1
|
Kupczyk D, Bilski R, Szeleszczuk Ł, Mądra-Gackowska K, Studzińska R. The Role of Diet in Modulating Inflammation and Oxidative Stress in Rheumatoid Arthritis, Ankylosing Spondylitis, and Psoriatic Arthritis. Nutrients 2025; 17:1603. [PMID: 40362911 PMCID: PMC12073256 DOI: 10.3390/nu17091603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2025] [Revised: 05/02/2025] [Accepted: 05/05/2025] [Indexed: 05/15/2025] Open
Abstract
Rheumatic diseases such as rheumatoid arthritis (RA), ankylosing spondylitis (AS), and psoriatic arthritis (PsA) are chronic autoimmune disorders characterized by persistent inflammation and oxidative stress, leading to joint damage and reduced quality of life. In recent years, increasing attention has been given to diet as a modifiable environmental factor that can complement pharmacological therapy. This review summarizes current evidence on how key dietary components-such as omega-3 fatty acids, fiber, polyphenols, and antioxidant vitamins-affect inflammatory pathways and oxidative balance. Special emphasis is placed on the Mediterranean diet, low-starch diets, and hypocaloric regimens, which have shown potential in improving disease activity. The gut microbiota emerges as a critical mediator between diet and immune function, with dietary interventions capable of restoring eubiosis and strengthening the intestinal barrier. Additionally, this paper discusses challenges in the clinical implementation of diet therapy, the need for personalized nutritional strategies, and the importance of integrating diet into holistic patient care. Collectively, findings suggest that dietary interventions may reduce disease activity, mitigate systemic inflammation, and enhance patients' overall well-being.
Collapse
Affiliation(s)
- Daria Kupczyk
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 24 Karłowicza St., 85-092 Bydgoszcz, Poland
| | - Rafał Bilski
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 24 Karłowicza St., 85-092 Bydgoszcz, Poland
| | - Łukasz Szeleszczuk
- Department of Organic and Physical Chemistry, Faculty of Pharmacy, Medical University of Warsaw, 1 Banacha Str., 02-093 Warsaw, Poland;
| | - Katarzyna Mądra-Gackowska
- Department of Geriatrics, Faculty of Health Sciences, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 9 Skłodowskiej Curie Str., 85-094 Bydgoszcz, Poland;
| | - Renata Studzińska
- Department of Organic Chemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 2 Jurasza Str., 85-089 Bydgoszcz, Poland;
| |
Collapse
|
2
|
Cominelli G, Sulas F, Pinto D, Rinaldi F, Favero G, Rezzani R. Neuro-Nutritional Approach to Neuropathic Pain Management: A Critical Review. Nutrients 2025; 17:1502. [PMID: 40362812 PMCID: PMC12073121 DOI: 10.3390/nu17091502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 04/15/2025] [Accepted: 04/25/2025] [Indexed: 05/15/2025] Open
Abstract
Pain is a significant global public health issue that can interfere with daily activities, sleep, and interpersonal relationships when it becomes chronic or worsens, ultimately impairing quality of life. Despite ongoing efforts, the efficacy of pain treatments in improving outcomes for patients remains limited. At present, the challenge lies in developing a personalized care and management plan that helps to maintain patient activity levels and effectively manages pain. Neuropathic pain is a chronic condition resulting from damage to the somatosensory nervous system, significantly impacting quality of life. It is partly thought to be caused by inflammation and oxidative stress, and clinical research has suggested a link between this condition and diet. However, these links are not yet well understood and require further investigation to evaluate the pathways involved in neuropathic pain. Specifically, the question remains whether supplementation with dietary antioxidants, such as melatonin, could serve as a potential adjunctive treatment for neuropathic pain modulation. Melatonin, primarily secreted by the pineal gland but also produced by other systems such as the digestive system, is known for its anti-inflammatory, antioxidant, and anti-aging properties. It is found in various fruits and vegetables, and its presence alongside other polyphenols in these foods may enhance melatonin intake and contribute to improved health. The aim of this review is to provide an overview of neuropathic pain and examine the potential role of melatonin as an adjunctive treatment in a neuro-nutritional approach to pain management.
Collapse
Affiliation(s)
- Giorgia Cominelli
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy; (G.C.); (F.S.); (G.F.)
| | - Francesca Sulas
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy; (G.C.); (F.S.); (G.F.)
| | - Daniela Pinto
- Human Microbiome Advanced Project Institute, 20129 Milan, Italy; (D.P.); (F.R.)
- Interdepartmental University Center of Research “Adaption and Regeneration of Tissues and Organs-(ARTO)”, University of Brescia, 25123 Brescia, Italy
| | - Fabio Rinaldi
- Human Microbiome Advanced Project Institute, 20129 Milan, Italy; (D.P.); (F.R.)
- Interdepartmental University Center of Research “Adaption and Regeneration of Tissues and Organs-(ARTO)”, University of Brescia, 25123 Brescia, Italy
| | - Gaia Favero
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy; (G.C.); (F.S.); (G.F.)
- Interdepartmental University Center of Research “Adaption and Regeneration of Tissues and Organs-(ARTO)”, University of Brescia, 25123 Brescia, Italy
| | - Rita Rezzani
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy; (G.C.); (F.S.); (G.F.)
- Interdepartmental University Center of Research “Adaption and Regeneration of Tissues and Organs-(ARTO)”, University of Brescia, 25123 Brescia, Italy
- Italian Society for the Study of Orofacial Pain (Società Italiana Studio Dolore Orofacciale–SISDO), 25123 Brescia, Italy
| |
Collapse
|
3
|
Chen S, Dan L, Xiang L, He Q, Hu D, Gao Y. The role of gut flora-driven Th cell responses in preclinical rheumatoid arthritis. J Autoimmun 2025; 154:103426. [PMID: 40300482 DOI: 10.1016/j.jaut.2025.103426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 03/24/2025] [Accepted: 04/22/2025] [Indexed: 05/01/2025]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune inflammatory disorder with an immune pathogenesis that evolves over decades. Preclinical RA (PreRA) represents a dynamic immune phase preceding clinical RA, marked by the loss of autoimmune tolerance, the appearance of tissue-invasive effector T cells, and the production of autoantibodies (such as antibodies against citrullinated proteins and rheumatoid factors). Extensive research has demonstrated that gut microbiota influence mucosal T-cell responses, driving the progression of PreRA through multiple mechanisms, including altered intestinal permeability, gene-environment interactions, bacterial antigenic specificity, molecular mimicry, and metabolite production. Environmental risk factors such as smoking, hormonal changes, and high-sodium (Na) diets, may contribute to RA pathogenesis via the gut microbiome. The next challenge in RA research lies in developing therapeutic strategies to intervene during the asymptomatic autoimmune phase, where dietary adjustments, natural compounds, probiotics, and other approaches could effectively modulate gut flora to prevent or delay RA onset.
Collapse
Affiliation(s)
- Shuanglan Chen
- Department of Rheumatology and Immunology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Lijuan Dan
- Department of Infection, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Li Xiang
- Department of Rheumatology and Immunology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Qingman He
- Department of Rheumatology and Immunology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Dongsen Hu
- Sichuan Jinxin Xi'nan Women's and Children's Hospital Co., Ltd, Chengdu, 610023, China
| | - Yongxiang Gao
- Department of Rheumatology and Immunology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China.
| |
Collapse
|
4
|
Karaca NB, Kahraman A, Buran S, Tüfekçi O, Kumbaroğlu FB, Bulut Zİ, Bulut S, Barlak A, Doğru A, Kalyoncu U, Akdoğan A, Apraş Bilgen Ş, Kiraz S, Ünal E. Comparison of functionality, mood, and biopsychosocial status in rheumatic patients with and without self-reported bowel problems and the evaluation of BETY's impact on these factors: a retrospective study. Curr Med Res Opin 2025:1-7. [PMID: 40151068 DOI: 10.1080/03007995.2025.2486164] [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: 11/11/2024] [Revised: 03/18/2025] [Accepted: 03/25/2025] [Indexed: 03/29/2025]
Abstract
OBJECTIVE This retrospective study aimed to evaluate the comparison of functionality, mood, and biopsychosocial status in rheumatic patients with and without self-reported bowel problems and the evaluation of Bilişsel Egzersiz Terapi Yaklaşımı (BETY) - (Cognitive Exercise Therapy Approach in English)'s impact on these factors. METHODS The study included 718 patients with eight different diagnoses of rheumatism. The presence of self-reported bowel problems in rheumatic patients was assessed with item 10 of the BETY-Biopsychosocial Questionnaire (BETY-BQ), functionality with the Health Assessment Questionnaire (HAQ), emotional status with the Hospital Anxiety and Depression Scale (HADS), and biopsychosocial status (BPS) with the BETY-BQ. Sixty-five rheumatic patients were included in BETY group exercise sessions for 3 months, 3 days a week. RESULTS The rate of self-reporting bowel problems in the total cohort was 61.6%. Among all BPS parameters examined, a significant difference was found in favor of rheumatic individuals who did not report bowel symptoms (p <.005). All patients included in the BETY sessions achieved improvement in all parameters, including bowel symptoms (p <.005). CONCLUSIONS Many patients suffered from bowel problems. The investigated parameters of rheumatic patients with bowel symptoms were negatively affected. BETY improved all parameters, including bowel symptoms. BPS features should be considered in disease management in rheumatic patients reporting bowel problems. BETY should be used as an exercise intervention based on the BPS model in these patients.
Collapse
Affiliation(s)
- Nur Banu Karaca
- Faculty of Physical Therapy and Rehabilitation, Hacettepe University, Ankara, Turkey
| | - Aysu Kahraman
- Faculty of Physical Therapy and Rehabilitation, Hacettepe University, Ankara, Turkey
| | - Sinan Buran
- Faculty of Physical Therapy and Rehabilitation, Hacettepe University, Ankara, Turkey
| | - Orkun Tüfekçi
- Faculty of Physical Therapy and Rehabilitation, Hacettepe University, Ankara, Turkey
| | | | - Zeynep İrem Bulut
- Faculty of Health Sciences, Department of Physical Therapy and Rehabilitation, Hasan Kalyoncu University, Gaziantep, Turkey
| | - Senem Bulut
- Faculty of Physical Therapy and Rehabilitation, Hacettepe University, Ankara, Turkey
| | - Aysima Barlak
- Faculty of Physical Therapy and Rehabilitation, Hacettepe University, Ankara, Turkey
| | - Atalay Doğru
- Faculty of Medicine, Department of Internal Medicine, Division of Rheumatology, Süleyman Demirel University, Isparta, Turkey
| | - Umut Kalyoncu
- Faculty of Medicine, Department of Internal Medicine, Division of Rheumatology, Hacettepe University, Ankara, Turkey
| | - Ali Akdoğan
- Faculty of Medicine, Department of Internal Medicine, Division of Rheumatology, Hacettepe University, Ankara, Turkey
| | - Şule Apraş Bilgen
- Faculty of Medicine, Department of Internal Medicine, Division of Rheumatology, Hacettepe University, Ankara, Turkey
| | - Sedat Kiraz
- Faculty of Medicine, Department of Internal Medicine, Division of Rheumatology, Hacettepe University, Ankara, Turkey
| | - Edibe Ünal
- Faculty of Physical Therapy and Rehabilitation, Hacettepe University, Ankara, Turkey
| |
Collapse
|
5
|
Charneca S, Hernando A, Almada-Correia I, Polido-Pereira J, Vieira A, Sousa J, Almeida AS, Motta C, Barreto G, Eklund KK, Alonso-Pérez A, Gómez R, Cicci F, Mauro D, Pinho SS, Fonseca JE, Costa-Reis P, Guerreiro CS. TASTY trial: protocol for a study on the triad of nutrition, intestinal microbiota and rheumatoid arthritis. Nutr J 2025; 24:52. [PMID: 40189532 PMCID: PMC11974026 DOI: 10.1186/s12937-025-01089-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Accepted: 02/06/2025] [Indexed: 04/09/2025] Open
Abstract
BACKGROUND The gut microbiota has been implicated in the onset and progression of Rheumatoid Arthritis (RA). It has been proposed that gut dysbiosis impairs gut barrier function, leading to alterations in mucosal integrity and immunity. This disruption allows bacterial translocation, contributing to the perpetuation of the inflammatory process. Since diet is recognised as a key environmental factor influencing the gut microbiota, nutritional interventions targeting RA activity are currently being explored. This study aims to investigate whether a dietary intervention based on a typical Mediterranean Diet enriched with fermented foods (MedDiet +) can impact the gut microbiota, intestinal permeability, and RA-related outcomes. METHODS One hundred RA patients are being recruited at Unidade Local de Saúde (ULS) Santa Maria in Lisbon, Portugal, and randomly assigned to either the intervention (MedDiet +) or the control group. The 12-week nutritional intervention includes a personalised dietary plan following the MedDiet + pattern, along with educational resources, food basket deliveries, and clinical culinary workshops, all developed and monitored weekly by registered dietitians. The control group receives standardised general healthy diet recommendations at baseline. The intervention's effects will be assessed by evaluating disease activity, functional status, quality of life, intestinal permeability, endotoxemia, inflammatory biomarkers, intestinal and oral microbiota, serum proteomics, and serum glycome profile characterisation. DISCUSSION We anticipate obtaining integrative insights into the interplay between diet, the gut, and RA, while also exploring the underlying mechanisms driving these changes. This study, conducted by a multidisciplinary research team of registered dietitians, rheumatologists, biologists, and immunologists, aims to bridge the current gap between nutrition-related knowledge and RA. TRIAL REGISTRATION Registered in ClinicalTrials.gov (NCT06758817; date of registry: January 6th 2025).
Collapse
Affiliation(s)
- Sofia Charneca
- Laboratório de Nutrição, Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
| | - Ana Hernando
- Laboratório de Nutrição, Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
| | - Inês Almada-Correia
- Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
- GIMM - Gulbenkian Institute for Molecular Medicine, Lisbon, Portugal
| | - Joaquim Polido-Pereira
- Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
- GIMM - Gulbenkian Institute for Molecular Medicine, Lisbon, Portugal
- Serviço de Reumatologia, ULS Santa Maria, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
| | - Adriana Vieira
- GIMM - Gulbenkian Institute for Molecular Medicine, Lisbon, Portugal
| | - Joana Sousa
- Laboratório de Nutrição, Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
- Instituto de Saúde Ambiental, Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
| | - Ana Santos Almeida
- Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
- GIMM - Gulbenkian Institute for Molecular Medicine, Lisbon, Portugal
| | - Carla Motta
- Departamento de Alimentação e Nutrição, Instituto Nacional de Saúde Doutor Ricardo Jorge, Lisbon, Portugal
| | - Gonçalo Barreto
- Clinicum, Faculty of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, 00029, Finland.
| | - Kari K Eklund
- Clinicum, Faculty of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, 00029, Finland
- Department of Rheumatology, Helsinki University Hospital and Helsinki University, Helsinki, Finland
| | - Ana Alonso-Pérez
- Musculoskeletal Pathology Group, Institute IDIS, Santiago University Clinical Hospital, 15706, Santiago de Compostela, Spain
| | - Rodolfo Gómez
- Musculoskeletal Pathology Group, Institute IDIS, Santiago University Clinical Hospital, 15706, Santiago de Compostela, Spain
| | - Francesco Cicci
- Dipartimento di Medicina di Precisione, Università Della Campania L. Vanvitelli, Naples, Italy
| | - Daniele Mauro
- Dipartimento di Medicina di Precisione, Università Della Campania L. Vanvitelli, Naples, Italy
| | - Salomé S Pinho
- Institute for Research and Innovation in Health (i3s), University of Porto, Porto, Portugal
- School of Medicine and Biomedical Sciences (ICBAS), University of Porto, Porto, Portugal
- Faculty of Medicine, University of Porto, Porto, Portugal
| | - João Eurico Fonseca
- Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
- GIMM - Gulbenkian Institute for Molecular Medicine, Lisbon, Portugal
- Serviço de Reumatologia, ULS Santa Maria, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
| | - Patrícia Costa-Reis
- Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
- GIMM - Gulbenkian Institute for Molecular Medicine, Lisbon, Portugal
- Pediatric Rheumatology Unit, ULS Santa Maria, Lisbon, Portugal
| | - Catarina Sousa Guerreiro
- Laboratório de Nutrição, Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
- Instituto de Saúde Ambiental, Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
| |
Collapse
|
6
|
Guo J, Zhao Y, Guo W, Sun Y, Zhang W, Zhao Q, Zhang Y, Jiang Y. Effects of Lactobacillus paracei JY062 Postbiotic on Intestinal Barrier, Immunity, and Gut Microbiota. Nutrients 2025; 17:1272. [PMID: 40219029 PMCID: PMC11990213 DOI: 10.3390/nu17071272] [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: 02/18/2025] [Revised: 03/21/2025] [Accepted: 04/03/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND/OBJECTIVES Research on postbiotics derived from probiotic fermented milk bases require further expansion, and the mechanisms through which they exert their effects have yet to be fully elucidated. This study utilized in vitro cell co-culture, digestion, and fermentation experiments, combined with targeted T500 technology, to elucidate the mechanism by which postbiotic Pa JY062 safeguards intestinal health. Compared to the LPS group, Pa JY062 boosted phagocytic ability in RAW264.7 macrophages, decreased NO levels, and alleviated LPS-induced excessive inflammation. Pa JY062 suppressed pro-inflammatory cytokines (IL-6, IL-17α, and TNF-α) while elevating anti-inflammatory IL-10. It prevented LPS-induced TEER reduction in Caco-2 monolayers, decreased FITC-dextran permeability, restored intestinal microvilli integrity, and upregulated tight junction genes (ZO-1, occludin, claudin-1, and E-cadherin). The hydrolysis rate of Pa JY062 progressively rose in gastrointestinal fluids in 0-120 min. At 5 mg/mL, it enriched gut microbiota diversity and elevated proportions of Limosilactobacillus, Lactobacillus, Pediococcus, and Lacticaseibacillus while augmenting the microbial production of acetic acid (120.2 ± 8.08 μg/mL), propionic acid (9.9 ± 0.35 μg/mL), and butyric acid (10.55 ± 0.13 μg/mL). Pa JY062 incorporated αs-casein/β-lactoglobulin hydrolysate (L-glutamic acid, alanine, lysine, tyrosine, phenylalanine, histidine, and arginine) to mitigate protein allergenic potential while harboring bioactive components, including tryptophan metabolites, vitamin B6 (VB6), and γ-aminobutyric acid (GABA). Pa JY062 represented a novel postbiotic with demonstrated intestinal health-promoting properties. These findings advance the current knowledge on postbiotic-mediated gut homeostasis regulation and expedite the translational development of dairy-derived postbiotic formulations.
