota and obsessive-compulsive disorder: A two-sample Mendelian randomization analysis. J Affect Disord 2025; 379:852-860. [PMID: 40056996 DOI: 10.1016/j.jad.2025.02.099] [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: 07/27/2024] [Revised: 02/25/2025] [Accepted: 02/27/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Previous studies have indicated a potential link between the gut microbiota and obsessive-compulsive disorder (OCD). However, the exact causal relationship remains uncertain. In this study, we employed a two-sample Mendelian randomization (MR) analysis to evaluate the causal connection between gut microbiota and OCD. METHODS We collected Genome-Wide Association Study (GWAS) summary data on gut microbiota (n = 18, 340) and OCD (n = 199, 169), using single nucleotide polymorphisms (SNPs) as the instrumental variable. SNPs with an F-statistic of <10 were deemed weak instrumental variables and subsequently excluded. The MR analysis was conducted using five methods: inverse variance weighting (IVW), MR Egger, weighted median, weighted mode, and simple mode. Heterogeneity and pleiotropy were assessed using Cochran's Q-test and MR Egger intercept test, while sensitivity analysis was performed using a leave-one-out approach. RESULTS The IVW analysis revealed that at the phylum level, Proteobacteria (OR = 0.545, 95%CI: 0.347-0.855, P = 0.008) served as a protective factor for OCD, whereas at the order level, Bacillales (OR = 1.327, 95%CI: 1.032-1.707, P = 0.027) was identified as a risk factor. At the family level, Ruminococcaceae (OR = 0.570, 95%CI: 0.354-0.918, P = 0.021) also acted as a protective factor. At the genus level, Bilophila (OR = 0.623, 95%CI: 0.425-0.911, P = 0.015) was a protective factor, while Eubacterium ruminantium group (OR = 1.347, 95%CI: 1.012-1.794, P = 0.041) and Lachnospiraceae UCG001 (OR = 1.384, 95%CI: 1.003-1.910, P = 0.048) were identified risk factors. Reverse MR analysis showed no significant causal relationship between OCD and the gut microbiota, with no significant heterogeneity or horizontal pleiotropy observed. CONCLUSION Our analysis suggested that specific gut microbiota might have a causal relationship with OCD, revealing potential intervention strategies for the prevention and treatment of this disorder.
Collapse
Affiliation(s)
- Mingjie He
- Department of Health Laboratory Technology, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China; Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Hongyang Zhang
- Department of Health Laboratory Technology, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China; Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Zheng Luo
- Department of Health Laboratory Technology, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China; Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Xinhao Duan
- Department of Health Laboratory Technology, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China; Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Feng Zhao
- Department of Health Laboratory Technology, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China; Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Peng Su
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China; Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Zhijun Zeng
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China; Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Lixiao Zhou
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China; Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Chengzhi Chen
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China; Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China.
| | - Jingfu Qiu
- Department of Health Laboratory Technology, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China; Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China.
| |
Collapse
|
22
|
Shen D, Su L, Ding H, Long Y, Hui C. Study on the growth and decline patterns and environmental drivers of pathogens during the stabilization process of simulated landfilling municipal solid waste. WASTE MANAGEMENT (NEW YORK, N.Y.) 2025; 201:114791. [PMID: 40222285 DOI: 10.1016/j.wasman.2025.114791] [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: 11/08/2024] [Revised: 03/18/2025] [Accepted: 04/04/2025] [Indexed: 04/15/2025]
Abstract
Waste and leachate in landfills are substantial reservoirs of pathogens, however information about the risk of pathogen contamination during the stabilization process under different landfill conditions is very limited. In this study, dynamic changes of culturable pathogens, bacteria community, and human bacterial pathogens (HBPs) during the stabilization process under different landfill conditions were investigated, and the environmental drivers were explored. Results showed that total coliforms, Enterococcus, and Staphylococcus aureus were the dominant pathogens detected in waste and leachate samples. During the landfill stabilization process, the concentration of culturable pathogens peaked at the hydrolysis-acidification stage (3.6 × 105 CFU·g-1) in the anaerobic condition, fluctuated from 4.18 × 104 to 5.35 × 105 CFU·g-1 in the anaerobic leachate-recirculation condition, and kept rising (from 4.18 × 104 to 2.12 × 106 CFU·g-1) in the micro-aerobic condition. Moreover, HBPs abundance and diversity in the waste and leachate under micro-aerobic conditions were higher than those under the other two conditions, suggesting a higher risk of pathogen contamination. Sulfate and pH were significantly (p < 0.05) correlated with the composition of bacterial communities and HBPs, likely serving as the major environmental driving factors. Additionally, the interactions between HBPs and functional bacterial groups tended towards cooperative symbiotic relationships, with hydrolytic-acidogenic bacteria promoting the growth and proliferation of most pathogens. These findings will help to understand the changes and environmental drivers of pathogens during landfill stabilization, which will provide a theoretical basis for the risk prevention and control of pathogens in waste disposal.
Collapse
Affiliation(s)
- Dongsheng Shen
- Zhejiang Provincial Key Laboratory of Solid Waste Treatment and Recycling, Zhejiang Engineering Research Center of Non-ferrous Metal Waste Recycling, School of Environmental Science and Engineering, Zhejiang Gongshang University, Hangzhou 310012, China
| | - Liya Su
- Zhejiang Provincial Key Laboratory of Solid Waste Treatment and Recycling, Zhejiang Engineering Research Center of Non-ferrous Metal Waste Recycling, School of Environmental Science and Engineering, Zhejiang Gongshang University, Hangzhou 310012, China
| | - Hening Ding
- Zhejiang Provincial Key Laboratory of Solid Waste Treatment and Recycling, Zhejiang Engineering Research Center of Non-ferrous Metal Waste Recycling, School of Environmental Science and Engineering, Zhejiang Gongshang University, Hangzhou 310012, China
| | - Yuyang Long
- Zhejiang Provincial Key Laboratory of Solid Waste Treatment and Recycling, Zhejiang Engineering Research Center of Non-ferrous Metal Waste Recycling, School of Environmental Science and Engineering, Zhejiang Gongshang University, Hangzhou 310012, China
| | - Cai Hui
- Zhejiang Provincial Key Laboratory of Solid Waste Treatment and Recycling, Zhejiang Engineering Research Center of Non-ferrous Metal Waste Recycling, School of Environmental Science and Engineering, Zhejiang Gongshang University, Hangzhou 310012, China.
| |
Collapse
|
23
|
Adıgüzel E, Yılmaz ŞG, Atabilen B, Şeref B. Microbiome modulation as a novel therapeutic modality for anxiety disorders: A review of clinical trials. Behav Brain Res 2025; 487:115595. [PMID: 40246176 DOI: 10.1016/j.bbr.2025.115595] [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: 11/18/2024] [Revised: 03/08/2025] [Accepted: 04/14/2025] [Indexed: 04/19/2025]
Abstract
Anxiety disorders are one of the major conditions in psychiatry characterized by symptoms such as worry, social and performance fears, unexpected and/or triggered panic attacks, anticipatory anxiety, and avoidance behaviors. Recent developments have drawn attention to the putative involvement of peripheral systems in the control of anxiety, and the gut microbiota has come to light as an emerging peripheral target for anxiety. The relationship between the gut-brain axis, a bidirectional communication network between the central nervous system (CNS) and enteric nervous system (ENS), and anxiety has been the subject of some recent studies. Therefore, this systematic review analyzed clinical trials evaluating the potential of microbiome modulation methods in mitigating and ameliorating anxiety disorders. Clinical studies on probiotic, prebiotic, synbiotic supplements, dietary interventions, and fecal microbiota transplantation in anxiety disorders were screened. All of the studies examined the effects of probiotic intervention. One of these studies compared a prebiotic-rich diet with probiotic supplementation. Longitudinal analyses showed that the probiotic intervention alleviated anxiety. However, most of the controlled studies reported that the probiotic intervention did not make a difference compared to placebo. Thus, the current findings suggest that it is too early to consider the promising role of microbiome modulation in the treatment of anxiety disorders. However, it is obvious that more clinical research is needed to clarify issues such as probiotic strains, prebiotic types, and their doses that may be effective on anxiety disorders.
Collapse
Affiliation(s)
- Emre Adıgüzel
- Karamanoğlu Mehmetbey University, Faculty of Health Sciences, Department of Nutrition and Dietetics, Karaman, Turkey.
| | - Şemsi Gül Yılmaz
- Karamanoğlu Mehmetbey University, Faculty of Health Sciences, Department of Nutrition and Dietetics, Karaman, Turkey.
| | - Büşra Atabilen
- Karamanoğlu Mehmetbey University, Faculty of Health Sciences, Department of Nutrition and Dietetics, Karaman, Turkey.
| | - Betül Şeref
- Karamanoğlu Mehmetbey University, Faculty of Health Sciences, Department of Nutrition and Dietetics, Karaman, Turkey.
| |
Collapse
|
24
|
Guo N, Niu S, Geng Y, Shan G, Wei N, Chen H. Non-destructive quantification of low colchicine concentrations in commercially available tablets using transmission raman spectroscopy with partial least squares. Int J Pharm X 2025; 9:100321. [PMID: 40094145 PMCID: PMC11910358 DOI: 10.1016/j.ijpx.2025.100321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 02/10/2025] [Accepted: 02/20/2025] [Indexed: 03/19/2025] Open
Abstract
The narrow therapeutic index and significant toxicity of colchicine (COL) underscore the importance of content uniformity of dosage units to ensure drug safety and efficacy. In this study, transmission Raman spectroscopy (TRS) technology combined with partial least squares (PLS) regression was used for the non-destructive determination of low concentration levels of COL in commercial tablets (0.83 % w/w). Based on a multifactor orthogonal design of experiment, one hundred calibration tablets ranging in drug content from 70 % to 130 % of the label claim were manufactured to develop an initial model which was further calibrated using the HPLC results. The quantitative model displayed good repeatability and high accuracy with a root-mean-standard error for calibration of 0.038 % and root-mean-standard error for cross-validation of 0.039 %. The limits of detection and quantification were 0.13 % and 0.40 % w/w, respectively. The absolute value of relative error of the TRS and HPLC content results for commercial tablets varied between 0 and 3.8 %. Notably, the relative standard deviation (RSD) of the TRS method was 1.2 %, lower than the RSD of 2.9 % observed with HPLC. The results demonstrated a fast and non-destructive method for the quality control of highly toxic and low content active pharmaceutical ingredients in commercial products, without human or environmental exposure to toxic substances during sample preparation.