Collapse
Affiliation(s)
- Jinfeng Guo
- Key Lab of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China; (J.G.); (Y.Z.); (W.G.); (Y.S.); (W.Z.); (Q.Z.)
| | - Ying Zhao
- Key Lab of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China; (J.G.); (Y.Z.); (W.G.); (Y.S.); (W.Z.); (Q.Z.)
| | - Wenqian Guo
- Key Lab of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China; (J.G.); (Y.Z.); (W.G.); (Y.S.); (W.Z.); (Q.Z.)
| | - Yilin Sun
- Key Lab of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China; (J.G.); (Y.Z.); (W.G.); (Y.S.); (W.Z.); (Q.Z.)
| | - Wei Zhang
- Key Lab of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China; (J.G.); (Y.Z.); (W.G.); (Y.S.); (W.Z.); (Q.Z.)
| | - Qianyu Zhao
- Key Lab of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China; (J.G.); (Y.Z.); (W.G.); (Y.S.); (W.Z.); (Q.Z.)
| | - Yu Zhang
- Key Lab of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China; (J.G.); (Y.Z.); (W.G.); (Y.S.); (W.Z.); (Q.Z.)
| | - Yujun Jiang
- Key Lab of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China; (J.G.); (Y.Z.); (W.G.); (Y.S.); (W.Z.); (Q.Z.)
- Food Laboratory of Zhongyuan, Luohe 462300, China
| |
Collapse
|
7
|
Rodolfi S, Selmi C. Environmental factors and rheumatic diseases. Best Pract Res Clin Rheumatol 2025:102053. [PMID: 40140341 DOI: 10.1016/j.berh.2025.102053] [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: 02/10/2025] [Accepted: 02/26/2025] [Indexed: 03/28/2025]
Abstract
The pathogenesis and pathophysiology of rheumatic diseases is complex and relies on the interaction of different factors. The common view is that the pathological autoimmunity develops in genetically predisposed individuals upon exposure to an environmental trigger. This highlights the importance of recognizing and deconstructing the effects of environmental agents in rheumatic diseases. Several factors have been identified in the last decades, with detrimental or protective effects, impacting not only on disease onset, but also on its natural history. Cigarette smoking has been identified as one of the strongest environmental risk factors, being associated with disease development and severity for several rheumatic diseases, including rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), and spondyloarthropathies. Moreover, other airborne pollutants, such as silica, solvents, asbestos and metals are recognized risk factors for rheumatic diseases. The effect of some other agents is however not straightforward, of which a remarkable example is alcohol consumption. Alcohol has been associated with both pro- and anti-inflammatory effects, exerting a variable effect on rheumatic diseases depending on quantity and frequency of consumption, as well as sex and ethnicity. Similarly, ultraviolet light exposure has been associated with a higher risk of SLE but lower risk of RA. The relationship between microbial exposure and autoimmunity is also complex: while some infectious agents increase the risk of rheumatic diseases, it is widely accepted that less exposure to microbial agents, particularly during immune system development, increases the risk of autoimmunity. Furthermore, in recent years the spotlight has switched to the human microbiome, as alterations in organ-specific microbiome composition are anticipated to be early participants in the onset of immune-mediated illnesses. The aim of this review is to highlight the most relevant environmental factors and their role in Rheumatology, with a specific focus on proposed pathophysiological effect and correlation with clinical outcomes.
Collapse
Affiliation(s)
- Stefano Rodolfi
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy; Department of Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Carlo Selmi
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy; Department of Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, Rozzano, Italy.
| |
Collapse
|
8
|
Momo Cabrera P, Bokulich NA, Zimmermann P. Evaluating stool microbiome integrity after domestic freezer storage using whole-metagenome sequencing, genome assembly, and antimicrobial resistance gene analysis. Microbiol Spectr 2025; 13:e0227824. [PMID: 39932403 PMCID: PMC11878046 DOI: 10.1128/spectrum.02278-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 01/04/2025] [Indexed: 03/05/2025] Open
Abstract
The gut microbiome is crucial for host health. Early childhood is a critical period for the development of a healthy gut microbiome, but it is particularly sensitive to external influences. Recent research has focused on using advanced techniques like shotgun metagenome sequencing to identify key microbial signatures and disruptions linked to disease. For accurate microbiome analysis, samples need to be collected and stored under specific conditions to preserve microbial integrity and composition, with -80°C storage considered the gold standard for stabilization. This study investigates the effect of domestic freezer storage on the microbial composition of stool samples obtained from 20 children under 4 years of age with the use of shotgun metagenome sequencing. Fresh stool samples were aliquoted into sterile tubes, with one aliquot stored at 4°C and analyzed within 24 hours, while others were frozen in domestic freezers (below -18°C) and analyzed after 1 week, 2 months, and 6 months. Assessments of contig assembly quality, microbial diversity, and antimicrobial resistance genes revealed no significant degradation or variation in microbial composition. IMPORTANCE Most prior studies on sample storage have relied on amplicon sequencing, which is less applicable to metagenome sequencing-given considerations of contig quality and functional gene detection-and less reliable in representing microbial composition. Moreover, the effects of domestic freezer storage for at-home stool collection on microbiome profiles, contig quality, and antimicrobial resistance gene profiles have not been previously investigated. Our findings suggest that stool samples stored in domestic freezers for up to 6 months maintain the integrity of metagenomic data. These findings indicate that domestic freezer storage does not compromise the integrity or reproducibility of metagenomic data, offering a reliable and accessible alternative for temporary sample storage. This approach enhances the feasibility of large-scale at-home stool collection and citizen science projects, even those focused on the more easily perturbed early life microbiome. This advancement enables more inclusive research into the gut microbiome, enhancing our understanding of its role in human health.
Collapse
Affiliation(s)
- Paula Momo Cabrera
- Department of Community Health, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
- Laboratory of Food Systems Biotechnology, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Nicholas A. Bokulich
- Laboratory of Food Systems Biotechnology, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Petra Zimmermann
- Department of Community Health, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
- Department of Paediatrics, Fribourg Hospital, Fribourg, Switzerland
- Infectious Diseases Research Group, Murdoch Children’s Research Institute, Parkville, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Australia
| |
Collapse
|
9
|
Shiro Y, Arai YC, Nakaso Y, Sakurai H, Inoue M, Owari K, Sato J, Ikemoto T, Ushida T. Differences in Gut Microbiota Composition Depending on the Site of Pain in Patients with Chronic Pain. J Pain Res 2025; 18:769-782. [PMID: 39991525 PMCID: PMC11846523 DOI: 10.2147/jpr.s494984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 01/31/2025] [Indexed: 02/25/2025] Open
Abstract
Background There are many factors associated with chronic pain, including changes in the nervous and musculoskeletal systems and so on. Recently, it has become clear that the gut microbiota (GM) influences these factors, and there are many reports of GM dysbiosis in patients with chronic pain. However, the relationship between pain and GM remains unclear. Our previous study reported that defecation status, which reflects GM composition, was associated with pain intensity and that this relationship was different for each pain site. Our study investigated the association between pain site and the GM composition of feces in chronic pain patients. Methods The subjects were 136 patients with chronic pain and 125 healthy controls. Patients were classified into four groups, whole body (WB) pain, lower back and lower extremity (LL) pain, headache, and upper back and upper extremity pain, based on the site of pain, and we investigated differences in GM taxonomy groups compared with healthy subject. Results Chronic pain patients had a lower alpha diversity (effect size=0.16, p=0.02). But each pain site group did not differ in alpha diversity. WB pain patients showed higher Eggerthellaceae (LDA=3.09, p<0.01) and lower Halomonas (LDA =-2.72, p<0.01). LL pain patients had increased Fusobacterium and Sellimonas (LDA=4.09,3.03 p<0.01, 0.01) but reduced Halomonas (LDA=-2.59, p<0.01), and other key taxa. Conclusion WB and LL patients may have GM compositions different from healthy controls, but larger studies are needed to confirm this.
Collapse
Affiliation(s)
- Yukiko Shiro
- Department of Pain Medicine, Aichi Medical University, Aichi, Japan
- Department of Physical Therapy, Faculty of Rehabilitation Sciences, Nagoya Gakuin University, Aichi, Japan
| | - Young-Chang Arai
- Department of Pain Medicine, Aichi Medical University, Aichi, Japan
| | - Yuichiro Nakaso
- Department of Pain Medicine, Aichi Medical University, Aichi, Japan
| | - Hiroki Sakurai
- Department of Pain Medicine, Aichi Medical University, Aichi, Japan
- Faculty of Health and Medical Sciences, Tokoha University, Shizuoka, Japan
| | - Masayuki Inoue
- Department of Pain Medicine, Aichi Medical University, Aichi, Japan
| | - Keiko Owari
- Department of Pain Medicine, Aichi Medical University, Aichi, Japan
| | - Jun Sato
- Department of Pain Medicine, Aichi Medical University, Aichi, Japan
| | - Tatsunori Ikemoto
- Department of Orthopedics, School of Medicine, Aichi Medical University, Aichi, Japan
| | - Takahiro Ushida
- Department of Pain Medicine, Aichi Medical University, Aichi, Japan
| |
Collapse
|
10
|
Deng X, Gong X, Zhou D, Hong Z. Perturbations in gut microbiota composition in patients with autoimmune neurological diseases: a systematic review and meta-analysis. Front Immunol 2025; 16:1513599. [PMID: 39981228 PMCID: PMC11839609 DOI: 10.3389/fimmu.2025.1513599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 01/16/2025] [Indexed: 02/22/2025] Open
Abstract
Studies suggest that gut dysbiosis occurs in autoimmune neurological diseases, but a comprehensive synthesis of the evidence is lacking. Our aim was to systematically review and meta-analyze the correlation between the gut microbiota and autoimmune neurological disorders to inform clinical diagnosis and therapeutic intervention. We searched the databases of PubMed, Embase, Web of Science, and the Cochrane Library until 1 March 2024 for research on the correlation between gut microbiota and autoimmune neurological disorders. A total of 62 studies provided data and were included in the analysis (n = 3,126 patients, n = 2,843 healthy individuals). Among the included studies, 42 studies provided data on α-diversity. Regarding α-diversity, except for Chao1, which showed a consistent small decrease (SMD = -0.26, 95% CI = -0.45 to -0.07, p < 0.01), other indices demonstrated no significant changes. While most studies reported significant differences in β-diversity, consistent differences were only observed in neuromyelitis optica spectrum disorders. A decrease in short-chain fatty acid (SCFA)-producing bacteria, including Faecalibacterium and Roseburia, was observed in individuals with autoimmune encephalitis, neuromyelitis optica spectrum disorders, myasthenia gravis, and multiple sclerosis. Conversely, an increase in pathogenic or opportunistic pathogens, including Streptococcus and Escherichia-Shigella, was observed in these patients. Subgroup analyses assessed the confounding effects of geography and immunotherapy use. These findings suggest that disturbances of the gut flora are associated with autoimmune neurological diseases, primarily manifesting as non-specific and shared microbial alterations, including a reduction in SCFA-producing bacteria and an increase in pathogenic or opportunistic pathogens. Systematic review registration https://www.crd.york.ac.uk/prospero/, identifier CRD42023410215.
Collapse
Affiliation(s)
- Xiaolin Deng
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Neurology, Chengdu Shangjin Nanfu Hospital, Chengdu, Sichuan, China
| | - Xue Gong
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Neurology, Chengdu Shangjin Nanfu Hospital, Chengdu, Sichuan, China
| | - Dong Zhou
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhen Hong
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Neurology, Chengdu Shangjin Nanfu Hospital, Chengdu, Sichuan, China
| |
Collapse
|
11
|
Jiang Y, Huang Z, Sun W, Huang J, Xu Y, Liao Y, Jin T, Li Q, Ho IHT, Zou Y, Zhu W, Li Q, Qin F, Zhang X, Shi S, Zhang N, Yang S, Xie W, Wu S, Tan L, Zhang L, Chen H, Gin T, Chan MTV, Wu WKK, Xiao L, Liu X. Roseburia intestinalis-derived butyrate alleviates neuropathic pain. Cell Host Microbe 2025; 33:104-118.e7. [PMID: 39706182 DOI: 10.1016/j.chom.2024.11.013] [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: 04/02/2024] [Revised: 10/30/2024] [Accepted: 11/25/2024] [Indexed: 12/23/2024]
Abstract
Approximately 20% of patients with shingles develop postherpetic neuralgia (PHN). We investigated the role of gut microbiota in shingle- and PHN-related pain. Patients with shingles or PHN exhibited significant alterations in their gut microbiota with microbial markers predicting PHN development among patients with shingles. Functionally, fecal microbiota transplantation from patients with PHN to mice heightened pain sensitivity. Administration of Roseburia intestinalis, a bacterium both depleted in patients with shingles and PHN, alleviated peripheral nerve injury-induced pain in mice. R. intestinalis enhanced vagal neurotransmission to the nucleus tractus solitarius (NTS) to suppress the central amygdala (CeA), a brain region involved in pain perception. R. intestinalis-generated butyrate activated vagal neurons through the receptor, G protein-coupled receptor 41 (GPR41). Vagal knockout of Gpr41 abolished the effects of R. intestinalis on the NTS-CeA circuit and reduced pain behaviors. Overall, we established a microbiota-based model for PHN risk assessment and identified R. intestinalis as a potential pain-alleviating probiotic.
Collapse
Affiliation(s)
- Yanjun Jiang
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ziheng Huang
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Wuping Sun
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, Shenzhen Nanshan People's Hospital and the 6th Affiliated Hospital of Shenzhen University Medical School, National Key Clinical Pain Medicine of China, Shenzhen 518060, China
| | - Jiabin Huang
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, Shenzhen Nanshan People's Hospital and the 6th Affiliated Hospital of Shenzhen University Medical School, National Key Clinical Pain Medicine of China, Shenzhen 518060, China
| | - Yunlong Xu
- Shenzhen Key Laboratory of Drug Addiction, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Department of Neonatology, Shenzhen Maternity & Child Healthcare Hospital, The First School of Clinical Medicine, Southern Medical University, Shenzhen 518055, China
| | - Yuliang Liao
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, Shenzhen Nanshan People's Hospital and the 6th Affiliated Hospital of Shenzhen University Medical School, National Key Clinical Pain Medicine of China, Shenzhen 518060, China
| | - Tingting Jin
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Qing Li
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China; Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Idy Hiu Ting Ho
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yidan Zou
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Wenyi Zhu
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Qian Li
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Fenfen Qin
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Xinyi Zhang
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Shuqi Shi
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, Shenzhen Nanshan People's Hospital and the 6th Affiliated Hospital of Shenzhen University Medical School, National Key Clinical Pain Medicine of China, Shenzhen 518060, China
| | - Na Zhang
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Shaomin Yang
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, Shenzhen Nanshan People's Hospital and the 6th Affiliated Hospital of Shenzhen University Medical School, National Key Clinical Pain Medicine of China, Shenzhen 518060, China
| | - Wenhui Xie
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, Shenzhen Nanshan People's Hospital and the 6th Affiliated Hospital of Shenzhen University Medical School, National Key Clinical Pain Medicine of China, Shenzhen 518060, China
| | - Songbin Wu
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, Shenzhen Nanshan People's Hospital and the 6th Affiliated Hospital of Shenzhen University Medical School, National Key Clinical Pain Medicine of China, Shenzhen 518060, China
| | - Likai Tan
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Lin Zhang
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Microbiota I-Center (MagIC), The Chinese University of Hong Kong, Hong Kong SAR, China; Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Huarong Chen
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Tony Gin
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Matthew Tak Vai Chan
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - William Ka Kei Wu
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China; State Key Laboratory of Digestive Disease Institute of Digestive Disease and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - Lizu Xiao
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, Shenzhen Nanshan People's Hospital and the 6th Affiliated Hospital of Shenzhen University Medical School, National Key Clinical Pain Medicine of China, Shenzhen 518060, China.
| | - Xiaodong Liu
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
12
|
Li R, Chen X, Shi C, Zhu Y. Study on the Effect of Radish Sprouts on Short-Chain Fatty Acids and Gut Microbial Diversity in Healthy Individuals. Foods 2025; 14:170. [PMID: 39856836 PMCID: PMC11765271 DOI: 10.3390/foods14020170] [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: 12/02/2024] [Revised: 12/18/2024] [Accepted: 01/04/2025] [Indexed: 01/27/2025] Open
Abstract
This study aimed to assess the impact of radish sprouts on the gut microbiota of healthy individuals. Radish sprout additives, subjected to short-term storage and steam treatment, were used to intervene in an in vitro culture of human gut microbiota. The influence of radish sprouts on the gut microbiota was evaluated by monitoring short-chain fatty acid (SCFA) content and proportion in the fermentation broth, and microbial diversity was assessed using 16S rDNA amplicon sequencing. The results indicated that the gut microbiota produced a substantial amount of SCFA within 48 h of fermentation, with a right-skewed distribution across all groups. The addition of both digestates enhanced Firmicutes diversity, while Bacteroidetes and Proteobacteria diversity remained stable between the control and fresh sprout groups. The 30 s steam treatment group showed an increase in Bacteroidetes and a decrease in Proteobacteria diversity. The abundance of Bacilli, Bacillaceae, and Bacillus was significantly higher in both the fresh and steam-treated groups compared to the control. Both fresh and steam-treated radish sprout digestates enriched gut microbiota diversity, with steam treatment showing superior effects. These findings suggest that radish sprout consumption may positively influence gut microbiota, with steam treatment potentially enhancing these benefits.
Collapse
Affiliation(s)
- Ru Li
- College of Food and Biological Engineering, Xuzhou Institute of Technology, Xuzhou 221018, China; (R.L.); (X.C.); (C.S.)
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Xuehong Chen
- College of Food and Biological Engineering, Xuzhou Institute of Technology, Xuzhou 221018, China; (R.L.); (X.C.); (C.S.)
| | - Cong Shi
- College of Food and Biological Engineering, Xuzhou Institute of Technology, Xuzhou 221018, China; (R.L.); (X.C.); (C.S.)
| | - Yi Zhu
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| |
Collapse
|
13
|
Duong QA, Curtis N, Zimmermann P. The association between prenatal antibiotic exposure and adverse long-term health outcomes in children: A systematic review and meta-analysis. J Infect 2025; 90:106377. [PMID: 39675435 DOI: 10.1016/j.jinf.2024.106377] [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: 07/06/2024] [Revised: 12/05/2024] [Accepted: 12/07/2024] [Indexed: 12/17/2024]
Abstract
BACKGROUND Antibiotics are the most commonly prescribed drugs during pregnancy. The long-term health risks to children associated with prenatal antibiotic exposure are uncertain. OBJECTIVE To identify the association between prenatal antibiotics and adverse long-term health outcomes in children. METHODS A systematic search was done to identify original studies investigating the association between prenatal antibiotic exposure and adverse long-term health outcomes in children. Studies were excluded if: (i) antibiotics were only given during delivery or (ii) the outcome was present before antibiotic exposure. RESULTS We included 158 studies, reporting 23 outcomes in 21,943,763 children, in our analysis. For the following adverse health outcomes, there was a significant association with antibiotic exposure found in two or more studies: atopic dermatitis (OR 1.27, 95% CI 1.06-1.52, p=0.01), food allergies (OR 1.25, 95% CI 1.09-1.44, p<0.01), allergic rhinoconjunctivitis (OR 1.16, 95% CI 1.15-1.17, p<0.01), wheezing (OR 1.39, 95% CI 1.14-1.69, p<0.01), asthma (OR 1.36, 95% CI 1.24-1.50, p<0.01), obesity (OR 1.36, 95% CI 1.12-1.64, p<0.01), cerebral palsy (OR 1.25, 95% CI 1.10-1.43, p<0.01), epilepsy or febrile seizure (OR 1.16, 95% CI 1.08-1.24, p<0.01), and cancer (OR 1.13, 95% CI 1.01-1.26, p=0.04). CONCLUSION Although causality cannot be implied, these findings support antibiotic stewardship efforts to ensure judicious use of antibiotics during pregnancy to avoid potential long-term health risks.
Collapse
Affiliation(s)
- Quynh A Duong
- Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Nigel Curtis
- Department of Paediatrics, The University of Melbourne, Parkville, Australia; Infectious Diseases Research Group, Murdoch Children's Research Institute, Parkville, Australia; Infectious Diseases Unit, The Royal Children's Hospital Melbourne, Parkville, Australia
| | - Petra Zimmermann
- Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland; Department of Paediatrics, The University of Melbourne, Parkville, Australia; Infectious Diseases Research Group, Murdoch Children's Research Institute, Parkville, Australia; Department of Paediatrics, Fribourg Hospital HFR, Fribourg, Switzerland.
| |
Collapse
|
14
|
Liu Q, Liu Y, Feng H, Zhao L, Wan T. Exploring genetic associations in systemic lupus erythematosus through Mendelian randomization: implications for novel biomarkers and therapeutic targets. Clin Rheumatol 2025; 44:193-205. [PMID: 39126578 DOI: 10.1007/s10067-024-07094-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/01/2024] [Accepted: 07/26/2024] [Indexed: 08/12/2024]
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease characterized by a significant health burden. There is an essential need for novel biomarkers and therapeutic targets to improve diagnosis and management. Mendelian randomization (MR) was applied to explore causal links between SLE and various biomarkers like immune cells, metabolites, and inflammatory cytokines using multiple databases. Initially, biomarkers significantly associated with SLE were identified. Bidirectional MR helped clarify these relationships, and a two-step mediation MR examined their effects on SLE risk. Intersection analysis was used to identify biomarkers with consistent effects across datasets. Four biomarkers were identified as having significant associations with SLE risk: 1-palmitoyl-2-arachidonoyl-GPI levels [odds ratio (OR), 1.379; 95% confidence interval (CI), 1.180 to 1.613; FDR, 0.046], IL-17A levels (OR, 2.197; 95% CI, 1.412 to 3.418; FDR, 0.044), N-acetyl-aspartyl-glutamate (NAAG) levels (OR, 0.882; 95% CI, 0.831 to 0.936; FDR, 0.030), and ribitol levels (OR, 0.743; 95% CI, 0.644 to 0.857; FDR, 0.012). Bidirectional MR showed an inverse effect of NAAG on IL-17A levels (OR, 0.978; 95% CI, 0.962 to 0.994; p = 0.006). Mediation analysis indicated that NAAG influenced SLE risk both directly (beta = - 0.108) and indirectly through IL-17A (beta = - 0.018), highlighting the potential mediating role of IL-17A. After expanding the significance criteria to p < 0.05, intersection analysis across multiple datasets revealed 29 biomarkers with consistent beta directions, including 19 potential risk factors (beta > 0) and 10 protective factors (beta < 0) for SLE. This research has revealed significant genetic associations with SLE and demonstrated that IL-17A mediates the relationship between NAAG levels and SLE risk, highlighting potential new targets for personalized therapeutic interventions. Key Points • This study employs MR to identify significant genetic associations between various biomarkers and SLE, providing novel insights into potential biomarkers and therapeutic targets. • Four key biomarkers were identified as significantly associated with SLE risk: 1-palmitoyl-2-arachidonoyl-GPI, IL-17A, N-acetyl-aspartyl-glutamate (NAAG), and ribitol. • The findings suggest that NAAG levels have a protective effect against SLE, partly mediated through IL-17A, indicating a complex interplay between these biomarkers in the pathogenesis of SLE. • Intersectional analysis across multiple datasets revealed 29 biomarkers with consistent effects on SLE risk, highlighting new directions for future research and potential personalized therapeutic strategies.
Collapse
Affiliation(s)
- Qi Liu
- Department of Hepatobiliary Surgery, 8th Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China
- Medical School of Chinese People's Liberation Army, Beijing, China
| | - Yuyang Liu
- Department of Neurosurgery, 920th Hospital of Joint Logistics Support Force, Kunming, China
| | - Hui Feng
- Department of Zhantansi Outpatient, Jingzhong Medical District of Chinese People's Liberation Army General Hospital, Beijing, China
| | - Lin Zhao
- Department of Endocrinology, 8th Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China
| | - Tao Wan
- Department of Hepatobiliary Surgery, 8th Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China.
| |
Collapse
|
15
|
Tsuji K, Uchida N, Nakanoh H, Fukushima K, Haraguchi S, Kitamura S, Wada J. The Gut-Kidney Axis in Chronic Kidney Diseases. Diagnostics (Basel) 2024; 15:21. [PMID: 39795549 PMCID: PMC11719742 DOI: 10.3390/diagnostics15010021] [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: 11/11/2024] [Revised: 12/23/2024] [Accepted: 12/24/2024] [Indexed: 01/13/2025] Open
Abstract
The gut-kidney axis represents the complex interactions between the gut microbiota and kidney, which significantly impact the progression of chronic kidney disease (CKD) and overall patient health. In CKD patients, imbalances in the gut microbiota promote the production of uremic toxins, such as indoxyl sulfate and p-cresyl sulfate, which impair renal function and contribute to systemic inflammation. Mechanisms like endotoxemia, immune activation and oxidative stress worsen renal damage by activating pro-inflammatory and oxidative pathways. Insights into these mechanisms highlight the impact of gut-derived metabolites, bacterial translocation, and immune response changes on kidney health, suggesting new potential approaches for CKD treatment. Clinical applications, such as dietary interventions, prebiotics, probiotics and fecal microbiota transplantation, are promising in adjusting the gut microbiota to alleviate CKD symptoms and slow disease progression. Current research highlights the clinical relevance of the gut-kidney axis, but further study is essential to clarify these mechanisms' diagnostic biomarkers and optimize therapeutic interventions. This review emphasizes the importance of an integrated approach to CKD management, focusing on the gut microbiota as a therapeutic target to limit kidney injury.
Collapse
Affiliation(s)
- Kenji Tsuji
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Okayama 700-8558, Japan
| | - Naruhiko Uchida
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Okayama 700-8558, Japan
| | - Hiroyuki Nakanoh
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Okayama 700-8558, Japan
| | - Kazuhiko Fukushima
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Okayama 700-8558, Japan
| | - Soichiro Haraguchi
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Okayama 700-8558, Japan
- Department of Nephrology, Aoe Clinic, Okayama 700-8607, Japan
| | - Shinji Kitamura
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Okayama 700-8558, Japan
- Department of Nursing Science, Faculty of Health and Welfare Science, Okayama Prefectural University, Okayama 719-1197, Japan
| | - Jun Wada
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Okayama 700-8558, Japan
| |
Collapse
|
16
|
Huo C, Li S, Liu A, Hong Y, Zhu Y. Acupuncture ameliorates synovitis in mice with collagen-induced arthritis by repressing ferroptosis via butyric acid. Int Immunopharmacol 2024; 143:113342. [PMID: 39405932 DOI: 10.1016/j.intimp.2024.113342] [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: 06/19/2024] [Revised: 09/29/2024] [Accepted: 10/04/2024] [Indexed: 10/30/2024]
Abstract
It has been reported that the symptoms of rheumatoid arthritis (RA) can be ameliorated by acupuncture, an external treatment of traditional Chinese medicine. However, the immune mechanism underlying its action is elusive. Accordingly, this study investigated the role and mechanism of manual acupuncture (MA) in collagen-induced arthritis (CIA) in mice. The results demonstrated that MA or NaB treatment reduced Articular Index scores and right paw thickness and alleviated synovial inflammation and cartilage damage. MA or NaB treatment altered the content and relative abundance of short-chain fatty acids, particularly butyric and propionic acids, in feces. Additionally, MA or NaB treatment elevated SCD1, SREBP1, and GPX4 protein expression in synovial tissues and GSH-px contents in serum while decreasing ROS fluorescence intensity and MDA contents in peripheral blood. A linear correlation was found between the relative expression of SCD1 and SREBP1 in synovial tissues and the contents of propionic acids and butyric acids in feces, as well as between the contents of propionic acids and butyric acids. In summary, MA regulates butyric acids to inhibit ferroptosis, therefore suppressing inflammation in RA.
Collapse
Affiliation(s)
- Chenlu Huo
- Anhui University of Chinese Medicine, Hefei 230031, China
| | - Shuang Li
- Anhui University of Chinese Medicine, Hefei 230031, China
| | - Anting Liu
- Anhui University of Chinese Medicine, Hefei 230031, China
| | - Yujie Hong
- Anhui University of Chinese Medicine, Hefei 230031, China
| | - Yan Zhu
- The Geriatrics, The Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230061, China.
| |
Collapse
|
17
|
Xie C, Liu X, Li W, Yao Z, Men H, Li Z. The role of miRNAs as biomarkers in heterotopic ossification. EFORT Open Rev 2024; 9:1120-1133. [PMID: 39620561 PMCID: PMC11619732 DOI: 10.1530/eor-22-0100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/08/2024] Open
Abstract
Fibrodysplasia ossificans progressiva and progressive osseous heteroplasia are genetic forms of heterotopic ossification (HO). Fibrodysplasia ossificans progressiva is caused by ACVR1 gene mutations, while progressive osseous heteroplasia is caused by GNAS gene mutations. Nongenetic HO typically occurs after trauma or surgery, with an occurrence rate of 20-60%. It can also be observed in conditions such as diffuse idiopathic skeletal hyperostosis, spinal ligament ossification, ankylosing spondylitis, and skeletal fluorosis. The exact cause of nongenetic HO is not entirely clear. More than 100 types of miRNAs have been identified as being linked to the development of HO. Some miRNAs are promising potential biomarkers for traumatic HO and ossification of the posterior longitudinal ligament. These findings further emphasize the significant role miRNAs play in the pathogenesis and progression of bone disorders. Repeated investigations into the function of a specific miRNA are infrequent and yield inconsistent results, possibly because of variable experimental conditions. It is hypothesized that miRNAs can enhance osteogenesis for the management of fractures and bone defects. However, further research is required to validate this hypothesis.
Collapse
Affiliation(s)
- Chen Xie
- Trauma center, The 960th Hospital of PLA, Jinan, Shandong, China
| | - Xiao Liu
- Department of Basic Medical Sciences, The 960th Hospital of PLA, Jinan, Shandong, China
| | - Wenbao Li
- Trauma center, The 960th Hospital of PLA, Jinan, Shandong, China
| | - Zhaozhe Yao
- Trauma center, The 960th Hospital of PLA, Jinan, Shandong, China
| | - Hongyue Men
- Trauma center, The 960th Hospital of PLA, Jinan, Shandong, China
| | - Zongyu Li
- Trauma center, The 960th Hospital of PLA, Jinan, Shandong, China
| |
Collapse
|
18
|
Levinson J, Buehring B. Pitfalls and pearls in diagnosing inflammatory arthritis in older patients. Joint Bone Spine 2024; 91:105719. [PMID: 38452885 DOI: 10.1016/j.jbspin.2024.105719] [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: 12/18/2023] [Revised: 02/22/2024] [Accepted: 02/26/2024] [Indexed: 03/09/2024]
Abstract
Given current demographic shifts, the number of older adults continues to grow, with almost half of patients over 65 being diagnosed with some form of arthritis. Rheumatic diseases pose unique diagnostic challenges in older patients due to the convergence of physiologic changes of aging, confounding difficulties to care, and atypical disease manifestations. This review summarizes the current published evidence to guide clinicians in evaluating geriatric patients with rheumatologic concerns, focusing on inflammatory arthritis. Using the background of epidemiologic data on various musculoskeletal diseases, clinical presentations, current diagnostic tests, and known physiologic changes of aging, this review highlights five diagnostic pitfalls in inflammatory polyarthritis among older patients. The pitfalls include: 1) broader differential diagnosis; 2) atypical presentations; 3) communication, cognitive, and social impairments; 4) the role of chronological vs. biological age; and 5) anchoring bias by assuming older adults are simply "older young adults". These pitfalls are discussed in the context of geriatric principles such as the "hallmarks of aging" and the expected pathophysiologic changes of organ systems. Furthermore, the review discusses the strengths and weaknesses of diagnostic tests used in arthritis and introduces some of the geriatric assessment tools that systematically evaluate multimorbidity and geriatric syndromes. With familiarity of the potential diagnostic pitfalls, knowledge of both normal and pathologic aging processes, awareness of the difference between biological and chronological age, and the ability to use geriatric assessment tools to better characterize older patients, clinicians will be better able to diagnose and manage rheumatic conditions in this population.