Collapse
Affiliation(s)
- Ningzi Guo
- National Institutes for Food and Drug Control, No. 31, Huatuo Road, Beijing 102629, China
| | - Sijing Niu
- National Institutes for Food and Drug Control, No. 31, Huatuo Road, Beijing 102629, China
| | - Ying Geng
- National Institutes for Food and Drug Control, No. 31, Huatuo Road, Beijing 102629, China
| | - Guangzhi Shan
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 1, Tian Tan Xi Li, Beijing 100050, China
| | - Ningyi Wei
- National Institutes for Food and Drug Control, No. 31, Huatuo Road, Beijing 102629, China
| | - Hua Chen
- National Institutes for Food and Drug Control, No. 31, Huatuo Road, Beijing 102629, China
| |
Collapse
|
25
|
Xu HS, Chen Y, Lin YJ, Eldefrawy F, Kramer NE, Siracusa JS, Kong F, Guo TL. Nanocellulose dysregulated glucose homeostasis in female mice on a Western diet: The role of gut microbiome. Life Sci 2025; 370:123567. [PMID: 40113076 DOI: 10.1016/j.lfs.2025.123567] [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: 11/14/2024] [Revised: 03/12/2025] [Accepted: 03/16/2025] [Indexed: 03/22/2025]
Abstract
There is currently increased interest in nanocellulose as a food emulsifier and dietary supplement. It was hypothesized that nanocellulose could modulate behaviors and glucose homeostasis in female mice using mechanisms of altered gut microbiome and immune modulation. An initial experiment was conducted with the objective of examining whether three common types of nanocellulose affected the gut microbiome of female C57BL/6 mice on a Western diet. Cellulose nanofibrils (CNF), TEMPO-CNF and cellulose nanocrystals were administered at the physiologically relevant dose of 30 mg/kg/day for 30 days by gavage, with cellulose and water groups as the positive and negative controls, respectively. Findings suggested that CNF had the strongest effect on the gut microbiome. CNF was therefore selected for a chronic 6-month study on the gut microbiome, immune system and behaviors in female NOD mice, a model for type 1 diabetes. Gut microbiome analysis suggested that there might be some beneficial changes following subchronic exposure (e.g., at the two-month timepoint), however, this effect was no longer seen after chronic consumption (e.g., at the six-month timepoint). CNF treatment also altered the immune homeostasis, including decreases in the splenic Mac-3+ population and serum level of proinflammatory chemokine LIX. Additionally, CNF consumption decreased diabetic incidences but had no effect on the depressive-like behavior and grip strength. However, further analysis, e.g., the insulin tolerance test, indicated that CNF-treated NOD mice might exhibit signs of insulin resistance. Taken together, nanocellulose dysregulated glucose homeostasis in female mice on a Western diet involving mechanisms related to alteration of the gut microbiome.
Collapse
Affiliation(s)
| | - Yingjia Chen
- Department of Veterinary Biomedical Sciences, USA
| | - Yu-Ju Lin
- Department of Pharmaceutical and Biomedical Sciences, USA
| | | | - Naomi E Kramer
- Department of Pharmaceutical and Biomedical Sciences, USA
| | | | - Fanbin Kong
- Department of Food Science and Technology, University of Georgia, Athens, GA 30602, USA
| | - Tai L Guo
- Department of Veterinary Biomedical Sciences, USA.
| |
Collapse
|
26
|
Duan H, Yang T, Li C, Xu N, Wang M, Zhang C, Zhao J, Chen Y, Yan J, Li W, Li Z, Ma F, Huang G. Supplementation of medium-chain triglycerides combined with docosahexaenoic acid improves cognitive function in Chinese older adults with mild cognitive impairment: A randomized double-blind, placebo-controlled trial. J Affect Disord 2025; 378:263-270. [PMID: 40044083 DOI: 10.1016/j.jad.2025.03.008] [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: 11/10/2024] [Revised: 02/26/2025] [Accepted: 03/01/2025] [Indexed: 03/21/2025]
Abstract
BACKGROUND Medium-chain triglycerides (MCT) and docosahexaenoic acid (DHA) could affect cognitive function, but their combination effects remain unclear. This randomized, double-blind, placebo-controlled trial aimed to evaluate effects of MCT and DHA supplementation, alone or in combination, on mild cognitive impairment (MCI) patients. METHODS A total of 280 MCI participants were randomly assigned to the placebo group, MCT group (14 g/d octanoic acid+10 g/d capric acid), DHA group (800 mg/d) and MCT + DHA (14 g/d octanoic acid, 10 g/d capric acid and 800 mg/d DHA) group, 70 individuals per group for 12 months. Cognitive function was assessed at baseline, 6 months and 12 months; blood indicators were analyzed at baseline and 12 months. This study is registered with Chinese Clinical Trial Registry (ChiCTR2200059641). RESULTS After 12 months intervention, compared with the placebo group, MCT group, DHA group and MCT + DHA group had statistically significant improvements in full scale IQ (β: 0.107, 95 % CI: 0.006, 0.208; β: 0.135, 95 % CI: 0.034, 0.235; β: 0.136, 95 % CI: 0.035, 0.237), serum total ketone body levels (β: 10.540, 95 % CI: 9.550, 11.531; β: 5.884, 95 % CI: 4.894, 6.875; β: 13.186, 95 % CI: 12.196, 14.176), and decreases in mtDNA deletions (β: -0.370, 95 % CI: -0.648, -0.092; β: -0.335, 95 % CI: -0.613, -0.056; β: -0.427, 95 % CI: -0.705, -0.148) and plasma Aβ42 levels (β: -3.318, 95 % CI: -5.571, -1.064; β: -3.218, 95 % CI: -5.472, -0.965; β: -2.906, 95 % CI: -5.160, -0.653). CONCLUSIONS Supplementation of MCT, DHA and their combination for 12 months can significantly improve cognitive function, mitochondria function and increase serum total ketone body levels in MCI individuals. Combined intervention was more beneficial than MCT or DHA alone.
Collapse
Affiliation(s)
- Huilian Duan
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin, China; Key Laboratory of Prevention and Control of Major Diseases in the Population, Ministry of Education, School of Public Health, Tianjin Medical University, Tianjin, China; Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China
| | - Tong Yang
- Department of Epidemiology & Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China; Key Laboratory of Prevention and Control of Major Diseases in the Population, Ministry of Education, School of Public Health, Tianjin Medical University, Tianjin, China; Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China
| | - Chenyu Li
- Department of Epidemiology & Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China; Key Laboratory of Prevention and Control of Major Diseases in the Population, Ministry of Education, School of Public Health, Tianjin Medical University, Tianjin, China; Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China
| | - Ning Xu
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin, China; Key Laboratory of Prevention and Control of Major Diseases in the Population, Ministry of Education, School of Public Health, Tianjin Medical University, Tianjin, China; Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China
| | - Moyan Wang
- Community Health Service Center, Sanhuailu Street, Binhai New District, Tianjin, China
| | - Chunlai Zhang
- Community Health Service Center, Sanhuailu Street, Binhai New District, Tianjin, China
| | - Jiangang Zhao
- Community Health Service Center, Sanhuailu Street, Binhai New District, Tianjin, China
| | - Yongjie Chen
- Department of Epidemiology & Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China; Key Laboratory of Prevention and Control of Major Diseases in the Population, Ministry of Education, School of Public Health, Tianjin Medical University, Tianjin, China; Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China
| | - Jing Yan
- Department of Social Medicine and Health Management, School of Public Health, Tianjin Medical University, Tianjin, China; Key Laboratory of Prevention and Control of Major Diseases in the Population, Ministry of Education, School of Public Health, Tianjin Medical University, Tianjin, China; Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China
| | - Wen Li
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin, China; Key Laboratory of Prevention and Control of Major Diseases in the Population, Ministry of Education, School of Public Health, Tianjin Medical University, Tianjin, China; Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China
| | - Zhenshu Li
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin, China; Key Laboratory of Prevention and Control of Major Diseases in the Population, Ministry of Education, School of Public Health, Tianjin Medical University, Tianjin, China; Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China
| | - Fei Ma
- Department of Epidemiology & Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China; Key Laboratory of Prevention and Control of Major Diseases in the Population, Ministry of Education, School of Public Health, Tianjin Medical University, Tianjin, China; Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China.
| | - Guowei Huang
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin, China; Key Laboratory of Prevention and Control of Major Diseases in the Population, Ministry of Education, School of Public Health, Tianjin Medical University, Tianjin, China; Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China; The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin, China.
| |
Collapse
|
27
|
Monsalve DM, Acosta-Ampudia Y, Acosta NG, Celis-Andrade M, Şahin A, Yilmaz AM, Shoenfeld Y, Ramírez-Santana C. NETosis: A key player in autoimmunity, COVID-19, and long COVID. J Transl Autoimmun 2025; 10:100280. [PMID: 40071133 PMCID: PMC11894324 DOI: 10.1016/j.jtauto.2025.100280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
NETosis, the process through which neutrophils release neutrophil extracellular traps (NETs), has emerged as a crucial mechanism in host defense and the pathogenesis of autoimmune responses. During the SARS-CoV-2 pandemic, this process received significant attention due to the central role of neutrophil recruitment and activation in infection control. However, elevated neutrophil levels and dysregulated NET formation have been linked to coagulopathy and endothelial damage, correlating with disease severity and poor prognosis in COVID-19. Moreover, it is known that SARS-CoV-2 can induce persistent low-grade systemic inflammation, known as long COVID, although the underlying causes remain unclear. It has been increasingly acknowledged that excessive NETosis and NET generation contribute to further pathophysiological abnormalities following SARS-CoV-2 infection. This review provides an updated overview of the role of NETosis in autoimmune diseases, but also the relationship between COVID-19 and long COVID with autoimmunity (e.g., latent and overt autoimmunity, molecular mimicry, epitope spreading) and NETosis (e.g., immune responses, NET markers). Finally, we discuss potential therapeutic strategies targeting dysregulated NETosis to mitigate the severe complications of COVID-19 and long COVID.
Collapse
Affiliation(s)
- Diana M. Monsalve
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Yeny Acosta-Ampudia
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Nicolás Guerrero Acosta
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Mariana Celis-Andrade
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Ali Şahin
- Selcuk University, Faculty of Medicine, Konya, Turkiye
| | - Ahsen Morva Yilmaz
- TUBITAK Marmara Research Center (TUBITAK-MAM), Life Sciences, Medical Biotechnology Unit, Kocaeli, Turkiye
| | - Yehuda Shoenfeld
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Reichman University, Herzelia, Israel
| | - Carolina Ramírez-Santana
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| |
Collapse
|
28
|
Luo F, Yang J, Song Z, Zhao Y, Wang P, Liu K, Mou X, Liu W, Li W. Renshen Zhuye decoction ameliorates high-fat diet-induced obesity and insulin resistance by modulating gut microbiota and metabolic homeostasis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156655. [PMID: 40120542 DOI: 10.1016/j.phymed.2025.156655] [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: 01/20/2025] [Revised: 03/09/2025] [Accepted: 03/15/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Obesity, characterized by excessive adipose tissue accumulation, has become a global health challenge with rapidly increasing prevalence. It contributes significantly to metabolic disorders including insulin resistance (IR). Renshen-zhuye decoction (RZD), a traditional Chinese medicine formula historically used for diabetes, shows potential for improving metabolic parameters, but its effects and mechanisms in obesity and insulin resistance remain unclear. PURPOSE This study aimed to evaluate the therapeutic benefits of RZD on obesity and insulin resistance, and to elucidate the underlying mechanisms through which it improves glucose and lipid metabolism. METHODS The role of RZD was evaluated in a high-fat diet (HFD) mouse model. The formula was characterized using UPLC-MS. Comprehensive analyses including histopathological staining, immunofluorescence, biochemical assays, 16S rRNA gene sequencing of gut microbiota, and non-targeted metabolomic analysis were performed. To validate the role of gut microbiota, we employed antibiotic treatment (ABX) to deplete intestinal flora and conducted fecal microbiota transplantation (FMT) experiments. RESULTS RZD treatment dose-dependently alleviated HFD-induced dyslipidemia and insulin resistance, improving glucose tolerance, insulin sensitivity, and energy expenditure. Gut microbiota analysis revealed that RZD significantly modulated the composition of intestinal flora and their metabolic profiles. Additionally, RZD reduced intestinal and systemic inflammation by enhancing intestinal barrier integrity, particularly through increased expression of tight junction proteins such as Occludin. Importantly, the beneficial effects of RZD on weight management and glucose homeostasis were antagonized by antibiotic intervention, while FMT experiments confirmed that these improvements were mediated through gut microbiota modulation. CONCLUSION This study provides new insights into RZD's modulatory effects on gut microbiota and subsequent improvements in obesity-related metabolic parameters. RZD alleviates HFD-induced obesity and insulin resistance in mice by modulating gut microbiota composition and function, which subsequently improves intestinal barrier integrity, reduces inflammation, and enhances metabolic homeostasis.