Collapse
Affiliation(s)
- Justin Levinson
- Rheumatology Division, Department of Medicine, University of Wisconsin School of Medicine and Public Health, 1685, Highland avenue, Madison, WI, USA.
| | - Bjoern Buehring
- Division of Rheumatology, Krankenhaus Sankt Josef, Wuppertal, Germany.
| |
Collapse
|
19
|
Cuevas-Martínez R, González-Chávez SA, Bermúdez M, Salas-Leiva JS, Vázquez-Olvera G, Hinojos-Gallardo LC, Chaparro-Barrera E, Pacheco-Silva C, Romero-Sánchez C, Villegas-Mercado CE, Pacheco-Tena C. Intermittent fasting reduces inflammation and joint damage in a murine model of rheumatoid arthritis: insights from transcriptomic and metagenomic analyses. BMC Rheumatol 2024; 8:64. [PMID: 39587696 PMCID: PMC11587710 DOI: 10.1186/s41927-024-00436-0] [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: 08/29/2024] [Accepted: 11/11/2024] [Indexed: 11/27/2024] Open
Abstract
BACKGROUND Intermittent fasting (IF) has shown benefits in various pathological conditions. Although its anti-inflammatory potential has been recognized, its effects on the mechanism underlying rheumatoid arthritis (RA) remain insufficiently characterized. This study aimed to investigate the effects of IF in a murine model of RA. METHODS Collagen-induced arthritis (CIA) was developed in sixteen male DBA/1 mice, randomly assigned to two groups, with one undergoing IF every other day for four weeks. The effects of IF on joint inflammation and remodeling were evaluated clinically, histologically, and through tomography. Transcriptomic changes were characterized using expression microarrays, validated by RT-qPCR, and confirmed by immunohistochemistry. Additionally, modifications in gut microbiota were assessed through 16 S sequencing. RESULTS Mice subjected to IF significantly reduced the incidence and severity of clinical arthritis. Histological and radiographic assessments confirmed a decrease in inflammation and joint damage. Transcriptomic analysis revealed that IF led to the upregulation of 364 genes and the downregulation of 543 genes, with notable reductions in inflammatory signaling pathways associated with RA-related genes, including Cd72, Cd79a, Ifna, Il33, and Bglap 2. Notably, IL33 emerged as a pivotal mediator in the inflammatory processes mitigated by fasting. Key regulators associated with IF effects, such as CEBPA, FOXO1, HIF1A, PPARG, and PPARA, were identified, indicating a complex interplay between metabolic and inflammatory pathways. Furthermore, differential expression of microRNAs and lncRNAs, including miR-15b, miR-103-2, miR-302a, miR-6985, and miR- 5624, was observed. Metagenomic analysis indicated that IF enhanced the abundance and diversity of the gut microbiome, explicitly promoting anti-inflammatory bacterial populations, notably within the genus Ruminococcaceae. CONCLUSION Our findings suggest that IF exerts significant anti-inflammatory and immunoregulatory effects in the context of CIA. Given its non-risky nature, further investigation into the potential benefits of IF in patients with RA is warranted. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Rubén Cuevas-Martínez
- PABIOM Laboratory, Faculty of Medicine and Biomedical Sciences, Autonomous University of Chihuahua, Circuito Universitario Campus II, Chihuahua, CP, 31125, Mexico
| | - Susana Aideé González-Chávez
- PABIOM Laboratory, Faculty of Medicine and Biomedical Sciences, Autonomous University of Chihuahua, Circuito Universitario Campus II, Chihuahua, CP, 31125, Mexico.
| | - Mercedes Bermúdez
- Faculty of Dentistry, Autonomous University of Chihuahua, Chihuahua, Mexico
| | | | | | | | - Eduardo Chaparro-Barrera
- PABIOM Laboratory, Faculty of Medicine and Biomedical Sciences, Autonomous University of Chihuahua, Circuito Universitario Campus II, Chihuahua, CP, 31125, Mexico
| | - César Pacheco-Silva
- PABIOM Laboratory, Faculty of Medicine and Biomedical Sciences, Autonomous University of Chihuahua, Circuito Universitario Campus II, Chihuahua, CP, 31125, Mexico
| | - Consuelo Romero-Sánchez
- Cellular and Molecular Immunology Group (INMUBO), School of Dentistry, Universidad El Bosque, Bogotá, Colombia
| | | | - César Pacheco-Tena
- PABIOM Laboratory, Faculty of Medicine and Biomedical Sciences, Autonomous University of Chihuahua, Circuito Universitario Campus II, Chihuahua, CP, 31125, Mexico.
| |
Collapse
|
20
|
Picchianti Diamanti A, Panebianco C, Di Gioia V, Bellofatto IA, Salemi S, Di Rosa R, Sesti G, Nalli G, Salerno G, Finocchiaro E, Laganà B. A Case Series Report on the Effect of Tofacitinib on Joint Inflammation and Gut Microbiota Composition in Psoriatic Arthritis Patients Naive to Biologic Agents. Microorganisms 2024; 12:2387. [PMID: 39770590 PMCID: PMC11676042 DOI: 10.3390/microorganisms12122387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 11/18/2024] [Accepted: 11/19/2024] [Indexed: 01/11/2025] Open
Abstract
INTRODUCTION Psoriatic arthritis (PsA) is a complex condition within the Spondyloarthritis (SpA) group. Recent studies have focused on the important role of the intestinal microbiota in maintaining immunological homeostasis, highlighting how intestinal dysbiosis may act as a trigger for autoimmune diseases. Tofacitinib is a Janus kinase inhibitor (JAK-i) with proven efficacy for the treatment of both rheumatoid arthritis and PsA. However, there is a lack of data on its ability to reduce joint remission through ultrasonography (US) and the effects it might have on the composition of the gut microbiota. METHODS Here, we present a case series of seven bio-naïve PsA patients who received tofacitinib treatment and were followed up for 12 months. The clinical response was assessed using validated scores (DAPSA, ASDAS, and BASDAI), laboratory tests, and US assessment of the target joint and enthesis. Finally, we evaluated changes in the composition of the intestinal microbiota using next-generation sequencing analysis of fecal samples. RESULTS The patients in the study showed a significant improvement in all clinical scores used; this improvement was also confirmed by a significant reduction in the US synovitis scores. The data on the microbiota analysis suggested that the effectiveness of tofacitinib in ameliorating PsA activity was associated with a relevant modification of some gut bacterial lineages. No cases of severe adverse effects were reported. CONCLUSIONS Treatment with tofacitinib proved to be effective, safe and capable of varying the composition of the gut microbiota by selecting bacterial strains considered beneficial in immune modulation.
Collapse
Affiliation(s)
- Andrea Picchianti Diamanti
- Department of Clinical and Molecular Medicine, S. Andrea University Hospital, “Sapienza” University of Rome, 00189 Rome, Italy; (V.D.G.); (S.S.); (R.D.R.); (G.S.); (G.N.); (G.S.); (B.L.)
| | - Concetta Panebianco
- Division of Gastroenterology, Fondazione IRCCS Casa Sollievo della Sofferenza Hospital, 71013 San Giovanni Rotondo, Italy;
| | - Valeria Di Gioia
- Department of Clinical and Molecular Medicine, S. Andrea University Hospital, “Sapienza” University of Rome, 00189 Rome, Italy; (V.D.G.); (S.S.); (R.D.R.); (G.S.); (G.N.); (G.S.); (B.L.)
| | - Ilaria Anna Bellofatto
- Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132 Genoa, Italy;
| | - Simonetta Salemi
- Department of Clinical and Molecular Medicine, S. Andrea University Hospital, “Sapienza” University of Rome, 00189 Rome, Italy; (V.D.G.); (S.S.); (R.D.R.); (G.S.); (G.N.); (G.S.); (B.L.)
| | - Roberta Di Rosa
- Department of Clinical and Molecular Medicine, S. Andrea University Hospital, “Sapienza” University of Rome, 00189 Rome, Italy; (V.D.G.); (S.S.); (R.D.R.); (G.S.); (G.N.); (G.S.); (B.L.)
| | - Giorgio Sesti
- Department of Clinical and Molecular Medicine, S. Andrea University Hospital, “Sapienza” University of Rome, 00189 Rome, Italy; (V.D.G.); (S.S.); (R.D.R.); (G.S.); (G.N.); (G.S.); (B.L.)
| | - Gabriele Nalli
- Department of Clinical and Molecular Medicine, S. Andrea University Hospital, “Sapienza” University of Rome, 00189 Rome, Italy; (V.D.G.); (S.S.); (R.D.R.); (G.S.); (G.N.); (G.S.); (B.L.)
| | - Gerardo Salerno
- Department of Clinical and Molecular Medicine, S. Andrea University Hospital, “Sapienza” University of Rome, 00189 Rome, Italy; (V.D.G.); (S.S.); (R.D.R.); (G.S.); (G.N.); (G.S.); (B.L.)
| | - Etta Finocchiaro
- Dietetic and Clinical Nutrition Unit, City of Health and Science University Hospital, 10126 Turin, Italy;
| | - Bruno Laganà
- Department of Clinical and Molecular Medicine, S. Andrea University Hospital, “Sapienza” University of Rome, 00189 Rome, Italy; (V.D.G.); (S.S.); (R.D.R.); (G.S.); (G.N.); (G.S.); (B.L.)
| |
Collapse
|
21
|
Ferrari S, Galla R, Mulè S, Uberti F. Analysis of the Beneficial Effects of Probiotics on the Gut-Prostate Axis Using Prostatic Co-Culture Model. Foods 2024; 13:3647. [PMID: 39594064 PMCID: PMC11593767 DOI: 10.3390/foods13223647] [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: 09/13/2024] [Revised: 11/08/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
The link between the gut environment and the prostate has recently been proposed as a potential therapeutic approach for treating benign prostatic hyperplasia (BPH). Therefore, this study examined the advantages of a novel oral probiotic supplement to improve intestinal health and treat BPH. A 3D intestinal barrier model that simulated oral intake was used to analyse the combined regulative abilities of Bifidobacterium longum and Bifidobacterium psychaerophilum. Then, a co-culture prostatic model was used to investigate the biological consequences of the combination under conditions mimicking BPH. The results show the connection between the gut microbiome and prostate disease since the probiotics successfully modulate the primary mechanism involved in the pathogenesis of BPH. Indeed, after the intestinal passage, the mediators released from B. longum and B. psychaerophilum induced a substantial decrease in reactive oxidative species of about 6 times and inflammation (about 5 times regarding interleukine-6 and 10) and a sharp increase in testosterone and serotonin levels (about 95%). Further, proliferation and BPH principal mediators (such as androgen and dihydrotestosterone) were highly affected and nearly restored to physiological levels. Thus, BPH can be directly affected by probiotic supplementation; specifically, B. longum and B. psychaerophilum, in combination, seem able to promote the mitigation of this disease.
Collapse
Affiliation(s)
- Sara Ferrari
- Laboratory of Physiology, Department for Sustainable Development and Ecological Transition, 13100 Vercelli, Italy
| | - Rebecca Galla
- Laboratory of Physiology, Department for Sustainable Development and Ecological Transition, 13100 Vercelli, Italy
- Noivita Srls, Spin Off, University of Piemonte Orientale, Via Solaroli 17, 28100 Novara, Italy
| | - Simone Mulè
- Laboratory of Physiology, Department for Sustainable Development and Ecological Transition, 13100 Vercelli, Italy
| | - Francesca Uberti
- Laboratory of Physiology, Department for Sustainable Development and Ecological Transition, 13100 Vercelli, Italy
| |
Collapse
|
22
|
Zhu X, Long W, Zhang J, Jian C, Chen J, Huang J, Li S, Zhang J, Wang L, Chen Y, Wu J, Wang T, Zou Q, Zhu J, Zeng F. Integrated multi-omics revealed that dysregulated lipid metabolism played an important role in RA patients with metabolic diseases. Arthritis Res Ther 2024; 26:188. [PMID: 39482717 PMCID: PMC11529425 DOI: 10.1186/s13075-024-03423-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 10/22/2024] [Indexed: 11/03/2024] Open
Abstract
OBJECTIVES Patients with rheumatoid arthritis (RA) commonly experience a high prevalence of multiple metabolic diseases (MD), leading to higher morbidity and premature mortality. Here, we aimed to investigate the pathogenesis of MD in RA patients (RA_MD) through an integrated multi-omics approach. METHODS Fecal and blood samples were collected from a total of 181 subjects in this study for multi-omics analyses, including 16S rRNA and internally transcribed spacer (ITS) gene sequencing, metabolomics, transcriptomics, proteomics and phosphoproteomics. Spearman's correlation and protein-protein interaction networks were used to assess the multi-omics data correlations. The Least Absolute Shrinkage and Selection Operator (LASSO) machine learning algorithm were used to identify disease-specific biomarkers for RA_MD diagnosis. RESULTS Our results found that RA_MD was associated with differential abundance of gut microbiota such as Turicibacter and Neocosmospora, metabolites including decreased unsaturated fatty acid, genes related to linoleic acid metabolism and arachidonic acid metabolism, as well as downregulation of proteins and phosphoproteins involved in cholesterol metabolism. Furthermore, a multi-omics classifier differentiated RA_MD from RA with high accuracy (AUC: 0.958). Compared to gouty arthritis and systemic lupus erythematosus, dysregulation of lipid metabolism showed disease-specificity in RA_MD. CONCLUSIONS The integration of multi-omics data demonstrates that lipid metabolic pathways play a crucial role in RA_MD, providing the basis and direction for the prevention and early diagnosis of MD, as well as new insights to complement clinical treatment options.
Collapse
Affiliation(s)
- Xiaoting Zhu
- Department of Clinical Research Center, Dazhou Central Hospital, Dazhou, Sichuan, China
| | - Wubin Long
- Department of Rheumatology and Immunology, Sichuan Provincial People's Hospital, Chengdu, China
| | - Jing Zhang
- Department of Rheumatology and Immunology, Sichuan Provincial People's Hospital, Chengdu, China
| | - Congcong Jian
- School of Basic Medical Science, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jianghua Chen
- School of Basic Medical Science, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Jiaxin Huang
- School of Basic Medical Science, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Shilin Li
- Department of Clinical Research Center, Dazhou Central Hospital, Dazhou, Sichuan, China
| | - Jie Zhang
- Department of Clinical Research Center, Dazhou Central Hospital, Dazhou, Sichuan, China
| | - Liang Wang
- Department of Rheumatology and Immunology, Sichuan Provincial People's Hospital, Chengdu, China
| | - Yan Chen
- Department of Rheumatology and Immunology, Sichuan Provincial People's Hospital, Chengdu, China
| | - Jianhong Wu
- Department of Rheumatology and Immunology, Dazhou Central Hospital, Dazhou, China
| | - Tingting Wang
- Department of Rheumatology and Immunology, Dazhou Central Hospital, Dazhou, China
| | - Qinghua Zou
- Department of Rheumatology and Immunology, First Affiliated Hospital of Army Medical University, Chongqing, China.
| | - Jing Zhu
- Department of Rheumatology and Immunology, Sichuan Provincial People's Hospital, Chengdu, China.
| | - Fanxin Zeng
- Department of Clinical Research Center, Dazhou Central Hospital, Dazhou, Sichuan, China.
| |
Collapse
|
23
|
Cao Y, Chen J, Xiao J, Hong Y, Xu K, Zhu Y. Butyrate: a bridge between intestinal flora and rheumatoid arthritis. Front Immunol 2024; 15:1475529. [PMID: 39478858 PMCID: PMC11521846 DOI: 10.3389/fimmu.2024.1475529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 09/30/2024] [Indexed: 11/02/2024] Open
Abstract
In patients with rheumatoid arthritis (RA), intestinal flora imbalance and butyrate metabolism disorders precede clinical arthritis and are associated with the pathogenesis of RA. This imbalance can alter the immunology and intestinal permeability of the intestinal mucosa, leading to damage to the intestinal barrier. In this context, bacteria and their metabolites can enter the bloodstream and reach the distant target tissues of the host, resulting in local inflammation and aggravating arthritis. Additionally, arthritis is also exacerbated by bone destruction and immune tolerance due to disturbed differentiation of osteoclasts and adaptive immune cells. Of note, butyrate is a metabolite of intestinal flora, which not only locally inhibits intestinal immunity and targets zonulin and tight junction proteins to alleviate intestinal barrier-mediated arthritis but also inhibits osteoclasts and autoantibodies and balances the immune responses of T and B lymphocytes throughout the body to repress bone erosion and inflammation. Therefore, butyrate is a key intermediate linking intestinal flora to the host. As a result, restoring the butyrate-producing capacity of intestinal flora and using exogenous butyrate are potential therapeutic strategies for RA in the future.
Collapse
Affiliation(s)
- Yang Cao
- Second Clinical College, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Jingjing Chen
- Second Clinical College, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Jing Xiao
- Second Clinical College, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Yujie Hong
- Second Clinical College, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Ke Xu
- School of Sports Health, Shenyang Sport University, Shenyang, Liaoning, China
| | - Yan Zhu
- The Second Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, Anhui, China
| |
Collapse
|
24
|
Ievina L, Fomins N, Gudra D, Kenina V, Vilmane A, Gravelsina S, Rasa-Dzelzkaleja S, Murovska M, Fridmanis D, Nora-Krukle Z. Human Herpesvirus-6B Infection and Alterations of Gut Microbiome in Patients with Fibromyalgia: A Pilot Study. Biomolecules 2024; 14:1291. [PMID: 39456224 PMCID: PMC11506677 DOI: 10.3390/biom14101291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 10/03/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
Fibromyalgia (FM) is a chronic disorder characterized by widespread musculoskeletal pain often accompanied by fatigue, sleep disturbances, memory issues, and mood disorders. The exact cause of FM remains unknown, and diagnosis is typically based on a history of persistent widespread pain, as there are no objective biomarkers usable in diagnosis of this disorder available. The aim of this study was to identify measurable indicators specific to FM with potential as biomarkers. This study included 17 individuals diagnosed with FM and 24 apparently healthy persons. Using real-time polymerase chain reaction (qPCR), we detected the presence of human herpesvirus (HHV)-6A and B genomic sequences in DNA isolated from peripheral blood mononuclear cells (PBMCs) and buccal swabs. HHV-6-specific IgG and IgM class antibodies, along with proinflammatory cytokine levels, were measured using enzyme-linked immunosorbent assay (ELISA) and bead-based multiplex assays. Additionally, the gut microbiome was analyzed through next-generation sequencing. HHV-6B was more frequently detected in the PBMCs of FM patients. FM patients with a body mass index (BMI) of 30 or higher exhibited elevated cytokine levels compared to the control group with the same BMI range. Gut microbiome analysis revealed significant differences in both α-diversity and β-diversity between the FM and control groups, indicating a shift in species abundance in the FM group.
Collapse
Affiliation(s)
- Lauma Ievina
- The Institute of Microbiology and Virology of the Riga Stradiņš University Science Hub, LV-1067 Riga, Latvia (M.M.)
| | - Nikita Fomins
- Latvian Biomedical Research and Study Center, LV-1067 Riga, Latvia; (N.F.); (D.G.)
| | - Dita Gudra
- Latvian Biomedical Research and Study Center, LV-1067 Riga, Latvia; (N.F.); (D.G.)
| | - Viktorija Kenina
- Center for Neuroimmunology and Immune Deficiencies, LV-1050 Riga, Latvia
- Department of Biology and Microbiology, Rīga Stradinš University, LV-1007 Riga, Latvia
- Institute of Oncology and Molecular Genetics, Rīga Stradinš University, LV-1002 Riga, Latvia
- Department of Neurology, Pauls Stradiņš Clinical University Hospital, LV-1002 Riga, Latvia
| | - Anda Vilmane
- The Institute of Microbiology and Virology of the Riga Stradiņš University Science Hub, LV-1067 Riga, Latvia (M.M.)
| | - Sabine Gravelsina
- The Institute of Microbiology and Virology of the Riga Stradiņš University Science Hub, LV-1067 Riga, Latvia (M.M.)
| | - Santa Rasa-Dzelzkaleja
- The Institute of Microbiology and Virology of the Riga Stradiņš University Science Hub, LV-1067 Riga, Latvia (M.M.)
| | - Modra Murovska
- The Institute of Microbiology and Virology of the Riga Stradiņš University Science Hub, LV-1067 Riga, Latvia (M.M.)
| | - Davids Fridmanis
- Latvian Biomedical Research and Study Center, LV-1067 Riga, Latvia; (N.F.); (D.G.)
| | - Zaiga Nora-Krukle
- The Institute of Microbiology and Virology of the Riga Stradiņš University Science Hub, LV-1067 Riga, Latvia (M.M.)
| |
Collapse
|
25
|
Wurm J, Curtis N, Zimmermann P. The effect of antibiotics on the intestinal microbiota in children - a systematic review. FRONTIERS IN ALLERGY 2024; 5:1458688. [PMID: 39435363 PMCID: PMC11491438 DOI: 10.3389/falgy.2024.1458688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 09/06/2024] [Indexed: 10/23/2024] Open
Abstract
Background Children are the age group with the highest exposure to antibiotics (ABX). ABX treatment changes the composition of the intestinal microbiota. The first few years of life are crucial for the establishment of a healthy microbiota and consequently, disturbance of the microbiota during this critical period may have far-reaching consequences. In this review, we summarise studies that have investigated the effect of ABX on the composition of the intestinal microbiota in children. Methods According to the PRISMA guidelines, a systematic search was done using MEDLINE and Embase to identify original studies that have investigated the effect of systemic ABX on the composition of the intestinal microbiota in children. Results We identified 89 studies investigating a total of 9,712 children (including 4,574 controls) and 14,845 samples. All ABX investigated resulted in a reduction in alpha diversity, either when comparing samples before and after ABX or children with ABX and controls. Following treatment with penicillins, the decrease in alpha diversity persisted for up to 6-12 months and with macrolides, up to the latest follow-up at 12-24 months. After ABX in the neonatal period, a decrease in alpha diversity was still found at 36 months. Treatment with penicillins, penicillins plus gentamicin, cephalosporins, carbapenems, macrolides, and aminoglycosides, but not trimethoprim/sulfamethoxazole, was associated with decreased abundances of beneficial bacteria including Actinobacteria, Bifidobacteriales, Bifidobacteriaceae, and/or Bifidobacterium, and Lactobacillus. The direction of change in the abundance of Enterobacteriaceae varied with ABX classes, but an increase in Enterobacteriaceae other than Escherichia coli was frequently observed. Conclusion ABX have profound effects on the intestinal microbiota of children, with notable differences between ABX classes. Macrolides have the most substantial impact while trimethoprim/sulfamethoxazole has the least pronounced effect.
Collapse
Affiliation(s)
- Juliane Wurm
- Department of Paediatrics, Fribourg Hospital, Fribourg, Switzerland
- Department of Health Science and Medicine, University Lucerne, Lucerne, Switzerland
| | - Nigel Curtis
- Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia
- Infectious Diseases Research Group, Murdoch Children’s Research Institute, Parkville, VIC, Australia
- Infectious Diseases Unit, The Royal Children’s Hospital Melbourne, Parkville, VIC, Australia
| | - Petra Zimmermann
- Department of Paediatrics, Fribourg Hospital, Fribourg, Switzerland
- Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia
- Infectious Diseases Research Group, Murdoch Children’s Research Institute, Parkville, VIC, Australia
- Department for Community Health, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
26
|
Su QY, Zhang Y, Qiao D, Song X, Shi Y, Wang Z, Wang CY, Zhang SX. Gut microbiota dysbiosis in ankylosing spondylitis: a systematic review and meta-analysis. Front Cell Infect Microbiol 2024; 14:1376525. [PMID: 39421642 PMCID: PMC11484232 DOI: 10.3389/fcimb.2024.1376525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 08/29/2024] [Indexed: 10/19/2024] Open
Abstract
Background Ankylosing spondylitis (AS) is a connective tissue disease that primarily affects spinal joints, peripheral joints, and paravertebral soft tissues, leading to joint stiffness and spinal deformity. Growing evidence has implicated gut microbiota in the regulation of AS, though the underlying mechanisms remain poorly understood. Methods We conducted a comprehensive search of PubMed, Embase, Web of Science, the Cochrane Library, MEDLINE, Wanfang Data, China Science and Technology Journal Database (VIP), and China National Knowledge Internet (CNKI) databases from the time the databases were created until 30 July 2023. To evaluate changes in α-diversity and the abundance of certain microbiota families in AS, standardized mean difference (SMD) and 95% confidence interval (CI) calculations were made. Meta-analyses were performed using STATA 12.0 and the quality of the literature was assessed by following systematic review guidelines. Results This systematic review and meta-analysis included 47 studies, providing insights into the gut microbiota composition in patients with AS compared to healthy controls (HCs). Our findings indicate a significant reduction in gut microbial diversity in patients with AS, as evidenced by a decrease in both richness and evenness. Specifically, the Shannon index showed a moderate decrease (SMD = -0.27, 95% CI: -0.49, -0.04; P < 0.001), suggesting a less diverse microbial ecosystem in patients with AS. The Chao1 index further confirmed this trend, with a larger effect size (SMD = -0.44, 95% CI: -0.80, -0.07; P < 0.001), indicating a lower species richness. The Simpson index also reflected a significant reduction in evenness (SMD = -0.30, 95% CI: -0.53, -0.06; P < 0.001). Additionally, patients with AS who received anti-rheumatic combination treatment exhibited a more pronounced reduction in α-diversity compared to untreated patients, highlighting the potential impact of this treatment on gut microbiota balance. In terms of specific microbial families, we observed a significant decrease in the abundance of Bifidobacterium (SMD = -0.42, 95% CI: -2.37, 1.52; P < 0.001), which is known for its beneficial effects on gut health. Conversely, an increase in the abundance of Bacteroidetes was noted (SMD = 0.42, 95% CI: -0.93, 1.76; P < 0.001), suggesting a possible shift in the gut microbiota composition that may be associated with AS pathophysiology. Conclusion Our analysis revealed changes in α-diversity and the relative abundance of specific bacteria in AS. This suggests that targeting the gut microbiota could provide new therapeutic opportunities for treating AS. Systematic review registration https://www.crd.york.ac.uk./PROSPERO/, identifier CRD42023450028.
Collapse
Affiliation(s)
- Qin-Yi Su
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, The Shanxi Medical University, Taiyuan, Shanxi, China
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, China
| | - Yan Zhang
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, The Shanxi Medical University, Taiyuan, Shanxi, China
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, China
| | - Dan Qiao
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, The Shanxi Medical University, Taiyuan, Shanxi, China
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, China
| | - Xia Song
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, The Shanxi Medical University, Taiyuan, Shanxi, China
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, China
| | - Yang Shi
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, The Shanxi Medical University, Taiyuan, Shanxi, China
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, China
| | - Zhe Wang
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, The Shanxi Medical University, Taiyuan, Shanxi, China
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, China
| | - Chen-Yan Wang
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, The Shanxi Medical University, Taiyuan, Shanxi, China
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, China
| | - Sheng-Xiao Zhang
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, The Shanxi Medical University, Taiyuan, Shanxi, China
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, China
- Shanxi Medical University (SXMU)-Tsinghua Collaborative Innovation Center for Frontier Medicine, Shanxi Medical University, Taiyuan, China
| |
Collapse
|
27
|
Luo X, Li J, Yang M, Tu L, Xie Y, Lv Q, Wen S, Wen X, Zhou L, Gu J. Alterations in peripheral T-cell and B-cell subsets in the ankylosing spondylitis patients with gut inflammation. Int J Rheum Dis 2024; 27:e15324. [PMID: 39380421 DOI: 10.1111/1756-185x.15324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 08/08/2024] [Accepted: 08/15/2024] [Indexed: 10/10/2024]
Abstract
AIM This study investigates changes in immune cell subsets in peripheral blood of ankylosing spondylitis (AS) patients with colitis or terminal ileitis. It aims to explore the connection between changes in lymphocyte subsets and gut inflammation, providing insights for early detection. METHODS Overall, 50 AS patients undergoing colonoscopy were enrolled. Flow cytometry was employed to analyze lymphocyte subsets, including T and B cells, in peripheral blood. Disease activity was assessed using CRP, ESR, BASDAI, ASDAS-CRP, and ASDAS-ESR. RESULTS Compared to AS patients without gut inflammation, those with colorectal inflammation showed a significant increase in total T cells (p < .05), an increase in exhausted CD4+ T cells (p < .05), and a decrease in Th2 cells and total Tc cells (p < .05). Notably, in AS patients with terminal ileitis, there was an increase in total B cells and classic switched B cells (p < .05), with a decrease in double-positive T cells (p < .05). However, no significant differences were observed in the distribution of Tfh-cell subpopulations (Tfh1, Tfh2, Tfh17) and Tc-cell subpopulations (Tc1, Tc2, Tc17) between AS patients with either colorectal inflammation or terminal ileitis (p > .05). We explored the relationship between disease activity scores, ESR, CRP, and lymphocyte subsets, but found no statistically significant correlation between them. CONCLUSION Distinct immune patterns may exist in AS with different types of intestinal inflammation. Colitis in AS is primarily characterized by a significant increase in exhausted CD4+ T cells, along with a decrease in Th2 cells. In contrast, terminal ileum inflammation in AS is marked by an increase in total B cells and classic switched B cells. These findings offer new insights for early detection and therapeutic intervention.
Collapse
Affiliation(s)
- Xiqing Luo
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jinwei Li
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Mingcan Yang
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Liudan Tu
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Ya Xie
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Qing Lv
- Department of Rheumatology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Shenghui Wen
- Department of Rheumatology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Xianghui Wen
- Shenzhen Institute of Immunomedicine Transformation (Longhua), Shenzhen, China
| | - Liuzhong Zhou
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jieruo Gu
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
28
|
Dabboussi N, Debs E, Bouji M, Rafei R, Fares N. Balancing the mind: Toward a complete picture of the interplay between gut microbiota, inflammation and major depressive disorder. Brain Res Bull 2024; 216:111056. [PMID: 39182696 DOI: 10.1016/j.brainresbull.2024.111056] [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: 06/03/2024] [Revised: 08/07/2024] [Accepted: 08/22/2024] [Indexed: 08/27/2024]
Abstract
The intricate interplay existing between gut microbiota and homeostasis extends to the realm of the brain, where emerging research underscores the significant impact of the microbiota on mood regulation and overall neurological well-being and vice-versa, with inflammation playing a pivotal role in mediating these complex interactions. This comprehensive review explores the complex interplay between inflammation, alterations in gut microbiota, and their impact on major depressive disorder (MDD). It provides a cohesive framework for the puzzle pieces of this triad, emphasizing recent advancements in understanding the gut microbiota and inflammatory states' contribution to the depressive features. Two directions of communication between the gut and the brain in depression are discussed, with inflammation serving as a potential modulator. Therapeutic implications were discussed as well, drawing insights from interventional studies on the effects of probiotics on gut bacterial composition and depressive symptoms. Ultimately, this review will attempt to provide a complete and valuable framework for future research and therapeutic interventions in MDD.
Collapse
Affiliation(s)
- Nour Dabboussi
- Laboratory of Research in Physiology and pathophysiology, Faculty of Medicine, Saint Joseph University of Beirut, POBox. 17-5208 - Mar Mikhaël, Beirut 1104 2020, Lebanon; Laboratoire Microbiologie Santé et Environnement (LMSE), Doctoral School of Science & Technology, Faculty of Public Health, Lebanese University, Tripoli, Lebanon.
| | - Espérance Debs
- Department of Biology, Faculty of Arts and Sciences, University of Balamand, P.O. Box 100, Tripoli, Lebanon.
| | - Marc Bouji
- Faculté des Sciences, Université Saint-Joseph de Beyrouth-Liban, Mar Roukos, Beirut, Lebanon.
| | - Rayane Rafei
- Laboratoire Microbiologie Santé et Environnement (LMSE), Doctoral School of Science & Technology, Faculty of Public Health, Lebanese University, Tripoli, Lebanon
| | - Nassim Fares
- Laboratory of Research in Physiology and pathophysiology, Faculty of Medicine, Saint Joseph University of Beirut, POBox. 17-5208 - Mar Mikhaël, Beirut 1104 2020, Lebanon.
| |
Collapse
|
29
|
Bakinowska E, Stańska W, Kiełbowski K, Szwedkowicz A, Boboryko D, Pawlik A. Gut Dysbiosis and Dietary Interventions in Rheumatoid Arthritis-A Narrative Review. Nutrients 2024; 16:3215. [PMID: 39339815 PMCID: PMC11435214 DOI: 10.3390/nu16183215] [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: 08/11/2024] [Revised: 09/20/2024] [Accepted: 09/21/2024] [Indexed: 09/30/2024] Open
Abstract
Rheumatoid arthritis (RA) is a chronic and progressive autoimmune disease. The pathogenesis of RA is complex and involves interactions between articular cells, such as fibroblast-like synoviocytes, and immune cells. These cells secrete pro-inflammatory cytokines, chemokines, metalloproteinases and other molecules that together participate in joint degradation. The current evidence suggests the important immunoregulatory role of the gut microbiome, which can affect susceptibility to diseases and infections. An altered microbiome, a phenomenon known as gut dysbiosis, is associated with the development of inflammatory diseases. Importantly, the profile of the gut microbiome depends on dietary habits. Therefore, dietary elements and interventions can indirectly impact the progression of diseases. This review summarises the evidence on the involvement of gut dysbiosis and diet in the pathogenesis of RA.
Collapse
Affiliation(s)
- Estera Bakinowska
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Wiktoria Stańska
- Department of Medical Biology, Medical University of Warsaw, 00-575 Warsaw, Poland
| | - Kajetan Kiełbowski
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Agata Szwedkowicz
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Dominika Boboryko
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| |
Collapse
|
30
|
Coccia C, Bonomi F, Lo Cricchio A, Russo E, Peretti S, Bandini G, Lepri G, Bartoli F, Moggi-Pignone A, Guiducci S, Del Galdo F, Furst DE, Matucci Cerinic M, Bellando-Randone S. The Potential Role of Butyrate in the Pathogenesis and Treatment of Autoimmune Rheumatic Diseases. Biomedicines 2024; 12:1760. [PMID: 39200224 PMCID: PMC11351188 DOI: 10.3390/biomedicines12081760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 07/30/2024] [Accepted: 07/31/2024] [Indexed: 09/02/2024] Open
Abstract
The gut microbiota is a complex ecosystem of microorganisms residing in the human gastrointestinal tract, playing a crucial role in various biological processes and overall health maintenance. Dysbiosis, an imbalance in the composition and function of the gut microbiota, is linked to systemic autoimmune diseases (SAD). Short-chain fatty acids (SCFAs), especially butyrate, produced by the gut microbiota through the fermentation of dietary fibers, play a significant role in immunomodulation and maintaining intestinal homeostasis. Butyrate is essential for colonocyte energy, anti-inflammatory responses, and maintaining intestinal barrier integrity. Studies show reduced butyrate-producing bacteria in SAD patients, suggesting that increasing butyrate levels could have therapeutic benefits. Butyrate's anti-inflammatory effects and its potential therapeutic role have been studied in rheumatoid arthritis, Sjogren's syndrome, systemic lupus erythematosus, systemic sclerosis, and Behçet's disease. Despite promising in vitro and animal model results, human studies are limited, and the optimal strategies for modulating dysbiosis in SADs remain elusive. This review explores the current evidence on the immunoregulatory role of butyrate and its potential therapeutic effects in SAD.