Collapse
Affiliation(s)
- Fei Luo
- School of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, PR China
| | - Jie Yang
- School of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, PR China
| | - Zhiping Song
- School of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, PR China
| | - Yuan Zhao
- School of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, PR China
| | - Panpan Wang
- Hangzhou Linping Hospital of Traditional Chinese Medicine, Hangzhou 310000, PR China
| | - Kaiyuan Liu
- Department of Endocrinology, Zhejiang Integrated Traditional Chinese and Western Medicine Hospital, Hangzhou, 310000, PR China
| | - Xin Mou
- Department of Endocrinology, Zhejiang Integrated Traditional Chinese and Western Medicine Hospital, Hangzhou, 310000, PR China.
| | - Wenhong Liu
- School of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, PR China.
| | - Wei Li
- School of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, PR China.
| |
Collapse
|
29
|
Pradeepkiran JA, Islam MA, Sehar U, Reddy AP, Vijayan M, Reddy PH. Impact of diet and exercise on mitochondrial quality and mitophagy in Alzheimer's disease. Ageing Res Rev 2025; 108:102734. [PMID: 40120948 DOI: 10.1016/j.arr.2025.102734] [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: 09/10/2024] [Revised: 11/26/2024] [Accepted: 03/15/2025] [Indexed: 03/25/2025]
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disorder that affects millions of people worldwide. It is characterized by the accumulation of beta-amyloid and phosphorylated tau, synaptic damage, and mitochondrial abnormalities in the brain, leading to the progressive loss of cognitive function and memory. In AD, emerging research suggests that lifestyle factors such as a healthy diet and regular exercise may play a significant role in delaying the onset and progression of the disease. Mitochondria are often referred to as the powerhouse of the cell, as they are responsible for producing the energy to cells, including neurons to maintain cognitive function. Our article elaborates on how mitochondrial quality and function decline with age and AD, leading to an increase in oxidative stress and a decrease in ATP production. Decline in mitochondrial quality can impair cellular functions contributing to the development and progression of disease with the loss of neuronal functions in AD. This article also covered mitophagy, the process by which damaged or dysfunctional mitochondria are selectively removed from the cell to maintain cellular homeostasis. Impaired mitophagy has been implicated in the progression and pathogenesis of AD. We also discussed the impact of impaired mitophagy implicated in AD, as the accumulation of damaged mitochondria can lead to increased oxidative stress. We expounded how dietary interventions and exercise can help to improve mitochondrial quality, and mitochondrial function and enhance mitophagy in AD. A diet rich in antioxidants, polyphenols, and mitochondria-targeted small molecules has been shown to enhance mitochondrial function and protect against oxidative stress, particularly in neurons with aged and mild cognitively impaired subjects and AD patients. Promoting a healthy lifestyle, mainly balanced diet and regular exercise that support mitochondrial health, in an individual can potentially delay the onset and progression of AD. In conclusion, a healthy diet and regular exercise play a crucial role in maintaining mitochondrial quality and mitochondrial function, in turn, enhancing mitophagy and synaptic activities that delay AD in the elderly populations.
Collapse
Affiliation(s)
| | - Md Ariful Islam
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Ujala Sehar
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Arubala P Reddy
- Nutritional Sciences Department, College Human Sciences, Texas Tech University, Lubbock, TX, USA
| | - Murali Vijayan
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Nutritional Sciences Department, College Human Sciences, Texas Tech University, Lubbock, TX, USA; Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Department of Public Health, Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Department of Speech, Language, and Hearing Sciences, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
| |
Collapse
|
30
|
Li P, Wang S, Li J, Xiao Z, Zhu H, Sheng D, Liu W, Xiao B, Zhou L. Appraising the Effects of Gut Microbiota on Insomnia Risk Through Genetic Causal Analysis. Am J Med Genet B Neuropsychiatr Genet 2025; 198:e33021. [PMID: 39754389 DOI: 10.1002/ajmg.b.33021] [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: 01/09/2024] [Revised: 11/12/2024] [Accepted: 12/16/2024] [Indexed: 01/06/2025]
Abstract
BackgroundInsomnia is a common neurological disorder that exhibits connections with the gut microbiota; however, the exact causal relationship remains unclear. MethodsWe conducted a Mendelian randomization (MR) study to systematically evaluate the causal effects of genus-level gut microbiota on insomnia risk in individuals of European ancestry. Summary-level datasets on gut microbiota were sourced from the genome-wide association study (GWAS) of MiBioGen, while datasets on insomnia were obtained from the GWAS of Neale Lab and FinnGen. The primary analytical approach used was the inverse-variance weighted (IVW) method, supplemented by MR-Egger, maximum likelihood, MR-robust adjusted profile score, and weighted median. Sensitivity analyses were conducted to ensure robustness. ResultsThe microbial taxa Enterorhabdus, Family XIII AD3011 group, Paraprevotella, and Lachnospiraceae UCG004 were associated with an increased risk of insomnia, whereas Coprococcus1, Coprobacter, Desulfovibrio, Flavonifractor, Olsenella, Odoribacter, and Oscillibacter were linked to a decreased risk. Regarding the insomnia phenotype characterized by trouble falling asleep, the microbial taxon Eisenbergiella was correlated with an increased risk, while Haemophilus and the Eubacterium brachy group were associated with a reduced risk. Furthermore, for the insomnia phenotype characterized by waking too early, the microbial taxa Family XIII UCG001, Lachnospiraceae FCS020 group, and Olsenella were linked to an increased risk, whereas the Eubacterium brachy group and Victivallis were associated with a lower risk. The results remained robust across all sensitivity analyses. ConclusionOur MR study identified multiple genus-level gut microbial taxa that may exhibit potential causal effects on insomnia from a genetic perspective. These findings provide evidence supporting the theory of the microbiota-gut-brain axis and offer new insights into potential prevention and therapeutic targets for insomnia.
Collapse
Affiliation(s)
- Peihong Li
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Song Wang
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jiaxin Li
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zheng Xiao
- Department of Pathology, First Hospital of Changsha, Changsha, Hunan, China
| | - Haoyue Zhu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Dandan Sheng
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Weiping Liu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Bo Xiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Luo Zhou
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
31
|
Shaffer A, Meyerowitz EA. Clinical Manifestations of SARS-CoV-2 Infection in Immunocompetent Adults in the Era of Widespread Population Immunity and Omicron Sublineage Viruses. Infect Dis Clin North Am 2025; 39:233-251. [PMID: 40068975 DOI: 10.1016/j.idc.2025.02.002] [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] [Indexed: 05/10/2025]
Abstract
While most SARS-CoV-2 infections and reinfections in the era of widespread population immunity with omicron subsub-lineage variants are mild for immunocompetent individuals, any manifestation previously seen during the pandemic phase is still possible. COVID-19 may affect any organ system. Previous infections and prior vaccines protect against symptomatic future SARS-CoV-2 infections, though this protection wanes over time.
Collapse
Affiliation(s)
- Alexander Shaffer
- Division of Infectious Diseases, Department of Medicine, Montefiore Medical Center, 111 East 210th Street, Bronx, NY 10467, USA; Albert Einstein College of Medicine, Bronx, NY, USA
| | - Eric A Meyerowitz
- Division of Infectious Diseases, Department of Medicine, Montefiore Medical Center, 111 East 210th Street, Bronx, NY 10467, USA; Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
32
|
Wang W, Zhang K, Zhang K, Wu R, Tang Y, Li Y. Gut microbiota promotes cholesterol gallstone formation through the gut-metabolism-gene axis. Microb Pathog 2025; 203:107446. [PMID: 40118296 DOI: 10.1016/j.micpath.2025.107446] [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: 06/17/2024] [Revised: 02/17/2025] [Accepted: 02/28/2025] [Indexed: 03/23/2025]
Abstract
BACKGROUND Gallstone disease, arising from the interplay between host metabolism and gut microbiota, represents a significant health concern. Dysbiosis of the gut microbiome and disruptions in circadian rhythm contribute to the pathogenesis of gallstones. This study conducted a comprehensive analysis of gut microbiota and metabolites derived from stool and serum samples of 28 patients with cholesterol gallstones (CGS) and 19 healthy controls, employing methodologies such as 16S rRNA sequencing, metaproteomics, metabolomics, and host genetic analysis. Additionally, a retrospective cohort study was utilized to assess the efficacy of probiotics or ursodeoxycholic acid (UDCA) in preventing CGS formation post-bariatric surgery. RESULTS In CGS patients, gut microbiota diversity shifted, with harmful bacteria rising and beneficial ones declining. The altered microbiota primarily affected amino acid, lipid, nucleotide, and carbohydrate metabolism. Metabolic abnormalities were noted in amino acids, glucose, lipids, and bile acids with decreased levels of ursodeoxycholic, glycosodeoxycholic, and glycolithocholic acids, and increased glycohyodeoxycholic and allocholic acids. Glutamine and alanine levels dropped, while phenylalanine and tyrosine rosed. Animal studies confirmed gene changes in gallbladder tissues related to bile acid, energy, glucose, and lipid metabolism. Importantly, UDCA and probiotics effectively reduced CGS risk post-bariatric surgery, especially when combined. CONCLUSIONS Multi-omics can clarify CGS pathology, by focusing on the gut-metabolism-gene axis, paving the way for future studies on CGS prevention and treatment through gut microbiota or metabolic interventions.