Collapse
Affiliation(s)
- Carmela Coccia
- Department of Experimental and Clinical Medicine, Division of Rheumatology, Scleroderma Unit, AOU Careggi, University of Florence, 50139 Florence, Italy; (C.C.); (F.B.); (S.P.); (G.L.); (F.B.); (S.G.)
| | - Francesco Bonomi
- Department of Experimental and Clinical Medicine, Division of Rheumatology, Scleroderma Unit, AOU Careggi, University of Florence, 50139 Florence, Italy; (C.C.); (F.B.); (S.P.); (G.L.); (F.B.); (S.G.)
| | - Anna Lo Cricchio
- Department of Experimental and Clinical Medicine, Division of Internal Medicine AOUC, University of Florence, 50134 Florence, Italy; (A.L.C.); (G.B.); (A.M.-P.)
| | - Edda Russo
- Department of Experimental and Clinical Medicine, University of Florence, 50139 Florence, Italy;
| | - Silvia Peretti
- Department of Experimental and Clinical Medicine, Division of Rheumatology, Scleroderma Unit, AOU Careggi, University of Florence, 50139 Florence, Italy; (C.C.); (F.B.); (S.P.); (G.L.); (F.B.); (S.G.)
| | - Giulia Bandini
- Department of Experimental and Clinical Medicine, Division of Internal Medicine AOUC, University of Florence, 50134 Florence, Italy; (A.L.C.); (G.B.); (A.M.-P.)
| | - Gemma Lepri
- Department of Experimental and Clinical Medicine, Division of Rheumatology, Scleroderma Unit, AOU Careggi, University of Florence, 50139 Florence, Italy; (C.C.); (F.B.); (S.P.); (G.L.); (F.B.); (S.G.)
| | - Francesca Bartoli
- Department of Experimental and Clinical Medicine, Division of Rheumatology, Scleroderma Unit, AOU Careggi, University of Florence, 50139 Florence, Italy; (C.C.); (F.B.); (S.P.); (G.L.); (F.B.); (S.G.)
| | - Alberto Moggi-Pignone
- Department of Experimental and Clinical Medicine, Division of Internal Medicine AOUC, University of Florence, 50134 Florence, Italy; (A.L.C.); (G.B.); (A.M.-P.)
| | - Serena Guiducci
- Department of Experimental and Clinical Medicine, Division of Rheumatology, Scleroderma Unit, AOU Careggi, University of Florence, 50139 Florence, Italy; (C.C.); (F.B.); (S.P.); (G.L.); (F.B.); (S.G.)
| | - Francesco Del Galdo
- Raynaud’s and Scleroderma Programme, NIHR Biomedical Research Centre, Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds LS9 7JT, UK;
| | - Daniel E. Furst
- Department of Rheumatology, University of California Los Angeles, Los Angeles, CA 90095, USA;
| | - Marco Matucci Cerinic
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS San Raffaele Hospital, 20132 Milan, Italy;
| | - Silvia Bellando-Randone
- Department of Experimental and Clinical Medicine, Division of Rheumatology, Scleroderma Unit, AOU Careggi, University of Florence, 50139 Florence, Italy; (C.C.); (F.B.); (S.P.); (G.L.); (F.B.); (S.G.)
| |
Collapse
|
31
|
Garcia AC, Six N, Ma L, Morel L. Intersection of the microbiome and immune metabolism in lupus. Immunol Rev 2024; 325:77-89. [PMID: 38873851 PMCID: PMC11338729 DOI: 10.1111/imr.13360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Abstract
Systemic lupus erythematosus is a complex autoimmune disease resulting from a dysregulation of the immune system that involves gut dysbiosis and an altered host cellular metabolism. This review highlights novel insights and expands on the interactions between the gut microbiome and the host immune metabolism in lupus. Pathobionts, invasive pathogens, and even commensal microbes, when in dysbiosis, can all trigger and modulate immune responses through metabolic reprogramming. Changes in the microbiota's global composition or individual taxa may trigger a cascade of metabolic changes in immune cells that may, in turn, reprogram their functions. Factors contributing to dysbiosis include changes in intestinal hypoxia, competition for glucose, and limited availability of essential nutrients, such as tryptophan and metal ions, all of which can be driven by host metabolism changes. Conversely, the accumulation of some host metabolites, such as itaconate, succinate, and free fatty acids, could further influence the microbial composition and immune responses. Overall, mounting evidence supports a bidirectional relationship between host immunometabolism and the microbiota in lupus pathogenesis.
Collapse
Affiliation(s)
- Abigail Castellanos Garcia
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - Natalie Six
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - Longhuan Ma
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - Laurence Morel
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health San Antonio, San Antonio, Texas, USA
| |
Collapse
|
32
|
Fan S, Xue B, Ma J. Causal relationship between gut microbiota and idiopathic pulmonary fibrosis: A two-sample Mendelian randomization. Medicine (Baltimore) 2024; 103:e39013. [PMID: 39029004 PMCID: PMC11398756 DOI: 10.1097/md.0000000000039013] [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: 01/09/2024] [Accepted: 06/19/2024] [Indexed: 07/21/2024] Open
Abstract
To explore the causal relationship between gut microbiota (GM) and Idiopathic pulmonary fibrosis (IPF), we performed a two-sample Mendelian randomization (MR). GM was used as an exposure factor, and instrumental variables were determined from the GWAS of 18,340 participants. GWAS of IPF (including 1028 IPF patients and 196,986 controls) from the FinnGen was used as the outcome factor. The primary analysis method is the inverse variance weighted (IVW) method, and sensitivity analysis was used to validate the reliability. Family Bacteroidaceae (OR = 1.917 95% CI = 1.083-3.393, P = .026), order Gastranaerophilales (OR = 1.441 95% CI = 1.019-2.036, P = .039), genus Senegalimassilia (OR = 2.28 95% CI = 1.174-4.427, P = .015), phylum Cyanobacteria (OR = 1.631 95% CI = 1.035-2.571, P = .035) were positively correlated with IPF. FamilyXIII(OR = 0.452 95% CI = 0.249-0.82, P = .009), order Selenomonadale (OR = 0.563 95% CI = 0.337-0.941, P = .029), genus Veillonella (OR = 0.546 95% CI = 0.304-0.982, P = .043) (OR = 0.717 95% CI = 0.527-0.976, P = .034), genus Ruminococcusgnavus (OR = 0.717 95% CI = 0.527-0.976, P = .034), genus Oscillibacter (OR = 0.571 95% CI = 0.405-0.806, P = .001) was negatively correlated with IPF. Sensitivity analysis showed no evidence of pleiotropy or heterogeneity (P > .05). The results of MR demonstrated a causal relationship between GM and IPF. Further studies are needed to investigate the intrinsic mechanisms of the GM in the pathogenesis of IPF.
Collapse
Affiliation(s)
- Shiqin Fan
- Department of Intensive Care Medicine, Liyuan Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Baorui Xue
- Department of Endocrinology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jing Ma
- Department of Intensive Care Medicine, Liyuan Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
33
|
Pan P, Wang Y, Nyirenda MH, Saiyed Z, Karimian Azari E, Sunderman A, Milling S, Harnett MM, Pineda M. Undenatured type II collagen protects against collagen-induced arthritis by restoring gut-joint homeostasis and immunity. Commun Biol 2024; 7:804. [PMID: 38961129 PMCID: PMC11222443 DOI: 10.1038/s42003-024-06476-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 06/20/2024] [Indexed: 07/05/2024] Open
Abstract
Oral administration of harmless antigens can induce suppression of reactive immune responses, a process that capitalises on the ability of the gastrointestinal tract to tolerate exposure to food and commensal microbiome without triggering inflammatory responses. Repeating exposure to type II collagen induces oral tolerance and inhibits induction of arthritis, a chronic inflammatory joint condition. Although some mechanisms underlying oral tolerance are described, how dysregulation of gut immune networks impacts on inflammation of distant tissues like the joints is unclear. We used undenatured type II collagen in a prophylactic regime -7.33 mg/kg three times/week- to describe the mechanisms associated with protective oral immune-therapy (OIT) in gut and joint during experimental Collagen-Induced Arthritis (CIA). OIT reduced disease incidence to 50%, with reduced expression of IL-17 and IL-22 in the joints of asymptomatic mice. Moreover, whilst the gut tissue of arthritic mice shows substantial damage and activation of tissue-specific immune networks, oral administration of undenatured type II collagen protects against gut pathology in all mice, symptomatic and asymptomatic, rewiring IL-17/IL-22 networks. Furthermore, gut fucosylation and microbiome composition were also modulated. These results corroborate the relevance of the gut-joint axis in arthritis, showing novel regulatory mechanisms linked to therapeutic OIT in joint disease.
Collapse
Affiliation(s)
- Piaopiao Pan
- Centre for the Cellular Microenvironment, School of Molecular Biology, University of Glasgow, Glasgow, UK
| | - Yilin Wang
- Department of Bacteriology and Immunology, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis & Thoracic Tumor Research Institute, Beijing, China
| | - Mukanthu H Nyirenda
- Institute of Infection and Immunity, University of Glasgow, Glasgow, UK
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Zainulabedin Saiyed
- Research and Development, Lonza Greenwood LLC, North Emerald Road, Greenwood, SC, USA
| | - Elnaz Karimian Azari
- Research and Development, Lonza Greenwood LLC, North Emerald Road, Greenwood, SC, USA
| | - Amy Sunderman
- Research and Development, Lonza Greenwood LLC, North Emerald Road, Greenwood, SC, USA
| | - Simon Milling
- Institute of Infection and Immunity, University of Glasgow, Glasgow, UK
| | | | - Miguel Pineda
- Centre for the Cellular Microenvironment, School of Molecular Biology, University of Glasgow, Glasgow, UK.
| |
Collapse
|
34
|
Hansdah K, Lui JC. Emerging Insights into the Endocrine Regulation of Bone Homeostasis by Gut Microbiome. J Endocr Soc 2024; 8:bvae117. [PMID: 38957653 PMCID: PMC11215793 DOI: 10.1210/jendso/bvae117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Indexed: 07/04/2024] Open
Abstract
Gut microbiota plays an important role in the regulation of bone homeostasis and bone health. Recent studies showed that these effects could be mediated through microbial metabolites released by the microbiota like short-chain fatty acids, metabolism of endogenous molecules such as bile acids, or a complex interplay between microbiota, the endocrine system, and the immune system. Importantly, some studies showed a reciprocal relationship between the endocrine system and gut microbiota. For instance, postmenopausal estrogen deficiency could lead to dysbiosis of the gut microbiota, which could in turn affect various immune response and bone remodeling. In addition, evidence showed that shift in the indigenous gut microbiota caused by antibiotics treatment may also impact normal skeletal growth and maturation. In this mini-review, we describe recent findings on the role of microbiome in bone homeostasis, with a particular focus on molecular mechanisms and their interactions with the endocrine and immune system. We will also discuss the recent findings on estrogen deficiency and microbiota dysbiosis, and the clinical implications for the development of new therapeutic strategies for osteoporosis and other bone disorders.
Collapse
Affiliation(s)
- Kirtal Hansdah
- Section on Growth and Development, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Julian C Lui
- Section on Growth and Development, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
35
|
Bixio R, Bertelle D, Bertoldo E, Morciano A, Rossini M. The potential pathogenic role of gut microbiota in rheumatic diseases: a human-centred narrative review. Intern Emerg Med 2024; 19:891-900. [PMID: 38141117 DOI: 10.1007/s11739-023-03496-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 11/23/2023] [Indexed: 12/24/2023]
Abstract
A growing amount of evidence suggests that gut microbiota plays an important role in human health, including a possible role in the pathogenesis of rheumatic and musculoskeletal diseases (RMD). We analysed the current evidence about the role of microbiota in rheumatoid arthritis (RA), spondyloarthritis (SpA), systemic lupus erythematosus (SLE) and systemic sclerosis (SSc). In RA, we found a general consensus regarding a reduction of diversity and a specific bacterial signature, with consistent changes according to the different ethnic and geographical areas. The major pathogenetic role in RA is recognised for P. copri, L. salivarius and Collinsella, even if findings become more heterogeneous when considering established disease. In SpA, we found a relative gut abundance of Akkermansia, Coprococcus, Ruminoccocus and a relative reduction in Bacterioides and Firmicutes spp. Human and preclinical data suggest loss of mucosal barrier, increased permeability and Th1- and Th17-mediated inflammation. Additionally, HLA-B27 seems to play a role in shaping the intestinal microbiota and the consequent inflammation. In SLE, the typical gut microbiota signature was characterised by a reduction in the Firmicutes/Bacteroidetes ratio and by enrichment of Rhodococcus, Eggerthella, Klebsiella, Prevotella, Eubacterium and Flavonifractor, even if their real pathogenic impact remains unclear. In SSc, gastrointestinal dysbiosis is well documented with an increase of pro-inflammatory species (Fusobacterium, Prevotella, Ruminococcus, Akkermansia, γ-Proteobacteria, Erwinia, Trabsulsiella, Bifidobacterium, Lactobacillus, Firmicutes and Actinobacteria) and a reduction of species as Faecalibacterium, Clostridium, Bacteroidetes and Rikenella. In conclusion, seems possible to recognise a distinct gut microbiota profile for each RMD, even if significant differences in bacterial species do exist between different studies and there is a high risk of bias due to the cross-sectional nature of such studies. Therefore longitudinal studies are needed, especially on patients with preclinical and early disease, to investigate the real role of gut microbiota in the pathogenesis of RMD.
Collapse
Affiliation(s)
- Riccardo Bixio
- Rheumatology Section, Department of Medicine, University of Verona, P.Le L.A. Scuro 10, 37134, Verona, Italy.
| | - Davide Bertelle
- Rheumatology Section, Department of Medicine, University of Verona, P.Le L.A. Scuro 10, 37134, Verona, Italy
- Rheumatology Section, Department of Medicine, Azienda Ospedaliera Friuli Occidentale, Pordenone, Italy
| | - Eugenia Bertoldo
- Rheumatology Section, Department of Medicine, University of Verona, P.Le L.A. Scuro 10, 37134, Verona, Italy
- Internal Medicine Unit, Department of Medicine, Mater Salutis Hospital, Legnago, Italy
| | - Andrea Morciano
- Rheumatology Section, Department of Medicine, University of Verona, P.Le L.A. Scuro 10, 37134, Verona, Italy
| | - Maurizio Rossini
- Rheumatology Section, Department of Medicine, University of Verona, P.Le L.A. Scuro 10, 37134, Verona, Italy
| |
Collapse
|
36
|
Longo UG, Lalli A, Bandini B, Angeletti S, Lustig S, Budhiparama NC. The influence of gut microbiome on periprosthetic joint infections: State-of-the art. J ISAKOS 2024; 9:353-361. [PMID: 38272392 DOI: 10.1016/j.jisako.2024.01.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 01/16/2024] [Accepted: 01/18/2024] [Indexed: 01/27/2024]
Abstract
Early periprosthetic joint infection constitutes one of the most frightening complications of joint replacement. Recently, some evidence has highlighted the potential link between dysregulation of the gut microbiota and degenerative diseases of joints. It has been hypothesized that microbiome dysbiosis may increase the risk of periprosthetic joint infection by facilitating bacterial translocation from these sites to the bloodstream or by impairing local or systemic immune responses. Although the processes tying the gut microbiome to infection susceptibility are still unknown, new research suggests that the presurgical gut microbiota-a previously unconsidered component-may influence the patient's ability to resist infection. Exploring the potential impact of the microbiome on periprosthetic joint infections may therefore bring new insights into the pathogenesis and therapy of these disorders. For a successful therapy, a proper surgical procedure in conjunction with an antibacterial concept is essential. As per the surgical approach, different treatment strategies include surgical irrigation, debridement, antibiotic therapy, and implant retention with or without polyethylene exchange. Other alternatives could be one-stage or two-stage revisions surgery. Interventions that either directly target gut microbes as well as interventions that modify the composition and/or function of the commensal microbes represent an innovative and potentially successful field to be explored. In recent times, innovative therapeutic methods have arisen in the realm of microbiome restoration and the management of gut-related ailments. These progressive approaches offer fresh perspectives on tackling intricate microbial imbalances in the gastrointestinal tract. These emerging therapies signify a shift towards more precise and individualized approaches to microbiome restoration and the management of gut-related disorders. Once a more advanced knowledge of the pathways linking the gut microbiota to musculoskeletal tissues is gained, relevant microbiome-based therapies can be developed. If dysbiosis is proven to be a significant contributor, developing treatments for dysbiosis may represent a new frontier in the prevention of periprosthetic joint infections.