Collapse
Affiliation(s)
- Wei Wang
- Department of Interventional, The Second Hospital of Shandong University, Shandong, 250033, China
| | - Kai Zhang
- Department of Hepatobiliary Surgery, Shandong Provincial Third Hospital, Shandong University, Shandong, 250033, China
| | - Kun Zhang
- Shanghai Biotree Biotech Co., Ltd., Shanghai, China
| | - Rui Wu
- Department of Hepatobiliary Surgery, Shandong Provincial Third Hospital, Shandong University, Shandong, 250033, China
| | - Yu Tang
- Department of Geriatrics, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| | - Yuliang Li
- Department of Interventional, The Second Hospital of Shandong University, Shandong, 250033, China.
| |
Collapse
|
33
|
Shahriar SMS, Haque N, Hasan T, Sufal MTA, Hassan MT, Hasan M, Salam SM. Heavy metal pollution in poultry feeds and broiler chickens in Bangladesh. Toxicol Rep 2025; 14:101932. [PMID: 39968052 PMCID: PMC11833629 DOI: 10.1016/j.toxrep.2025.101932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 01/18/2025] [Accepted: 01/25/2025] [Indexed: 02/20/2025] Open
Abstract
The poultry industry poses a significant threat of heavy metal poisoning for the people of Bangladesh. The research was performed to assess the levels of heavy metals in chicken feed as well as other consumable sections of poultry fowl, and to determine the possible health hazards implicated. The levels of seven metals were evaluated in sixteen commercially available poultry feeds and in three edible portions of chicken obtained from several local markets in Rajshahi city. The metal concentrations were investigated via an atomic absorption spectrophotometer following the wet digestion method. The amount of Cr, Cd, Pb, Cu, Mn, Ni, and Fe in poultry feeds were observed from 0.03 to 12.85 mg/kg, 0.01-1.64 mg/kg, 0.15-4.21 mg/kg, 2.65-45.83 mg/kg, 22.63-188.85 mg/kg, 0.09-2.64 mg/kg, and 0.54-41.01 mg/kg, respectively. In broiler chickens, the concentrations were determined from 0.87 to 3.15 mg/kg, 0.01-0.05 mg/kg, 0.19-1.09 mg/kg, 0.96-3.78 mg/kg, 4.45-23.53 mg/kg, 0.07-0.56 mg/kg, and 2.70-92.32 mg/kg, respectively. With the exception of Cu, Mn, and Fe, most heavy metal concentrations in chickens exceeded the highest allowed concentration set by FAO/WHO. The estimated EDI, THQ and TTHQ numbers for all metals examined were found to be below MTDI, indicating that consumption of chicken meat poses noncarcinogenic risk to individuals. Comparatively, ILCR associated with Cd and Pb are around the safety threshold, but Cr exceeds the permissible range and poses a significant risk.
Collapse
Affiliation(s)
- Sha Md. Shahan Shahriar
- Department of Applied Chemistry and Chemical Engineering, University of Rajshahi, Rajshahi 6205, Bangladesh
| | - Nazmul Haque
- Department of Applied Chemistry and Chemical Engineering, University of Rajshahi, Rajshahi 6205, Bangladesh
| | - Tafsir Hasan
- Department of Applied Chemistry and Chemical Engineering, University of Rajshahi, Rajshahi 6205, Bangladesh
| | - Md Tasif Amir Sufal
- Department of Applied Chemistry and Chemical Engineering, University of Rajshahi, Rajshahi 6205, Bangladesh
| | - Mohammad Tariqul Hassan
- Institute of Food Science and Technology (IFST), Bangladesh Council of Scientific and Industrial Research (BCSIR), Dhaka, Bangladesh
| | - Mahfujul Hasan
- Institute of Food Science and Technology (IFST), Bangladesh Council of Scientific and Industrial Research (BCSIR), Dhaka, Bangladesh
| | - Sayed M.A. Salam
- Department of Applied Chemistry and Chemical Engineering, University of Rajshahi, Rajshahi 6205, Bangladesh
| |
Collapse
|
34
|
Xu C, Ezzi SHA, Zou X, Dong Y, Alhaskawi A, Zhou H, Kota VG, Abdulla MHAH, Abdalbary SA, Lu H. The role of TNF in metabolic disorders and liver diseases. Cytokine 2025; 190:156933. [PMID: 40174483 DOI: 10.1016/j.cyto.2025.156933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 03/23/2025] [Accepted: 03/26/2025] [Indexed: 04/04/2025]
Abstract
Tumor necrosis factor (TNF) is identified as a pro-inflammatory cytokine critical to the pathology of liver disease. In the carbohydrate metabolism, TNF has been demonstrated to impede the insulin signaling pathway, thereby precipitating glucose intolerance and insulin resistance. In lipid metabolism, TNF upregulates genes implicated in fatty acid synthesis, resulting in increased lipid accumulation within the liver. In amino acid metabolism, TNF has shown to promote the gene expression for amino acid catabolism, leading to decreased protein synthesis. Additionally, TNF stimulates the production of other chemokines and inflammatory cytokines that can further exacerbate liver injury. Overall, TNF is crucial in developing liver diseases by disrupting various metabolic pathways in the liver, causing insulin resistance, lipid accumulation, and decreased protein synthesis. This review summarizes the present understanding of TNF's role in the regulation of carbohydrate, lipid and amino acid metabolism in liver disease together with its potential therapeutic implications.
Collapse
Affiliation(s)
- Chuze Xu
- School of Optometry and Ophthalmology, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | | | - Xiaodi Zou
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Yanzhao Dong
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Ahmad Alhaskawi
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Haiying Zhou
- Faculty of Medicine, The Chinese University of Hong Kong School of Biomedical Science, Hong Kong, China
| | | | | | - Sahar Ahmed Abdalbary
- Department of Orthopedic Physical Therapy, Faculty of Physical Therapy, Nahda University, Beni Suef, Egypt
| | - Hui Lu
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University, Hangzhou, China; Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Zhejiang University, Hangzhou, China.
| |
Collapse
|
35
|
Wang C, Fan S, Li M, Ye Y, Li Z, Long W, Li Y, Huang Z, Jiang Q, Yang W, Yang R, Tang D. A 7-year feed study on the long-term effects of genetically modified maize containing cry1Ab/cry2Aj and EPSPS genes on gut microbiota and metabolite profiles across two generations of cynomolgus macaques. Food Chem Toxicol 2025; 200:115419. [PMID: 40157594 DOI: 10.1016/j.fct.2025.115419] [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: 12/18/2024] [Revised: 03/26/2025] [Accepted: 03/26/2025] [Indexed: 04/01/2025]
Abstract
The health implications of genetically modified (GM) crops remain controversial relative to their non-GM counterparts, particularly regarding long-term dietary exposure. Although the gut microbiome is a key health indicator, studies investigating the impact of GM crop consumption on intestinal microbiota remain limited. This study presents a comprehensive 7-year evaluation of GM maize expressing cry1Ab/cry2Aj and G10evo-EPSPS proteins through metagenomic and metabolomic analyses. We assessed the effects of GM maize consumption on gut microbiota diversity and metabolite profiles in cynomolgus macaques (Macaca fascicularis) compared with non-GM maize. Three diet regimens were implemented: a conventional compound feed (CK group), diet formulation containing 70 % non-GM maize (Corn group), and diet formulation containing 70 % GM maize (Tg group). The results demonstrated that feeding GM maize to the first (F0) and second (F1) generations of monkeys did not substantially affect the composition, community structure, or function of the intestinal microbiome, as indicated by species composition and diversity analyses. Minor differences in intestinal metabolites were observed but were not directly linked to transgenic maize consumption. Collectively, long-term intake of maize with cry1Ab/cry2Aj and g10evo-epsps genes had no adverse effects on macaques or their offspring.
Collapse
Affiliation(s)
- Chenyun Wang
- Medical Primate Research Center, Drug Safety Evaluation Center, Institute of Medical Biology, Chinese Academy of Medical Sciences/Peking Union Medical College, Kunming, 650118, China
| | - Shengtao Fan
- Medical Primate Research Center, Drug Safety Evaluation Center, Institute of Medical Biology, Chinese Academy of Medical Sciences/Peking Union Medical College, Kunming, 650118, China
| | - Minghao Li
- Medical Primate Research Center, Drug Safety Evaluation Center, Institute of Medical Biology, Chinese Academy of Medical Sciences/Peking Union Medical College, Kunming, 650118, China
| | - Yousong Ye
- Medical Primate Research Center, Drug Safety Evaluation Center, Institute of Medical Biology, Chinese Academy of Medical Sciences/Peking Union Medical College, Kunming, 650118, China
| | - Zheli Li
- Medical Primate Research Center, Drug Safety Evaluation Center, Institute of Medical Biology, Chinese Academy of Medical Sciences/Peking Union Medical College, Kunming, 650118, China
| | - Weihu Long
- Medical Primate Research Center, Drug Safety Evaluation Center, Institute of Medical Biology, Chinese Academy of Medical Sciences/Peking Union Medical College, Kunming, 650118, China
| | - Yongjie Li
- Medical Primate Research Center, Drug Safety Evaluation Center, Institute of Medical Biology, Chinese Academy of Medical Sciences/Peking Union Medical College, Kunming, 650118, China
| | - Zhangqiong Huang
- Medical Primate Research Center, Drug Safety Evaluation Center, Institute of Medical Biology, Chinese Academy of Medical Sciences/Peking Union Medical College, Kunming, 650118, China
| | - Qinfang Jiang
- Medical Primate Research Center, Drug Safety Evaluation Center, Institute of Medical Biology, Chinese Academy of Medical Sciences/Peking Union Medical College, Kunming, 650118, China
| | - Wanjing Yang
- Medical Primate Research Center, Drug Safety Evaluation Center, Institute of Medical Biology, Chinese Academy of Medical Sciences/Peking Union Medical College, Kunming, 650118, China
| | - Rujia Yang
- Medical Primate Research Center, Drug Safety Evaluation Center, Institute of Medical Biology, Chinese Academy of Medical Sciences/Peking Union Medical College, Kunming, 650118, China
| | - Donghong Tang
- Medical Primate Research Center, Drug Safety Evaluation Center, Institute of Medical Biology, Chinese Academy of Medical Sciences/Peking Union Medical College, Kunming, 650118, China.
| |
Collapse
|
36
|
Zhang Z, Zhang J, Yan X, Wang J, Huang H, Teng M, Liu Q, Han S. Dissecting the genetic basis and mechanisms underlying the associations between multiple extrahepatic factors and autoimmune liver diseases. J Transl Autoimmun 2025; 10:100260. [PMID: 39741931 PMCID: PMC11683281 DOI: 10.1016/j.jtauto.2024.100260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 11/02/2024] [Accepted: 11/03/2024] [Indexed: 01/03/2025] Open
Abstract
Background Autoimmune liver diseases (AILDs) encompass autoimmune hepatitis (AIH), primary biliary cholangitis (PBC), and primary sclerosing cholangitis (PSC). The onset of these diseases is fundamentally influenced by genetic susceptibility. Although various extrahepatic factors are potentially linked to AILDs, the genetic underpinnings and mechanisms of these associations remain unclear. Methods Utilizing large-scale genome-wide association study (GWAS) data, this study systematically investigated the relationships between extrahepatic autoimmune diseases (EHAIDs), immune cells, and various triggering factors with AILDs. Mendelian randomization (MR) was employed to assess the causal effects of these extrahepatic factors on AILDs, complemented by linkage disequilibrium score (LDSC) regression to uncover shared genetic architecture and causal effects underlying the associations between autoimmune diseases. We employed colocalization, enrichment analysis, and protein-protein interaction (PPI) network to identify the functions of shared loci. Additionally, we proposed that activated immune cells in the circulation may contribute to liver and biliary tract inflammation via migration, mediating the impact of extrahepatic factors on AILDs. This hypothesis was tested using two mediation analysis methods: two-step MR (TSMR) and multivariable MR (MVMR). Results Causal associations between multiple extrahepatic factors and AILDs were identified. Notably, CD27+ B cells were found to be a risk factor for PBC, while PSC progression was associated with CD28+ CD8+ T cells exhaustion and increased levels of CD28- CD8+ T cells. Mediation analyses revealed 64 pathways via TSMR and 15 pathways via MVMR, indicating that the effects of extrahepatic factors on AILDs may be mediated by circulating immune cells. The shared genetic architecture also contributed to these associations. Analysis of shared loci and gene functions identified ATXN2 as being shared between PBC and 9 EHAIDs, while SH2B3 and PSMG1 were shared with 6 and 5 EHAIDs, respectively, in PSC. Conclusions Our research compared three distinct AILDs, enhancing the understanding of their etiology and providing new evidence on risk factors, diagnostic markers, and potential therapeutic targets.
Collapse
Affiliation(s)
- Zheng Zhang
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi Province, China
| | - Jiayi Zhang
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi Province, China
| | - Xinyang Yan
- Department of Neurosurgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi Province, China
| | - Jiachen Wang
- Department of Joint Surgery, HongHui Hospital, Xi'an Jiaotong University, Xi'an, 710061, Shaanxi Province, China
| | - Haoxiang Huang
- Department of urology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi Province, China
| | - Menghao Teng
- Department of Orthopedics, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi Province, China
| | - Qingguang Liu
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi Province, China
| | - Shaoshan Han
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi Province, China
| |
Collapse
|
37
|
Formichi C, Caprio S, Nigi L, Dotta F. The impact of environmental pollution on metabolic health and the risk of non-communicable chronic metabolic diseases in humans. Nutr Metab Cardiovasc Dis 2025; 35:103975. [PMID: 40180824 DOI: 10.1016/j.numecd.2025.103975] [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: 01/20/2025] [Revised: 02/25/2025] [Accepted: 02/28/2025] [Indexed: 04/05/2025]
Abstract
AIMS This review aims to provide a comprehensive overview to understand the role of pollution in the development of noncommunicable diseases (NCDs), with a focus on metabolic diseases. DATA SYNTHESIS In the context of NCDs, the incidence of metabolic diseases such as obesity and diabetes are increasing at an alarming rate. In addition to the well-known role of the so-called "obesogenic" environment, characterized by unhealthy diet and physical inactivity, great attention has been paid in recent years to the effects of pollution. Indeed, progressive urbanization has been associated with increased exposure to pollutants. The harmful effects of some pollutants on the endocrine system have been known for decades, but data on the metabolic impact of pollution are rather recent. Pollution in its various forms promotes a systemic inflammatory state, insulin resistance, and oxidative stress, which appear to be closely associated with increased risk of NCD, particularly obesity and diabetes. CONCLUSIONS In conclusion, urbanization has so far had a predominantly negative impact on collective health, but a better understanding of the mechanisms linking pollution to metabolic health is crucial to implement preventive strategies, including careful urban planning to improve community health, understood not only as the absence of disease but also as psychological and social well-being, overcoming the risks associated with urbanization.
Collapse
Affiliation(s)
- Caterina Formichi
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Viale Bracci 1-16, 53100, Siena, Italy.
| | - Sonia Caprio
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Viale Bracci 1-16, 53100, Siena, Italy
| | - Laura Nigi
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Viale Bracci 1-16, 53100, Siena, Italy
| | - Francesco Dotta
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Viale Bracci 1-16, 53100, Siena, Italy
| |
Collapse
|
38
|
Skawratananond S, Xiong DX, Zhang C, Tonk S, Pinili A, Delacruz B, Pham P, Smith SC, Navab R, Reddy PH. Mitophagy in Alzheimer's disease and other metabolic disorders: A focus on mitochondrial-targeted therapeutics. Ageing Res Rev 2025; 108:102732. [PMID: 40122398 DOI: 10.1016/j.arr.2025.102732] [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: 11/15/2024] [Revised: 02/19/2025] [Accepted: 03/15/2025] [Indexed: 03/25/2025]
Abstract
Mitochondria, as central regulators of cellular processes such as energy production, apoptosis, and metabolic homeostasis, are essential to cellular function and health. The maintenance of mitochondrial integrity, especially through mitophagy-the selective removal of impaired mitochondria-is crucial for cellular homeostasis. Dysregulation of mitochondrial function, dynamics, and biogenesis is linked to neurodegenerative and metabolic diseases, notably Alzheimer's disease (AD), which is increasingly recognized as a metabolic disorder due to its shared pathophysiologic features: insulin resistance, oxidative stress, and chronic inflammation. In this review, we highlight recent advancements in pharmacological interventions, focusing on agents that modulate mitophagy, mitochondrial uncouplers that reduce oxidative phosphorylation, compounds that directly scavenge reactive oxygen species to alleviate oxidative stress, and molecules that ameliorate amyloid beta plaque accumulation and phosphorylated tau pathology. Additionally, we explore dietary and lifestyle interventions-MIND and ketogenic diets, caloric restriction, physical activity, hormone modulation, and stress management-that complement pharmacological approaches and support mitochondrial health. Our review underscores mitochondria's central role in the pathogenesis and potential treatment of neurodegenerative and metabolic diseases, particularly AD. By advocating for an integrated therapeutic model that combines pharmacological and lifestyle interventions, we propose a comprehensive approach aimed at mitigating mitochondrial dysfunction and improving clinical outcomes in these complex, interrelated diseases.
Collapse
Affiliation(s)
- Shadt Skawratananond
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States; Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States.
| | - Daniel X Xiong
- Department of Integrative Biology, The University of Texas at Austin, Austin, TX 78712, United States.
| | - Charlie Zhang
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States; Honors College, Texas Tech University, Lubbock, TX 79401, United States; Department of Biology, Texas Tech University, Lubbock, TX 79401, USA, Texas Tech University, Lubbock, TX 79401, United States.
| | - Sahil Tonk
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States.
| | - Aljon Pinili
- Honors College, Texas Tech University, Lubbock, TX 79401, United States; Department of Biology, Texas Tech University, Lubbock, TX 79401, USA, Texas Tech University, Lubbock, TX 79401, United States.
| | - Brad Delacruz
- Honors College, Texas Tech University, Lubbock, TX 79401, United States; Department of Biology, Texas Tech University, Lubbock, TX 79401, USA, Texas Tech University, Lubbock, TX 79401, United States.
| | - Patrick Pham
- Honors College, Texas Tech University, Lubbock, TX 79401, United States; Department of Biology, Texas Tech University, Lubbock, TX 79401, USA, Texas Tech University, Lubbock, TX 79401, United States.
| | - Shane C Smith
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States.
| | - Rahul Navab
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States; Department of Internal Medicine, PES Institute of Medical Sciences and Research, Kuppam, India.
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States; Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States; Nutritional Sciences Department, College Human Sciences, Texas Tech University, Lubbock, TX 79409, United States; Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States; Department of Public Health, Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States; Department of Speech, Language, and Hearing Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| |
Collapse
|
39
|
Lei F, Zheng M, Zhang T, Wang S, Li B, He D, Zhang S, Zhou L, Zhang Q, Hu Z. Physicochemical analysis of beef tallow and its liquid fraction, comparing frying performance with high oleic acid rapeseed oil and rice bran oil. Food Chem 2025; 476:143515. [PMID: 40015054 DOI: 10.1016/j.foodchem.2025.143515] [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: 12/24/2024] [Revised: 02/07/2025] [Accepted: 02/18/2025] [Indexed: 03/01/2025]
Abstract
Fractionation allows the separation of components in beef tallow. This study compared the physicochemical characteristics and cholesterol content of beef tallow and its liquid fraction, evaluating their frying performance as potential deep-fat frying oils against plant oils. Results showed effective separation of unsaturated components from beef tallow through fractionation. Beef tallow exhibited superior physicochemical properties during frying, with lower deterioration levels than plant oils. Benzo[a]pyrene content increased in plant oils but remained low in beef tallow and its liquid fraction. The liquid fraction had a significantly shorter oxidative induction time of 0.38 h compared to 5.85 h and 5.24 h for plant oils. This study revealed that alterations were observed in beef tallow and its liquid fraction when used as frying oils, with beef tallow demonstrating stronger antioxidative properties compared to the liquid fraction, which exhibited lower levels of cholesterol and saturated fatty acids.
Collapse
Affiliation(s)
- Fenfen Lei
- Key Laboratory of Edible Oil Quality and Safety, State Administration for Market Regulation, School of Food Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, China; Grain and Oil Resources Comprehensive Exploitation and Engineering Technology Research Center of State Administration of Grain, College of Food Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, China; Wuhan Institute for Food and Cosmetic Control, Wuhan 430012, China
| | - Meiyu Zheng
- Key Laboratory of Edible Oil Quality and Safety, State Administration for Market Regulation, School of Food Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, China; Grain and Oil Resources Comprehensive Exploitation and Engineering Technology Research Center of State Administration of Grain, College of Food Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Tianyu Zhang
- Key Laboratory of Edible Oil Quality and Safety, State Administration for Market Regulation, School of Food Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, China; Grain and Oil Resources Comprehensive Exploitation and Engineering Technology Research Center of State Administration of Grain, College of Food Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Shu Wang
- Wuhan Institute for Food and Cosmetic Control, Wuhan 430012, China
| | - Bin Li
- School of Mechanical Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Dongping He
- Key Laboratory of Edible Oil Quality and Safety, State Administration for Market Regulation, School of Food Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, China; Grain and Oil Resources Comprehensive Exploitation and Engineering Technology Research Center of State Administration of Grain, College of Food Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, China; Wuhan Institute for Food and Cosmetic Control, Wuhan 430012, China
| | - Sihong Zhang
- Key Laboratory of Edible Oil Quality and Safety, State Administration for Market Regulation, School of Food Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, China; Grain and Oil Resources Comprehensive Exploitation and Engineering Technology Research Center of State Administration of Grain, College of Food Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Li Zhou
- Key Laboratory of Edible Oil Quality and Safety, State Administration for Market Regulation, School of Food Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, China; Grain and Oil Resources Comprehensive Exploitation and Engineering Technology Research Center of State Administration of Grain, College of Food Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, China.
| | - Qinfeng Zhang
- Key Laboratory of Edible Oil Quality and Safety, State Administration for Market Regulation, School of Food Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, China; Hubei Key Laboratory of Resources and Eco-Environment Geology, Hubei Geological Research Laboratory, Hubei, Geological Bureau, Wuhan 430034, China.
| | - Zhigang Hu
- School of Mechanical Engineering, Wuhan Polytechnic University, Wuhan 430023, China.
| |
Collapse
|
40
|
Hu Y, Sang N, Wu A, Pu J, Yan H, Luo J, Zheng P, Luo Y, Yu J, He J, Yu B, Chen D. Different types of bile acids exhibit opposite regulatory effects on lipid metabolism in finishing pigs through bile acid receptors. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2025; 21:25-36. [PMID: 40135169 PMCID: PMC11930731 DOI: 10.1016/j.aninu.2024.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 11/20/2024] [Accepted: 12/16/2024] [Indexed: 03/27/2025]
Abstract
The purpose of this research was to investigate how different bile acids impact lipid metabolism and carcass characteristics in finishing pigs, along with the potential mechanisms involved. Twenty-one finishing pigs (Duroc×Landrace×Yorkshire [DLY]; average BW = 144.38 ± 8.92 kg) were assigned to three dietary treatments, with each treatment containing seven replicates, each consisting of one pig. The three dietary treatments included: a basic diet, a basic diet supplemented with 500 mg/kg of hyodeoxycholic acid (HDCA), and a basic diet supplemented with 500 mg/kg of lithocholic acid (LCA). The trial lasted for 28 d. Hyodeoxycholic acid was used in the in vitro experiments and added to mature 3T3-L1 adipocytes for 4 d to elucidate the mechanism by which bile acids regulate lipid metabolism. The results suggested that HDCA tended to decrease backfat thickness in finishing pigs (P = 0.094) and reduced the size of lipid droplets in 3T3-L1 adipocytes (P = 0.012), whereas LCA increased backfat thickness (P = 0.016) and induced larger lipid droplets in the abdominal adipose tissue (P = 0.003). Furthermore, HDCA enhanced the expression of Takeda G-protein-coupled receptor 5 protein and hormone-sensitive lipase (HSL) gene in backfat of pigs (P < 0.05) and increased the protein expression of phosphorylated HSL (p-HSL) in vitro (P = 0.093). Compared to HDCA, LCA addition increased the gene and protein expression of peroxisome proliferator activated receptor gamma in backfat of pigs (P < 0.05) and enhanced the expression of hepatic genes sterol regulatory element-binding protein-1c and fatty acid synthase (P < 0.05). In conclusion, HDCA enhanced lipolysis and partially decreased backfat thickness in finishing pigs, while LCA promoted lipid synthesis and increased backfat thickness of pigs. The variations in the effects of various bile acids on bile acid receptors could explain these functional differences.