Collapse
Affiliation(s)
- Umile Giuseppe Longo
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy; Research Unit of Orthopaedic and Trauma Surgery, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Alberto Lalli
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy; Research Unit of Orthopaedic and Trauma Surgery, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy.
| | - Benedetta Bandini
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy; Research Unit of Orthopaedic and Trauma Surgery, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy.
| | - Silvia Angeletti
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy; Research Unit of Orthopaedic and Trauma Surgery, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy.
| | - Sebastien Lustig
- Orthopaedic Department, Lyon North University Hospital, Hôpital de La Croix Rousse, Hospices Civils de Lyon, 103 Grande Rue de la Croix Rousse, 69004 Lyon, France.
| | - Nicolaas Cyrillus Budhiparama
- Department of Orthopaedic and Traumatology, Faculty of Medicine, Unversitas Airlangga, Jl. Mayjend. Prof. Dr. Moestopo 6-8, Surabaya 60286, Indonesia; Department of Orthopaedics, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, the Netherlands.
| |
Collapse
|
37
|
Tang C, Li L, Jin X, Wang J, Zou D, Hou Y, Yu X, Wang Z, Jiang H. Investigating the Impact of Gut Microbiota on Gout Through Mendelian Randomization. Orthop Res Rev 2024; 16:125-136. [PMID: 38766545 PMCID: PMC11100514 DOI: 10.2147/orr.s454211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 05/07/2024] [Indexed: 05/22/2024] Open
Abstract
Background The relationship between gout and gut microbiota has attracted significant attention in current research. However, due to the diverse range of gut microbiota, the specific causal effect on gout remains unclear. This study utilizes Mendelian randomization (MR) to investigate the causal relationship between gut microbiota and gout, aiming to elucidate the underlying mechanism of microbiome-mediated gout and provide valuable guidance for clinical prevention and treatment. Materials and Methods The largest genome-wide association study meta-analysis conducted by the MiBioGen Consortium (n=18,340) was utilized to perform a two-sample Mendelian randomization investigation on aggregate statistics of intestinal microbiota. Summary statistics for gout were utilized from the data released by EBI. Various methods, including inverse variance weighted, weighted median, weighted model, MR-Egger, and Simple-mode, were employed to assess the causal relationship between gut microbiota and gout. Reverse Mendelian randomization analysis revealed a causal association between bacteria and gout in forward Mendelian randomization analysis. Cochran's Q statistic was used to quantify instrumental variable heterogeneity. Results The inverse variance weighted estimation revealed that Rikenellaceae exhibited a slight protective effect on gout, while the presence of Ruminococcaceae UCG_011 is associated with a marginal increase in the risk of gout. According to the reverse Mendelian Randomization results, no significant causal relationship between gout and gut microbiota was observed. No significant heterogeneity of instrumental variables or level pleiotropy was detected. Conclusion Our MR analysis revealed a potential causal relationship between the development of gout and specific gut microbiota; however, the causal effect was not robust, and further research is warranted to elucidate its underlying mechanism in gout development. Considering the significant association between diet, gut microbiota, and gout, these findings undoubtedly shed light on the mechanisms of microbiota-mediated gout and provide new insights for translational research on managing and standardizing treatment for this condition.
Collapse
Affiliation(s)
- Chaoqun Tang
- The First Clinical Medical School, Anhui University of Chinese Medicine, Hefei, Anhui, People’s Republic of China
| | - Lei Li
- Department of Orthopedics, Shandong Wendeng Osteopathic Hospital, Wendeng, Weihai, Shandong, People’s Republic of China
| | - Xin Jin
- Department of Orthopedics, Shandong Wendeng Osteopathic Hospital, Wendeng, Weihai, Shandong, People’s Republic of China
| | - Jinfeng Wang
- Department of Orthopedics, Shandong Wendeng Osteopathic Hospital, Wendeng, Weihai, Shandong, People’s Republic of China
| | - Debao Zou
- Department of Orthopedics, Shandong Wendeng Osteopathic Hospital, Wendeng, Weihai, Shandong, People’s Republic of China
| | - Yan Hou
- Department of Orthopedics, Shandong Wendeng Osteopathic Hospital, Wendeng, Weihai, Shandong, People’s Republic of China
| | - Xin Yu
- Department of Orthopedics, Shandong Wendeng Osteopathic Hospital, Wendeng, Weihai, Shandong, People’s Republic of China
| | - Zhizhou Wang
- Department of Orthopedics, Shandong Wendeng Osteopathic Hospital, Wendeng, Weihai, Shandong, People’s Republic of China
| | - Hongjiang Jiang
- The First Clinical Medical School, Anhui University of Chinese Medicine, Hefei, Anhui, People’s Republic of China
- Department of Orthopedics, Shandong Wendeng Osteopathic Hospital, Wendeng, Weihai, Shandong, People’s Republic of China
| |
Collapse
|
38
|
Romano L, Napolitano L, Crocetto F, Sciorio C, Sio MD, Miranda A, Romano M, Priadko K. Prostate and gut: Any relationship? A narrative review on the available evidence and putative mechanisms. Prostate 2024; 84:513-524. [PMID: 38353479 DOI: 10.1002/pros.24675] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 12/26/2023] [Accepted: 01/30/2024] [Indexed: 03/22/2024]
Abstract
BACKGROUND Gut microbiome is a community of microorganisms that lives in the human intestine and exerts various functions on the host, including metabolic, immunoregulatory, and control over cell proliferation. Gut microbiome alterations have been associated with various pathological conditions, such as diabetes mellitus, obesity, and cardiovascular diseases. Gut-prostate axis is explained by the association between gut microbiome quantitative and functional alterations along with increased intestinal epithelial permeability with prostatediseases. However, the pathophysiological mechanisms and clinical importance of this association are not completely clarified yet. METHODS We conducted a narrative review of the most relevant articles in the Medline (US National Library of Medicine, Bethesda, MD, USA), Scopus (Elsevier, Amsterdam, The Netherlands) and Web of Science Core Collection (Thomson Reuters, Toronto, ON, Canada) databases. No chronological restrictions were applied, and the most related papers published until December 2023 were included. RESULTS Gut microbiota (GM) and its metabolites are capable of modifying host androgen level, as well as prostate cancer (PCa) therapy response. Moreover, patients with inflammatory bowel disease have higher rates of prostatitis-like symptoms and a potential risk of developing PCa. CONCLUSIONS There is evidence that interventions on the GM and its metabolites have a high potential to serve as diagnostic and therapeutic tools for prostate diseases, including PCa.
Collapse
Affiliation(s)
- Lorenzo Romano
- Department of Neurosciences, Reproductive Sciences and Odontostomatology and Urology Unit, Federico II University, Naples, Italy
- Department of Woman, Child and General and Specialized Surgery, Unit of Urology, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Luigi Napolitano
- Department of Neurosciences, Reproductive Sciences and Odontostomatology and Urology Unit, Federico II University, Naples, Italy
| | - Felice Crocetto
- Department of Neurosciences, Reproductive Sciences and Odontostomatology and Urology Unit, Federico II University, Naples, Italy
| | | | - Marco De Sio
- Department of Woman, Child and General and Specialized Surgery, Unit of Urology, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Agnese Miranda
- Department of Precision Medicine and Hepatogastroenterology Unit, University of Campania "L. Vanvitelli", Naples, Italy
| | - Marco Romano
- Department of Precision Medicine and Hepatogastroenterology Unit, University of Campania "L. Vanvitelli", Naples, Italy
| | - Kateryna Priadko
- Department of Precision Medicine and Hepatogastroenterology Unit, University of Campania "L. Vanvitelli", Naples, Italy
| |
Collapse
|
39
|
Bordbar A, Manches O, Nowatzky J. Biology of HLA class I associated inflammatory diseases. Best Pract Res Clin Rheumatol 2024; 38:101977. [PMID: 39085016 PMCID: PMC11441793 DOI: 10.1016/j.berh.2024.101977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 07/15/2024] [Accepted: 07/15/2024] [Indexed: 08/02/2024]
Abstract
Human leukocyte antigen (HLA) class I association is a well-established feature of common and uncommon inflammatory diseases, but it is unknown whether it impacts the pathogenesis of these disorders. The "arthritogenic peptide" hypothesis proposed initially for HLA-B27-associated ankylosing spondylitis (AS) seems the most intuitive to serve as a model for other HLA class I-associated diseases, but evidence supporting it has been scarce. Recent technological advances and the discovery of epistatic relationships between disease-associated HLA class I and endoplasmic reticulum aminopeptidase (ERAP) coding variants have led to the generation of new data and conceptual approaches to the problem requiring its re-examination. Continued success in these endeavors holds promise to resolve a Gordian Knot in human immunobiology. It may ultimately benefit patients by enabling the development of new therapies and precision tools for assessing disease risk and predicting treatment responses.
Collapse
Affiliation(s)
- Ali Bordbar
- New York University Grossman School of Medicine, Department of Medicine, New York, NY, USA
| | - Olivier Manches
- New York University Grossman School of Medicine, Department of Medicine, New York, NY, USA
| | - Johannes Nowatzky
- New York University Grossman School of Medicine, Department of Medicine, New York, NY, USA; New York University Grossman School of Medicine, Department of Pathology, USA; New York University Grossman School of Medicine, Department of Medicine Division of Rheumatology, NYU Langone Ocular Rheumatology Program, New York, NY, USA; New York University Grossman School of Medicine, Department of Medicine, Division of Rheumatology, NYU Langone Center for Behçet's Disease, New York, NY, USA.
| |
Collapse
|
40
|
Jin XY, Li DD, Quan W, Chao Y, Zhang B. Leaky gut, circulating immune complexes, arthralgia, and arthritis in IBD: coincidence or inevitability? Front Immunol 2024; 15:1347901. [PMID: 38571963 PMCID: PMC10987687 DOI: 10.3389/fimmu.2024.1347901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 03/07/2024] [Indexed: 04/05/2024] Open
Abstract
Most host-microbiota interactions occur within the intestinal barrier, which is essential for separating the intestinal epithelium from toxins, microorganisms, and antigens in the gut lumen. Gut inflammation allows pathogenic bacteria to enter the blood stream, forming immune complexes which may deposit on organs. Despite increased circulating immune complexes (CICs) in patients with inflammatory bowel disease (IBD) and discussions among IBD experts regarding their potential pathogenic role in extra-intestinal manifestations, this phenomenon is overlooked because definitive evidence demonstrating CIC-induced extra-intestinal manifestations in IBD animal models is lacking. However, clinical observations of elevated CICs in newly diagnosed, untreated patients with IBD have reignited research into their potential pathogenic implications. Musculoskeletal symptoms are the most prevalent extra-intestinal IBD manifestations. CICs are pivotal in various arthritis forms, including reactive, rheumatoid, and Lyme arthritis and systemic lupus erythematosus. Research indicates that intestinal barrier restoration during the pre-phase of arthritis could inhibit arthritis development. In the absence of animal models supporting extra-intestinal IBD manifestations, this paper aims to comprehensively explore the relationship between CICs and arthritis onset via a multifaceted analysis to offer a fresh perspective for further investigation and provide novel insights into the interplay between CICs and arthritis development in IBD.
Collapse
Affiliation(s)
- Xi-ya Jin
- Department of Gastroenterology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Dan-dan Li
- Department of Gastroenterology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Wei Quan
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yang Chao
- Department of Gastroenterology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Bin Zhang
- Department of Gastroenterology, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
41
|
Schreiber F, Balas I, Robinson MJ, Bakdash G. Border Control: The Role of the Microbiome in Regulating Epithelial Barrier Function. Cells 2024; 13:477. [PMID: 38534321 PMCID: PMC10969408 DOI: 10.3390/cells13060477] [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: 02/16/2024] [Revised: 03/01/2024] [Accepted: 03/03/2024] [Indexed: 03/28/2024] Open
Abstract
The gut mucosal epithelium is one of the largest organs in the body and plays a critical role in regulating the crosstalk between the resident microbiome and the host. To this effect, the tight control of what is permitted through this barrier is of high importance. There should be restricted passage of harmful microorganisms and antigens while at the same time allowing the absorption of nutrients and water. An increased gut permeability, or "leaky gut", has been associated with a variety of diseases ranging from infections, metabolic diseases, and inflammatory and autoimmune diseases to neurological conditions. Several factors can affect gut permeability, including cytokines, dietary components, and the gut microbiome. Here, we discuss how the gut microbiome impacts the permeability of the gut epithelial barrier and how this can be harnessed for therapeutic purposes.
Collapse
Affiliation(s)
| | | | | | - Ghaith Bakdash
- Microbiotica Ltd., Cambridge CB10 1XL, UK; (F.S.); (I.B.); (M.J.R.)
| |
Collapse
|
42
|
Coskun Benlidayi I. Why is polymyalgia rheumatica a disease of older adults? Explanations through etiology and pathogenesis: a narrative review. Clin Rheumatol 2024; 43:851-861. [PMID: 37470883 DOI: 10.1007/s10067-023-06708-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/23/2023] [Accepted: 07/16/2023] [Indexed: 07/21/2023]
Abstract
Polymyalgia rheumatica is one of the most common inflammatory rheumatic conditions in older adults. The disease is characterized by pain and stiffness in the shoulder and pelvic girdle. Polymyalgia rheumatica is almost always observed in adults over the age of 50. The current article aimed to provide explanations for the age preference of polymyalgia rheumatica by reviewing the literature regarding disease etiology and pathogenesis. Potential factors related to the association between polymyalgia rheumatica and aging include immunosenescence/inflammaging, increased risk of infections by aging, endocrinosenescence, and age-related changes in gut microbiota. These factors and their potential contributions to immune-mediated inflammation will be discussed.
Collapse
Affiliation(s)
- Ilke Coskun Benlidayi
- Faculty of Medicine, Department of Physical Medicine and Rehabilitation, Cukurova University, Adana, Turkey.
| |
Collapse
|
43
|
Chen P, Luo Z, Lu C, Jian G, Qi X, Xiong H. Gut-immunity-joint axis: a new therapeutic target for gouty arthritis. Front Pharmacol 2024; 15:1353615. [PMID: 38464719 PMCID: PMC10920255 DOI: 10.3389/fphar.2024.1353615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 02/05/2024] [Indexed: 03/12/2024] Open
Abstract
Gouty arthritis (GA) is an inflammatory disease characterized by pain. The primary goal of current treatment strategies during GA flares remains the reduction of inflammation and pain. Research suggests that the gut microbiota and microbial metabolites contribute to the modulation of the inflammatory mechanism associated with GA, particularly through their effect on macrophage polarization. The increasing understanding of the gut-joint axis emphasizes the importance of this interaction. The primary objective of this review is to summarize existing research on the gut-immune-joint axis in GA, aiming to enhance understanding of the intricate processes and pathogenic pathways associated with pain and inflammation in GA, as documented in the published literature. The refined comprehension of the gut-joint axis may potentially contribute to the future development of analgesic drugs targeting gut microbes for GA.
Collapse
Affiliation(s)
- Pei Chen
- Hunan University of Chinese Medicine, Changsha, Hunan, China
- The Second Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
- The First Hospital of Hunan University Chinese Medicine, Changsha, Hunan, China
| | - Zhiqiang Luo
- Hunan University of Chinese Medicine, Changsha, Hunan, China
- The Second Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Chengyin Lu
- Hunan University of Chinese Medicine, Changsha, Hunan, China
- The Second Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Gonghui Jian
- Hunan University of Chinese Medicine, Changsha, Hunan, China
- College of Integrative Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Xinyu Qi
- Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Hui Xiong
- Hunan University of Chinese Medicine, Changsha, Hunan, China
- The First Hospital of Hunan University Chinese Medicine, Changsha, Hunan, China
| |
Collapse
|
44
|
Chu XJ, Song DD, Zhou MH, Chen XZ, Chu N, Li M, Li BZ, Liu SH, Hou S, Wu JB, Gong L. Perturbations in gut and respiratory microbiota in COVID-19 and influenza patients: a systematic review and meta-analysis. Front Med (Lausanne) 2024; 11:1301312. [PMID: 38405190 PMCID: PMC10884097 DOI: 10.3389/fmed.2024.1301312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 01/17/2024] [Indexed: 02/27/2024] Open
Abstract
Objectives Coronavirus disease-19 (COVID-19)/influenza poses unprecedented challenges to the global economy and healthcare services. Numerous studies have described alterations in the microbiome of COVID-19/influenza patients, but further investigation is needed to understand the relationship between the microbiome and these diseases. Herein, through systematic comparison between COVID-19 patients, long COVID-19 patients, influenza patients, no COVID-19/influenza controls and no COVID-19/influenza patients, we conducted a comprehensive review to describe the microbial change of respiratory tract/digestive tract in COVID-19/influenza patients. Methods We systematically reviewed relevant literature by searching the PubMed, Embase, and Cochrane Library databases from inception to August 12, 2023. We conducted a comprehensive review to explore microbial alterations in patients with COVID-19/influenza. In addition, the data on α-diversity were summarized and analyzed by meta-analysis. Results A total of 134 studies comparing COVID-19 patients with controls and 18 studies comparing influenza patients with controls were included. The Shannon indices of the gut and respiratory tract microbiome were slightly decreased in COVID-19/influenza patients compared to no COVID-19/influenza controls. Meanwhile, COVID-19 patients with more severe symptoms also exhibited a lower Shannon index versus COVID-19 patients with milder symptoms. The intestinal microbiome of COVID-19 patients was characterized by elevated opportunistic pathogens along with reduced short-chain fatty acid (SCFAs)-producing microbiota. Moreover, Enterobacteriaceae (including Escherichia and Enterococcus) and Lactococcus, were enriched in the gut and respiratory tract of COVID-19 patients. Conversely, Haemophilus and Neisseria showed reduced abundance in the respiratory tract of both COVID-19 and influenza patients. Conclusion In this systematic review, we identified the microbiome in COVID-19/influenza patients in comparison with controls. The microbial changes in influenza and COVID-19 are partly similar.