Collapse
Affiliation(s)
- Yaolian Hu
- Key Laboratory of Animal Disease-resistant Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Ya'an 625014, China
| | - Ni Sang
- Key Laboratory of Animal Disease-resistant Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Ya'an 625014, China
| | - Aimin Wu
- Key Laboratory of Animal Disease-resistant Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Ya'an 625014, China
| | - Junning Pu
- Key Laboratory of Animal Disease-resistant Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Ya'an 625014, China
| | - Hui Yan
- Key Laboratory of Animal Disease-resistant Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Ya'an 625014, China
| | - Junqiu Luo
- Key Laboratory of Animal Disease-resistant Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Ya'an 625014, China
| | - Ping Zheng
- Key Laboratory of Animal Disease-resistant Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Ya'an 625014, China
| | - Yuheng Luo
- Key Laboratory of Animal Disease-resistant Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Ya'an 625014, China
| | - Jie Yu
- Key Laboratory of Animal Disease-resistant Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Ya'an 625014, China
| | - Jun He
- Key Laboratory of Animal Disease-resistant Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Ya'an 625014, China
| | - Bing Yu
- Key Laboratory of Animal Disease-resistant Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Ya'an 625014, China
| | - Daiwen Chen
- Key Laboratory of Animal Disease-resistant Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Ya'an 625014, China
| |
Collapse
|
41
|
Pérez-Tierra G, Calo J, Comesaña S, Velasco C, Blanco AM, Soengas JL. Evidence for the involvement of secondary bile acids on peripheral and central regulation of feed intake in rainbow trout. Am J Physiol Endocrinol Metab 2025; 328:E787-E803. [PMID: 40257242 DOI: 10.1152/ajpendo.00072.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 03/16/2025] [Accepted: 04/01/2025] [Indexed: 04/22/2025]
Abstract
Recent studies suggest that secondary bile acids (SBAs) play a role in energy metabolism and feed intake regulation, but their effects in fish remain largely unknown. This study evaluates the impact of intragastric administration of the main SBAs [500 µM lithocholic acid (LCA), 1,500 µM deoxycholic acid (DCA), and their taurine conjugates: 1,000 µM T-LCA and 600 µM T-DCA] on feed intake, regulatory pathways, and bile acid-related elements in rainbow trout. Results show that all tested SBAs influenced bile acid transporters [apical sodium-dependent bile acid transporter (Asbt), Na+ taurocholate co-transporting polypeptide (Ntcp), organic solute transporter α and β (Ostα, and Ostβ)] and receptors [farnesoid X receptor like-α and β (Fxrα, Fxrβ), and Takeda G protein-coupled receptor 5 (Tgr5)], with DCA and T-DCA mainly affecting the gastrointestinal tract and LCA modulating hypothalamic pathways, suggesting a putative orexigenic role. Plasma analysis confirmed SBA absorption from the gastrointestinal tract into the bloodstream. This study provides the first evidence in fish of SBAs modulating gene and protein expression linked to appetite regulation, underscoring their role in gut-brain communication. Although all SBAs influenced fxr expression, gpbar1 remained unaffected, differing from mammals where BAs suppress appetite. Notably, despite taurine-conjugated SBAs being the most abundant in rainbow trout, only nonconjugated LCA showed significant effects. Taken together, these results provide new information on the emerging importance of SBA in feed intake regulation and bile acid mechanisms.NEW & NOTEWORTHY This study determined for the first time in teleost fish (specifically in rainbow trout): 1) the role of secondary bile acids in the regulation of feed intake and associated signaling pathways, highlighting a putative orexigenic role of lithocholic acid (LCA); 2) the response of Asbt and Ostα transporters and Tgr5 receptor in hypothalamus after LCA administration; and 3) the reabsorption of LCA, DCA, and their taurine conjugates from the gastrointestinal tract into the bloodstream.
Collapse
Affiliation(s)
- Gabriel Pérez-Tierra
- Centro de Investigación Mariña, Laboratorio de Fisioloxía Animal, Departamento de Bioloxía Funcional e Ciencias da Saúde, Facultade de Bioloxía, Universidade de Vigo, Vigo, Spain
| | - Jessica Calo
- Centro de Investigación Mariña, Laboratorio de Fisioloxía Animal, Departamento de Bioloxía Funcional e Ciencias da Saúde, Facultade de Bioloxía, Universidade de Vigo, Vigo, Spain
| | - Sara Comesaña
- Centro de Investigación Mariña, Laboratorio de Fisioloxía Animal, Departamento de Bioloxía Funcional e Ciencias da Saúde, Facultade de Bioloxía, Universidade de Vigo, Vigo, Spain
| | - Cristina Velasco
- Centro de Investigación Mariña, Laboratorio de Fisioloxía Animal, Departamento de Bioloxía Funcional e Ciencias da Saúde, Facultade de Bioloxía, Universidade de Vigo, Vigo, Spain
| | - Ayelén M Blanco
- Centro de Investigación Mariña, Laboratorio de Fisioloxía Animal, Departamento de Bioloxía Funcional e Ciencias da Saúde, Facultade de Bioloxía, Universidade de Vigo, Vigo, Spain
| | - José L Soengas
- Centro de Investigación Mariña, Laboratorio de Fisioloxía Animal, Departamento de Bioloxía Funcional e Ciencias da Saúde, Facultade de Bioloxía, Universidade de Vigo, Vigo, Spain
| |
Collapse
|
42
|
Cuffe MS, Staudacher HM, Aziz I, Adame EC, Krieger-Grubel C, Madrid AM, Ohlsson B, Black CJ, Ford AC. Efficacy of dietary interventions in irritable bowel syndrome: a systematic review and network meta-analysis. Lancet Gastroenterol Hepatol 2025; 10:520-536. [PMID: 40258374 DOI: 10.1016/s2468-1253(25)00054-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 04/23/2025]
Abstract
BACKGROUND Patients with irritable bowel syndrome (IBS) are often interested in dietary interventions as a means of managing their symptoms. However, the relative efficacy of available diets for the management of IBS is unclear. We aimed to examine the relative efficacy of various dietary interventions in IBS. METHODS For this systematic review and network meta-analysis we searched MEDLINE, EMBASE, EMBASE Classic, and the Cochrane Central Register of Controlled Trials from database inception to Feb 7, 2025, to identify randomised controlled trials comparing an active dietary intervention requiring changes to the intake of more than one food in IBS with either a control intervention, such as a habitual diet, sham diet, a high fermentable oligosaccharides, disaccharides, monosaccharides, and polyols (FODMAP) diet, or alternative miscellaneous dietary advice, or any other active dietary intervention requiring changes to the intake of more than one food. We assessed efficacy using dichotomous assessments of improvement in global IBS symptoms or improvement in individual IBS symptoms, including abdominal pain, abdominal bloating or distension, and bowel habit. We pooled data using a random-effects model, with the efficacy of each intervention reported as pooled relative risks (RRs) with 95% CIs. We ranked interventions according to their P-score, which measures the mean extent of certainty that one intervention is better than another, averaged over all competing interventions. FINDINGS We identified 28 eligible randomised controlled trials (comprising 2338 patients) of 11 different dietary interventions compared with four control interventions, of which six (low FODMAP diet, British Dietetic Association/National Institute for Health and Care Excellence [BDA/NICE] diet, lactose-reduced diet, starch-reduced and sucrose-reduced diet, a personalised diet, and a Mediterranean diet) were studied in more than one trial. For global IBS symptoms, assessed in 28 randomised controlled trials and when considering only the dietary interventions studied in more than one trial, a starch-reduced and sucrose-reduced diet ranked first (RR of global IBS symptoms not improving 0·41 [95% CI 0·26-0·67]; P-score 0·84; two trials), a low FODMAP diet ranked fourth (0·51 [0·37-0·70]; P-score 0·71; 24 trials), and a BDA/NICE diet ranked tenth (0·62 [0·43-0·90]; P-score 0·44; eight trials), versus a habitual diet. For abdominal pain, assessed in 26 trials and when considering only the dietary interventions studied in more than one randomised controlled trial, a starch-reduced and sucrose-reduced diet ranked second (RR of abdominal pain not improving 0·54 [95% CI 0·33-0·90]; P-score 0·73; two trials), and a low FODMAP diet ranked fifth (0·61 [0·42-0·89]; P-score 0·64; 23 trials), versus a habitual diet. For abdominal bloating or distension, assessed in 26 trials and when considering only the dietary interventions studied in more than one randomised trial, only a low FODMAP diet (RR of abdominal bloating or distension not improving 0·55 [95% CI 0·37-0·80]; P-score 0·64; 23 trials) was superior to a habitual diet and ranked fourth. For bowel habit, assessed in 23 randomised trials, none of the dietary interventions was superior to any of the control interventions, but a low FODMAP diet was superior to a BDA/NICE diet (RR of bowel habit not improving 0·79 [95% CI 0·63-0·99]). All comparisons across the network were rated as low or very low confidence, except for direct comparisons between a low FODMAP diet or a starch-reduced and sucrose-reduced diet and habitual diet, both of which were rated as moderate confidence. INTERPRETATION In terms of dietary interventions for IBS, the most evidence exists for a low FODMAP diet, but other promising therapies are emerging and should be the subject of further study. FUNDING None.
Collapse
Affiliation(s)
- Melanie S Cuffe
- Leeds Gastroenterology Institute, St James's University Hospital, Leeds, UK; Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - Heidi M Staudacher
- Monash University, Department of Medicine, School of Translational Medicine, Melbourne, VIC, Australia
| | - Imran Aziz
- Academic Unit of Gastroenterology, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK; Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield, UK
| | - Enrique Coss Adame
- Department of Gastroenterology/GI Motility Lab, Instituto Nacional de Ciencias Médicas y Nutrición "Salvador Zubirán", México City, México
| | - Claudia Krieger-Grubel
- Department of Gastroenterology/Hepatology, Cantonal Hospital St Gallen, St Gallen, Switzerland
| | - Ana Maria Madrid
- Section of Gastroenterology, Department of Medicine, Hospital Clinico Universidad de Chile, Santiago, Chile
| | - Bodil Ohlsson
- Department of Internal Medicine, Skåne University Hospital, Malmo, Sweden; Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Christopher J Black
- Leeds Gastroenterology Institute, St James's University Hospital, Leeds, UK; Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - Alexander C Ford
- Leeds Gastroenterology Institute, St James's University Hospital, Leeds, UK; Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK.
| |
Collapse
|
43
|
Zhu Y, Tian Q, Huang Q, Wang J. Bile-processed Rhizoma Coptidis alleviates type 2 diabetes mellitus through modulating the gut microbiota and short-chain fatty acid metabolism. Int Immunopharmacol 2025; 156:114645. [PMID: 40253770 DOI: 10.1016/j.intimp.2025.114645] [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: 11/10/2024] [Revised: 03/27/2025] [Accepted: 04/07/2025] [Indexed: 04/22/2025]
Abstract
BACKGROUND Bile-Processed Rhizoma Coptidis (BPRC) is a processed products of Rhizoma Coptidis (RC) commonly used to treat type 2 diabetes mellitus (T2DM). However, the synergistic mechanism of its processing remains unknown. Current research indicates that the gut microbiota and its metabolites, such as short-chain fatty acids (SCFAs), are closely associated with the progression of T2DM. PURPOSE This study aims to investigate the effects of BPRC on the gut microbiota and its metabolite SCFAs in T2DM rats. METHODS T2DM rat model was induced by a high-fat diet (HFD) combined with streptozotocin (STZ), followed by a 4-week treatment with BPRC to observe its therapeutic effects. The impact of BPRC on the gut microbiota was studied through metagenomic sequencing. Quantitative analysis of SCFAs was conducted using GC-MS. Western blot and quantitative real-time PCR (qRT-PCR) were conducted to investigate the potential mechanisms of BPRC. RESULTS BPRC significantly improved insulin resistance in T2DM rats, downregulated levels of pancreatic cell apoptosis factors, and upregulated the abundance of Bacteroides uniformis, Bacteroides sp A1C1, Anaerostipes caccae, Alistipes finegoldii and Blautia sp.N6H1-15 in T2DM rats. Additionally, BPRC increased the levels of seven SCFAs in the intestines of T2DM rats. It activated intestinal TGR5, GPR41, GPR43, and GPR109a receptors, collectively upregulating GLP-1 protein expression, and exerted therapeutic effects on T2DM. CONCLUSION The results indicate that the synergistic mechanism of BPRC in treating T2DM is associated with modulating the gut microbiota, increasing SCFAs content in the intestines, and regulating intestinal GLP-1 production.
Collapse
MESH Headings
- Animals
- Gastrointestinal Microbiome/drug effects
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/microbiology
- Male
- Drugs, Chinese Herbal/therapeutic use
- Drugs, Chinese Herbal/pharmacology
- Rats
- Fatty Acids, Volatile/metabolism
- Rats, Sprague-Dawley
- Diabetes Mellitus, Experimental/drug therapy
- Diabetes Mellitus, Experimental/metabolism
- Hypoglycemic Agents/pharmacology
- Hypoglycemic Agents/therapeutic use
- Bile/metabolism
- Diet, High-Fat
- Glucagon-Like Peptide 1/metabolism
- Insulin Resistance
- Blood Glucose/metabolism
- Coptis chinensis
Collapse
Affiliation(s)
- Ying Zhu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China; College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qingqing Tian
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China; College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qinwan Huang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China; College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Jin Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China; College of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
44
|
de Oliveira MR. Pre-clinical evidence for mitochondria as a therapeutic target for luteolin: A mechanistic view. Chem Biol Interact 2025; 413:111492. [PMID: 40154935 DOI: 10.1016/j.cbi.2025.111492] [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: 12/20/2024] [Revised: 03/05/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025]
Abstract
Pre-clinical evidence indicates that mitochondria may be a therapeutic target for luteolin (3',4',5,7-tetrahydroxyflavone; LUT) in different conditions. LUT modulates mitochondrial physiology in in vitro, ex vivo, and in vivo experimental models. This flavone exerted mitochondria-related antioxidant and anti-apoptotic effects, stimulated mitochondrial fusion and fission, induced mitophagy, and promoted mitochondrial biogenesis in human and animal cells and tissues. Moreover, LUT modulated the activity of components of the oxidative phosphorylation (OXPHOS) system, improving the ability of mitochondria to produce adenosine triphosphate (ATP) in certain circumstances. The mechanism of action by which LUT promoted mitochondrial benefits and protection are not completely clear yet. Nonetheless, LUT is a potential candidate to be utilized in mitochondrial therapy in the future. In this work, it is explored the mechanisms of action by which LUT modulates mitochondrial physiology in different pre-clinical experimental models.
Collapse
Affiliation(s)
- Marcos Roberto de Oliveira
- Grupo de Estudos em Neuroquímica e Neurobiologia de Moléculas Bioativas, Departamento de Química, Universidade Federal de Mato Grosso (UFMT), CEP 78060-900, Cuiaba, Mato Grosso, Brazil; Grupo de Estudos em Terapia Mitocondrial, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul (UFRGS), CEP 90035-003, Porto Alegre, Rio Grande do Sul, Brazil; Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Universidade Federal do Rio Grande do Sul (UFRGS), CEP 90035-003, Porto Alegre, Rio Grande do Sul, Brazil.
| |
Collapse
|
45
|
Liu Y, Pei Y, Wang H, Yang Z. Lead promoted bile acid deconjugation by modulating gut bacteria encoding bile salt hydrolase (BSH) in Rana chensinensis tadpoles. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 373:126187. [PMID: 40185186 DOI: 10.1016/j.envpol.2025.126187] [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: 01/26/2025] [Revised: 03/13/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
Bile salt hydrolase (BSH) is produced by gut bacteria and is responsible for deconjugating amino acids from the aliphatic side chains of conjugated bile acids (BAs), initiating the critical first step in BAs metabolism. Lead (Pb) is known to cause gut microbial dysbiosis, but whether it affects BAs profiles by reshaping the gut microbiota remains elusive. Here, using targeted BAs metabolomics and metagenomics sequencing, we found that 200 μg/L Pb treatment led to a significant increase in the abundance of BSH-producing microbiota (e.g., Eubacterium and Yersinia), thus promoting the deconjugation of taurocholic acid (TCA) and taurochenodeoxycholic acid (TCDCA). Consequently, the accumulation of relatively hydrophobic BAs cholic acid (CA) and chenodeoxycholic acid (CDCA) may cause damage to enterocytes (e.g., reduced microvilli and enterocyte heights), which attenuated tadpole digestion and ultimately led to significant reductions in morphological parameters. The inhibition of tadpole growth by Pb toxicity may negatively affect their survival and even increase their risk of death. Overall, these results revealed for the first time the toxicological mechanism by which Pb reshapes the gut microbiota and thus disrupts the BAs profile, shedding new insights into the detrimental effects of Pb toxicity on amphibian growth.
Collapse
Affiliation(s)
- Ying Liu
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, 710119, China
| | - Yuebin Pei
- Cotton Research Institute, Shanxi Agriculture University, Yuncheng, Shanxi, 044000, China
| | - Hongyuan Wang
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, 710119, China.
| | - Zhangmin Yang
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, 710119, China
| |
Collapse
|
46
|
Wylie AC, Murgueitio N, Carlson AL, Fry RC, Propper CB. The role of the gut microbiome in the associations between lead exposure and child neurodevelopment. Toxicol Lett 2025; 408:95-104. [PMID: 40250742 DOI: 10.1016/j.toxlet.2025.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 03/10/2025] [Accepted: 04/14/2025] [Indexed: 04/20/2025]
Abstract
Lead is highly toxic to the developing brain. Given its persistence in the environment, new intervention strategies are needed to mitigate the impacts of lead on child neurodevelopment. The gut microbiome, referring to the bacteria and microorganisms residing in the gastrointestinal system, may be a viable target for intervention. This short review summarizes recent evidence linking the gut-brain axis to child developmental outcomes. We explore how lead-induced effects to the gut microbiome could indirectly affect child neurodevelopment, such that disrupting or offsetting this mediating process could buffer the effects of lead on child developmental outcomes. Unexpected findings with respect to child microbiota diversity and child cognitive and behavioral outcomes as well as lead exposure and adult microbiota diversity are discussed. When possible, we draw connections between observed changes to relative bacterial abundance, proposed bacterial functions, and downstream effects to brain development. We also explore how the gut microbiome might modify the toxicity of lead by impeding the uptake of lead across the gastrointestinal tract or through indirect mechanisms in such ways that the gut microbiome does not fit within a mediating pathway. In this case, promoting the buffering capacity of the gut microbiome may reduce the impacts of lead on child neurodevelopment. The goal of this short review is to bring attention to the potential role of the gut microbiome in the associations between lead exposure and child neurodevelopment with an eye towards intervention.
Collapse
Affiliation(s)
- Amanda C Wylie
- RTI International, Research Triangle Park, North Carolina, United States; Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, United States.
| | - Nicolas Murgueitio
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, United States
| | | | - Rebecca C Fry
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, United States; Institute for Environmental Health Solutions, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, United States
| | - Cathi B Propper
- School of Nursing, University of North Carolina at Chapel Hill, United States; Frank Porter Graham Child Development Institute, University of North Carolina at Chapel Hill, United States
| |
Collapse
|
47
|
Yuan L, Wang S, Wang D, Wang E. Association of cardiometabolic index with gallstone disease and insulin resistance based on NHANES data. BMC Gastroenterol 2025; 25:354. [PMID: 40346457 DOI: 10.1186/s12876-025-03950-8] [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: 01/15/2025] [Accepted: 04/28/2025] [Indexed: 05/11/2025] Open
Abstract
BACKGROUND Cardiometabolic index (CMI) is an index integrating visceral obesity and dyslipidemia. This study intends to scrutinize the connection between CMI and gallstone disease (GSD) and to elucidate the association between CMI and insulin resistance (IR) in patients with GSD. METHODS To explore the potential nonlinear association and determine the inflection point, a restricted cubic spline (RCS) analysis was performed. Following categorization of CMI based on the identified inflection point, multivariate logistic regression models, subgroup analyses, and interaction tests were utilized to assess the connection between CMI and GSD, as well as between CMI and IR in GSD patients. The homeostasis model assessment for IR (HOMA-IR) and triglyceride-glucose (TyG) index was applied to evaluate IR. Spearman analysis was implemented to investigate the connection between CMI and HOMA-IR. The predictive performance of each indicator was evaluated by the receiver operating characteristic (ROC) curve and the area under the curve (AUC). RESULTS The study included 2311 individuals, with a GSD prevalence of 10.90%. RCS analysis revealed a nonlinear positive correlation between CMI and GSD (nonlinear P < 0.001), as well as between CMI and IR (nonlinear P < 0.001). In the fully adjusted multivariable logistic regression analysis of covariates, compared with the low-category CMI group, the high-category CMI was significantly associated with the risk of GSD (OR = 1.547, 95% CI: 1.143-2.092, P = 0.005), IR (OR = 4.990, 95% CI: 2.517-9.892, P < 0.001). Subgroup analysis demonstrated that the correlation between CMI and GSD was stronger in females. Spearman correlation analysis showed a positive association between CMI and HOMA-IR in GSD patients (r = 0.548, P < 0.001). The ROC curve demonstrated the predictive performance of the CMI model for GSD (AUC = 0.743), which was superior to conventional indicators such as Body Mass Index and Waist Circumference; the predictive performance of CMI (AUC = 0.772) for IR was consistent with that of TyG (AUC = 0.772). CONCLUSION Our research demonstrates that CMI exhibits a nonlinear positive correlation with the incidence of GSD and IR. This suggests that CMI may serve as a novel and valuable indicator for further investigating the intricate relationships among metabolic syndrome, obesity, and GSD.
Collapse
Affiliation(s)
- Liu Yuan
- China Medical University, Shenyang, China
- Central Hospital of Dalian University of Technology (Dalian Municipal Central Hospital), Dalian, China
| | - Shuqi Wang
- Central Hospital of Dalian University of Technology (Dalian Municipal Central Hospital), Dalian, China
| | - Dong Wang
- Central Hospital of Dalian University of Technology (Dalian Municipal Central Hospital), Dalian, China
| | - Enbo Wang
- Central Hospital of Dalian University of Technology (Dalian Municipal Central Hospital), Dalian, China.
| |
Collapse
|
48
|
Richtsmeier P, Nedielkov R, Haring M, Yücel O, Elsner L, Lülf RH, Wöhlbrand L, Rabus R, Moeller H, Philipp B, Mueller FM. 7β-Hydroxysteroid dehydratase Hsh3 eliminates the 7-hydroxy group of the bile salt ursodeoxycholate during degradation by Sphingobium sp. strain Chol11 and other Sphingomonadaceae. Appl Environ Microbiol 2025:e0018525. [PMID: 40340444 DOI: 10.1128/aem.00185-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 04/11/2025] [Indexed: 05/10/2025] Open
Abstract
Bile salts are steroids with distinctive hydroxylation patterns that are produced and excreted by vertebrates. In contrast to common human bile salts, ursodeoxycholate (UDCA) has a 7-hydroxy group in β-configuration and is used in large amounts for the treatment of diverse gastrointestinal diseases. We isolated the 7β-hydroxysteroid dehydratase Hsh3 that is involved in UDCA degradation by Sphingobium sp. strain Chol11. Hsh3 eliminates the 7β-hydroxy group as water, leading to a double bond in the B-ring. This is similar to 7α-hydroxysteroid dehydratases in this and other strains, but Hsh3 is evolutionarily different from these. Purified Hsh3 accepted steroids with and without side chains as substrates and had minor activity with 7α-hydroxy groups. The deletion mutant strain Chol11 Δhsh3 had impacted growth with UDCA and accumulated a novel compound. The compound was identified as 3',5-dihydroxy-H-methyl-hexahydro-5-indene-1-one-propanoate, consisting of steroid rings C and D with a C3-side chain carrying the former 7β-hydroxy group, indicating that Hsh3 activity is important especially for the later stages of bile salt degradation. Hsh3 homologs were found in other sphingomonads that were also able to degrade UDCA as well as in environmental metagenomes. Thus, Hsh3 adds to the bacterial enzyme repertoire for degrading a variety of differently hydroxylated bile salts.IMPORTANCEThe bacterial degradation of different bile salts is not only important for the removal of these steroidal compounds from the environment but also harbors interesting enzymes for steroid biotechnology. The 7β-hydroxy bile salt ursodeoxycholate (UDCA) naturally occurs in high concentration in the feces of black bears and has important pharmaceutical relevance for the treatment of different liver-related diseases, for which it is administered in high and increasing amounts. Additionally, it is present in the bile salt pool of humans in trace amounts. While UDCA degradation is environmentally important, the enzyme Hsh3 modifies the hydroxy group that confers the medically relevant properties and thus might be interesting for microbiome analyses and biotechnological applications.
Collapse
Affiliation(s)
- Phil Richtsmeier
- Microbial Biotechnology and Ecology, Institute for Molecular Microbiology and Biotechnology, University of Münster, Münster, Germany
| | - Ruslan Nedielkov
- Institute for Chemistry, University of Potsdam, Potsdam, Germany
| | - Malte Haring
- Microbial Biotechnology and Ecology, Institute for Molecular Microbiology and Biotechnology, University of Münster, Münster, Germany
| | - Onur Yücel
- Microbial Biotechnology and Ecology, Institute for Molecular Microbiology and Biotechnology, University of Münster, Münster, Germany
| | - Lea Elsner
- Microbial Biotechnology and Ecology, Institute for Molecular Microbiology and Biotechnology, University of Münster, Münster, Germany
| | - Rebekka Herdis Lülf
- Microbial Biotechnology and Ecology, Institute for Molecular Microbiology and Biotechnology, University of Münster, Münster, Germany
| | - Lars Wöhlbrand
- General and Molecular Microbiology, Institute for Chemistry and Biology of the Marine Environment (ICBM), Carl von Ossietzky Universität Oldenburg, Oldenburg, Germany
| | - Ralf Rabus
- General and Molecular Microbiology, Institute for Chemistry and Biology of the Marine Environment (ICBM), Carl von Ossietzky Universität Oldenburg, Oldenburg, Germany
| | - Heiko Moeller
- Institute for Chemistry, University of Potsdam, Potsdam, Germany
| | - Bodo Philipp
- Microbial Biotechnology and Ecology, Institute for Molecular Microbiology and Biotechnology, University of Münster, Münster, Germany
- Applied Ecology and Bioresources, Fraunhofer-Institute for Molecular and Applied Ecology IME, Schmallenberg, Germany
| | - Franziska Maria Mueller
- Microbial Biotechnology and Ecology, Institute for Molecular Microbiology and Biotechnology, University of Münster, Münster, Germany
| |
Collapse
|
49
|
Crawford MS, Ulu A, Ramirez BM, Santos AN, Chatterjee P, Canale V, Manz S, Lei H, Soriano SM, Nordgren TM, McCole DF. Respiratory Exposure to Agriculture Dust Extract Alters Gut Commensal Species and Key Metabolites in Mice. J Appl Toxicol 2025. [PMID: 40344252 DOI: 10.1002/jat.4808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2025] [Revised: 04/29/2025] [Accepted: 04/29/2025] [Indexed: 05/11/2025]
Abstract
Exposure to agricultural dust containing antimicrobial-resistant pathogens poses significant health risks for workers in animal agriculture production. Beyond causing severe airway inflammation, pollutants are linked to intestinal diseases. Swine farm dust is rich in ultrafine particles, gram-positive and gram-negative bacteria, and bacterial components such as lipopolysaccharides (LPS; endotoxins). In our previous study, we demonstrated that intranasal exposure of male and female C57BL/6J mice to 12.5% hog dust extract (HDE, containing 22.1-91.1 EU/mL) for 3 weeks resulted in elevated total cell and neutrophil counts in bronchoalveolar lavage fluid and increased intestinal permeability compared to saline controls. Now, we report that 16S and metagenomic analyses of Week 3 stool samples from HDE-treated mice indicate a reduced abundance of the beneficial species Akkermansia muciniphila and Clostridium sp. ASF356 and Lachnospiraceae bacterium. Bacterial alpha diversity showed increased species evenness in fecal samples from HDE-treated mice (Pielou's evenness, p = 0.047, n = 5-6/group). Metabolomic analysis also indicated significant reductions in key metabolites involved in energy metabolism, including riboflavin (p = 0.027, n = 11) and nicotinic acid (p = 0.049, n = 11), as well as essential amino acids, such as inosine (p = 0.043, n = 11) and leucine (p = 0.018, n = 11). While HDE exposure does not robustly alter overall microbial abundance or community structure, it leads to specific reductions in beneficial bacterial species and critical metabolites necessary for maintaining intestinal homeostasis by supporting energy metabolism, gut barrier function, microbiota balance, and immune regulation. The results of this study underscore the potential risks for gut health posed by inhalation of agricultural dust.
Collapse
Affiliation(s)
- Meli'sa S Crawford
- School of Medicine, Division of Biomedical Sciences, University of California, Riverside, California, USA
- School of Environmental and Biological Sciences, Department of Animal Sciences, Rutgers-New Brunswick, New Brunswick, New Jersey, USA
| | - Arzu Ulu
- School of Medicine, Division of Biomedical Sciences, University of California, Riverside, California, USA
| | - Briana M Ramirez
- Department of Biochemistry and Molecular Biology, University of California, Riverside, California, USA
| | - Alina N Santos
- School of Medicine, Division of Biomedical Sciences, University of California, Riverside, California, USA
| | - Pritha Chatterjee
- School of Medicine, Division of Biomedical Sciences, University of California, Riverside, California, USA
| | - Vinicius Canale
- School of Medicine, Division of Biomedical Sciences, University of California, Riverside, California, USA
| | - Salomon Manz
- School of Medicine, Division of Biomedical Sciences, University of California, Riverside, California, USA
| | - Hillmin Lei
- School of Medicine, Division of Biomedical Sciences, University of California, Riverside, California, USA
| | - Sarah Mae Soriano
- School of Medicine, Division of Biomedical Sciences, University of California, Riverside, California, USA
- Bishop Gorman High School, Las Vegas, Nevada, USA
| | - Tara M Nordgren
- School of Medicine, Division of Biomedical Sciences, University of California, Riverside, California, USA
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Declan F McCole
- School of Medicine, Division of Biomedical Sciences, University of California, Riverside, California, USA
| |
Collapse
|
50
|
Wu Z, Jiang S, Li J, Wang P, Chen Y. Association between urinary cadmium levels and increased gallstone disease in US adults. Sci Rep 2025; 15:15974. [PMID: 40341372 PMCID: PMC12062283 DOI: 10.1038/s41598-025-00648-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 04/29/2025] [Indexed: 05/10/2025] Open
Abstract
Heavy metal exposure is acknowledged as a risk factor for poor health. However, the effect of heavy metal exposure on the prevalence of gallstones is still unknown. Therefore, we investigated the relationship between heavy metal concentrations and the prevalence of gallstones among US adults. Multivariate logistic regression indicated that only urinary cadmium was an independent risk factor for gallstones. Compared to the low urine cadmium group, the high cadmium group had a elevated increased risk of gallstone formation. Furthermore, the weighted quantile sum model showed that heavy metal mixtures were not associated with gallstone prevalence. Additionally, urinary cadmium levels were associated with an increased risk of gallstone formation in young individuals, males, Mexican Americans, Non-Hispanic Whites, as well as smokers and drinkers. Moreover, nine machine learning methods were utilized to construct an interpretable predictive model for gallstone prevalence. Among these models, the XGBoost model exhibited the highest performance and was selected for further investigation. Subsequently, shapely additive explanations was used for model interpretation. The results also indicated that urinary cadmium concentrations were the most important variable for gallstones. Thus, our results indicated that long-term chronic cadmium exposure is a risk factor for gallstone prevalence.
Collapse
Affiliation(s)
- Zhaowei Wu
- Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Medical College Street, Yuzhong District, Chongqing, 404100, China
| | - Shiming Jiang
- Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Medical College Street, Yuzhong District, Chongqing, 404100, China
| | - Jinzhi Li
- Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Medical College Street, Yuzhong District, Chongqing, 404100, China
| | - Panguo Wang
- Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Medical College Street, Yuzhong District, Chongqing, 404100, China
| | - Yong Chen
- Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Medical College Street, Yuzhong District, Chongqing, 404100, China.
| |
Collapse
|