Collapse
Affiliation(s)
- Xiu-Jie Chu
- Department of Acute Infectious Disease Prevention and Control, Anhui Provincial Center for Disease Control and Prevention, Hefei, Anhui, China
| | - Dan-Dan Song
- Department of Acute Infectious Disease Prevention and Control, Anhui Provincial Center for Disease Control and Prevention, Hefei, Anhui, China
| | - Ming-Hua Zhou
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Xiu-Zhi Chen
- Department of Acute Infectious Disease Prevention and Control, Anhui Provincial Center for Disease Control and Prevention, Hefei, Anhui, China
| | - Na Chu
- Department of Acute Infectious Disease Prevention and Control, Anhui Provincial Center for Disease Control and Prevention, Hefei, Anhui, China
| | - Ming Li
- Department of Acute Infectious Disease Prevention and Control, Anhui Provincial Center for Disease Control and Prevention, Hefei, Anhui, China
| | - Bao-Zhu Li
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Song-Hui Liu
- School of Public Health, Bengbu Medical College, Bengbu, Anhui, China
| | - Sai Hou
- Department of Acute Infectious Disease Prevention and Control, Anhui Provincial Center for Disease Control and Prevention, Hefei, Anhui, China
| | - Jia-Bing Wu
- Department of Acute Infectious Disease Prevention and Control, Anhui Provincial Center for Disease Control and Prevention, Hefei, Anhui, China
| | - Lei Gong
- Department of Acute Infectious Disease Prevention and Control, Anhui Provincial Center for Disease Control and Prevention, Hefei, Anhui, China
| |
Collapse
|
45
|
Shen Y, Yu X, Wang Q, Yao X, Lu D, Zhou D, Wang X. Association between primary Sjögren's syndrome and gut microbiota disruption: a systematic review and meta-analysis. Clin Rheumatol 2024; 43:603-619. [PMID: 37682372 DOI: 10.1007/s10067-023-06754-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 08/02/2023] [Accepted: 08/18/2023] [Indexed: 09/09/2023]
Abstract
Evidence of gut microbiota disruption for numerous autoimmune diseases has accumulated. Recently, the relationship between the microbiota and primary Sjögren's disease has been increasingly investigated but has yet to be systematically elucidated. Therefore, a meta-analysis of publications dealing on topic was conducted. Case-control studies comparing primary Sjögren's syndrome patients and healthy controls (HCs) were systematically searched in nine databases from inception to March 1, 2023. The primary result quantitatively evaluated in this meta-analysis was the α-diversity. The secondary results qualitatively extracted and analyzed were the β-diversity and relative abundance. In total, 22 case-control studies covering 915 pSS patients and 2103 HCs were examined. The quantitative analysis revealed a slight reduction in α-diversity in pSS patients compared to HCs, with a lower Shannon-Wiener index (SMD = - 0.46, (- 0.68, - 0.25), p < 0.0001, I2 = 71%), Chao1 richness estimator (SMD = - 0.59, (- 0.86, - 0.32), p < 0.0001, I2 = 81%), and ACE index (SMD = - 0.92, (- 1.64, - 0.19), p = 0.01, I2 = 86%). However, the Simpson index (SMD = 0.01, (- 0.43, 0.46) p = 0.95, I2 = 86%) was similar in the two groups. The β-diversity significantly differed between pSS patients and HCs. Variations in the abundance of specific microbes and their metabolites and potential functions contribute to the pSS pathogenesis. Notably, the abundance of the phylum Firmicutes decreased, while that of Proteobacteria increased. SCFA-producing microbes including Ruminococcaceae, Lachnospiraceae, Faecalibacterium, Butyricicoccus, and Eubacterium hallii were depleted. In addition to diversity, the abundances of some specific microbes were related to clinical parameters. According to this systematic review and meta-analysis, gut microbiota dysbiosis, including reduced diversity, was associated with proinflammatory bacterium enrichment and anti-inflammatory bacterium depletion in pSS patients. Further research on the relationship between the gut microbiota and pSS is warranted.
Collapse
Affiliation(s)
- Yue Shen
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xue Yu
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qiao Wang
- School of Basic Medical Sciences, Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xinyi Yao
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Dingqi Lu
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Donghai Zhou
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China.
| | - Xinchang Wang
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
46
|
Navarro-Ledesma S, Carroll JD, González-Muñoz A, Burton P. Outcomes of whole-body photobiomodulation on pain, quality of life, leisure physical activity, pain catastrophizing, kinesiophobia, and self-efficacy: a prospective randomized triple-blinded clinical trial with 6 months of follow-up. Front Neurosci 2024; 18:1264821. [PMID: 38356644 PMCID: PMC10864543 DOI: 10.3389/fnins.2024.1264821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 01/12/2024] [Indexed: 02/16/2024] Open
Abstract
Background The management of fibromyalgia (FM) symptoms on a global scale remains a complex endeavor. This study endeavors to assess the impact of whole-body photobiomodulation (PBM) compared to placebo PBM on pain, functionality, and psychological symptoms in individuals afflicted with fibromyalgia. Objectives The primary objectives of this research were to conduct a comparative analysis of the effects of whole-body photobiomodulation (PBM) and placebo PBM on pain, functionality, and psychological symptoms in patients suffering from fibromyalgia (FM). Methods A total of 42 subjects were recruited from a private care practice for participation in this triple-blinded, placebo-controlled, randomized clinical trial. Participants underwent 12 treatment sessions, and assessments were conducted at various intervals, including baseline (T0), midway through the 12-session treatment (T1), at the completion of the 12 sessions (T2), and follow-ups at 2 weeks (T3), 3 months (T4), and 6 months (T5). Results Statistical analysis revealed significant reductions in pain at T2, T3, and T5. Additionally, quality of life exhibited marked improvements after sessions at T1, T2, T3, T4, and T5. Leisure activity also demonstrated statistically significant improvements at T2, T3, T4, and T5. Furthermore, kinesiophobia showed significant differences between groups immediately after treatment at T2, T3, T4, and T5. Self-efficacy, when compared between groups, demonstrated significant differences at T3, T4, and T5 (two weeks after treatment). Lastly, pain catastrophizing exhibited significant differences only at T5. Conclusion The findings of this study indicate that whole-body PBM treatment for 4 weeks resulted in significant pain reduction and improved quality of life in individuals suffering from FM. Furthermore, kinesiophobia and self-efficacy demonstrated improvements in both short-term and long-term assessments, while pain catastrophizing showed improvement at the 6-month follow-up. Consequently, whole-body PBM emerges as a promising multifactorial treatment option for FM patients, though further studies are required to validate and strengthen these results.Clinical Trial Registration:Clinicaltrials.gov, NCT0424897.
Collapse
Affiliation(s)
| | | | - Ana González-Muñoz
- Clinical Medicine and Public Health PhD Program, Faculty of Health Sciences, University of Granada, Av. de la Ilustración, Granada, Spain
- Clínica Ana González, Malaga, Spain
| | | |
Collapse
|
47
|
Goudman L, Demuyser T, Pilitsis JG, Billot M, Roulaud M, Rigoard P, Moens M. Gut dysbiosis in patients with chronic pain: a systematic review and meta-analysis. Front Immunol 2024; 15:1342833. [PMID: 38352865 PMCID: PMC10862364 DOI: 10.3389/fimmu.2024.1342833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 01/08/2024] [Indexed: 02/16/2024] Open
Abstract
Introduction Recent evidence supports the contribution of gut microbiota dysbiosis to the pathophysiology of rheumatic diseases, neuropathic pain, and neurodegenerative disorders. The bidirectional gut-brain communication network and the occurrence of chronic pain both involve contributions of the autonomic nervous system and the hypothalamic pituitary adrenal axis. Nevertheless, the current understanding of the association between gut microbiota and chronic pain is still not clear. Therefore, the aim of this study is to systematically evaluate the existing knowledge about gut microbiota alterations in chronic pain conditions. Methods Four databases were consulted for this systematic literature review: PubMed, Web of Science, Scopus, and Embase. The Newcastle-Ottawa Scale was used to assess the risk of bias. The study protocol was prospectively registered at the International prospective register of systematic reviews (PROSPERO, CRD42023430115). Alpha-diversity, β-diversity, and relative abundance at different taxonomic levels were summarized qualitatively, and quantitatively if possible. Results The initial database search identified a total of 3544 unique studies, of which 21 studies were eventually included in the systematic review and 11 in the meta-analysis. Decreases in alpha-diversity were revealed in chronic pain patients compared to controls for several metrics: observed species (SMD= -0.201, 95% CI from -0.04 to -0.36, p=0.01), Shannon index (SMD= -0.27, 95% CI from -0.11 to -0.43, p<0.001), and faith phylogenetic diversity (SMD -0.35, 95% CI from -0.08 to -0.61, p=0.01). Inconsistent results were revealed for beta-diversity. A decrease in the relative abundance of the Lachnospiraceae family, genus Faecalibacterium and Roseburia, and species of Faecalibacterium prausnitzii and Odoribacter splanchnicus, as well as an increase in Eggerthella spp., was revealed in chronic pain patients compared to controls. Discussion Indications for gut microbiota dysbiosis were revealed in chronic pain patients, with non-specific disease alterations of microbes. Systematic review registration https://www.crd.york.ac.uk/prospero/, identifier CRD42023430115.
Collapse
Affiliation(s)
- Lisa Goudman
- STIMULUS (Research and Teaching Neuromodulation Uz Brussel) Research Group, Vrije Universiteit Brussel, Brussels, Belgium
- Department of Neurosurgery, Universitair Ziekenhuis Brussel, Brussels, Belgium
- Center for Neurosciences (C4N), Vrije Universiteit Brussel, Brussels, Belgium
- Pain in Motion (PAIN) Research Group, Department of Physiotherapy, Human Physiology and Anatomy, Faculty of Physical Education and Physiotherapy, Vrije Universiteit Brussel, Brussels, Belgium
- Research Foundation—Flanders (FWO), Brussels, Belgium
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, United States
| | - Thomas Demuyser
- Department Microbiology and Infection Control, Universitair Ziekenhuis Brussel, Brussels, Belgium
- AIMS Lab, Center for Neurosciences, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Julie G. Pilitsis
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, United States
| | - Maxime Billot
- PRISMATICS Lab (Predictive Research in Spine/Neuromodulation Management and Thoracic Innovation/Cardiac Surgery), Poitiers University Hospital, Poitiers, France
| | - Manuel Roulaud
- PRISMATICS Lab (Predictive Research in Spine/Neuromodulation Management and Thoracic Innovation/Cardiac Surgery), Poitiers University Hospital, Poitiers, France
| | - Philippe Rigoard
- PRISMATICS Lab (Predictive Research in Spine/Neuromodulation Management and Thoracic Innovation/Cardiac Surgery), Poitiers University Hospital, Poitiers, France
- Department of Spine Surgery and Neuromodulation, Poitiers University Hospital, Poitiers, France
- Pprime Institute UPR 3346, CNRS, ISAE-ENSMA, University of Poitiers, Chasseneuil-du-Poitou, France
| | - Maarten Moens
- STIMULUS (Research and Teaching Neuromodulation Uz Brussel) Research Group, Vrije Universiteit Brussel, Brussels, Belgium
- Department of Neurosurgery, Universitair Ziekenhuis Brussel, Brussels, Belgium
- Center for Neurosciences (C4N), Vrije Universiteit Brussel, Brussels, Belgium
- Pain in Motion (PAIN) Research Group, Department of Physiotherapy, Human Physiology and Anatomy, Faculty of Physical Education and Physiotherapy, Vrije Universiteit Brussel, Brussels, Belgium
- Department of Radiology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| |
Collapse
|
48
|
Zou Y, Xiao W, Liu D, Li X, Li L, Peng L, Xiong Y, Gan H, Ren X. Human umbilical cord mesenchymal stem cells improve disease characterization of Sjogren's syndrome in NOD mice through regulation of gut microbiota and Treg/Th17 cellular immunity. Immun Inflamm Dis 2024; 12:e1139. [PMID: 38270310 PMCID: PMC10777879 DOI: 10.1002/iid3.1139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 09/28/2023] [Accepted: 12/22/2023] [Indexed: 01/26/2024] Open
Abstract
BACKGROUND For the unclear pathogenesis of Sjogren's syndrome (SS), further exploration is necessary. Mesenchymal stem cells (MSCs) and derived exosomes (MSCs-exo) have exhibited promising results in treating SS. OBJECT This study aimed to investigate the effect and mechanism of human umbilical cord MSCs (UC-MSCs) on SS. METHODS Nonobese Diabetic (NOD) mouse splenic T cells were co-cultured with UC-MSCs and UC-MSCs-exo, and interferon-gamma (IFN-γ), interleukin (IL)-6, IL-10, prostaglandin E2 (PGE2), and transforming growth factor-β1 (TGF-β1) levels in the supernatant were assessed by quantitative real-time polymerase chain reaction and enzyme-linked immunosorbent assay. Co-cultured T cells were injected into NOD mice via the tail vein. The inflammatory cell infiltration in the intestine and the submandibular gland was characterized by hematoxylin-eosin staining. Treg/Th17 homeostasis within the spleen was determined by flow cytometry. Gut microbiota was detected by 16S rRNA sequencing, and the relationship between differential microbiota and Treg/Th17 cytokines was analyzed by the Pearson correlation coefficient. RESULTS UC-MSCs, UC-MSCs-exo, and NOD mouse splenic T cells were successfully cultured and identified. After T cells were co-cultured with UC-MSCs and UC-MSCs-exo, both IFN-γ and IL-6 were decreased while IL-10, PGE2, and TGF-β1 were increased in transcriptional and translational levels. UC-MSCs and UC-MSCs-exo partially restored salivary secretion function, reduced Ro/SSA antibody and α-Fodrin immunoglobulin A levels, reduced inflammatory cell infiltration in the intestine and submandibular gland, raised proportion of Treg cells, decreased IFN-γ, IL-6, IL-2, IL-17, lipopolysaccharide, and tumor necrosis factor-alpha levels, and raised IL-10, Foxp3, and TGF-β1 levels by affecting co-cultured T cells. The intervention of UC-MSCs and UC-MSCs-exo improved intestinal homeostasis in NOD mice by increasing microbiota diversity and richness. Additionally, differential microbiota was significantly associated with Treg/Th17 cytokine levels. CONCLUSION Human UC-MSCs and UC-MSCs-exo improved disease characterization of SS in NOD mice through regulation of gut microbiota and Treg/Th17 cellular immunity.
Collapse
Affiliation(s)
- Yao Zou
- Jinan UniversityGuangzhouGuangdongChina
- Department of Rheumatology and Immunology, Changde Hospital, Xiangya School of MedicineCentral South UniversityChangdeHunanChina
| | - Wei Xiao
- Department of Rheumatology and Immunology, Changde Hospital, Xiangya School of MedicineCentral South UniversityChangdeHunanChina
| | - Dongzhou Liu
- Department of Rheumatology and ImmunologyShenzhen People's HospitalShenzhenGuangdongChina
| | - Xianyao Li
- Department of Rheumatology and Immunology, Changde Hospital, Xiangya School of MedicineCentral South UniversityChangdeHunanChina
| | - Lihua Li
- Department of Rheumatology and Immunology, Changde Hospital, Xiangya School of MedicineCentral South UniversityChangdeHunanChina
| | - Lijuan Peng
- Department of Rheumatology and Immunology, Changde Hospital, Xiangya School of MedicineCentral South UniversityChangdeHunanChina
| | - Ying Xiong
- Department of Rheumatology and Immunology, Changde Hospital, Xiangya School of MedicineCentral South UniversityChangdeHunanChina
| | - Haina Gan
- Department of Rheumatology and Immunology, Changde Hospital, Xiangya School of MedicineCentral South UniversityChangdeHunanChina
| | - Xiang Ren
- Department of Rheumatology and Immunology, Changde Hospital, Xiangya School of MedicineCentral South UniversityChangdeHunanChina
| |
Collapse
|
49
|
Budzinski L, von Goetze V, Chang HD. Single-cell phenotyping of bacteria combined with deep sequencing for improved contextualization of microbiome analyses. Eur J Immunol 2024; 54:e2250337. [PMID: 37863831 DOI: 10.1002/eji.202250337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 10/17/2023] [Accepted: 10/19/2023] [Indexed: 10/22/2023]
Abstract
Great effort was made to characterize the bacterial communities inhabiting the human body as a factor in disease, resulting in the realization that a wide spectrum of diseases is associated with an altered composition of the microbiome. However, the identification of disease-relevant bacteria has been hindered by the high cross-sectional diversity of individual microbiomes, and in most cases, it remains unclear whether the observed alterations are cause or consequence of disease. Hence, innovative analysis approaches are required that enable inquiries of the microbiome beyond mere taxonomic cataloging. This review highlights the utility of microbiota flow cytometry, a single-cell analysis platform to directly interrogate cellular interactions, cell conditions, and crosstalk with the host's immune system within the microbiome to take into consideration the role of microbes as critical interaction partners of the host and the spectrum of microbiome alterations, beyond compositional changes. In conjunction with advanced sequencing approaches it could reveal the genetic potential of target bacteria and advance our understanding of taxonomic diversity and gene usage in the context of the microenvironment. Single-cell bacterial phenotyping has the potential to change our perspective on the human microbiome and empower microbiome research for the development of microbiome-based therapy approaches and personalized medicine.
Collapse
Affiliation(s)
- Lisa Budzinski
- Schwiete Laboratory for Microbiota and Inflammation, German Rheumatism Research Centre Berlin - A Leibniz Institute, Berlin, Germany
| | - Victoria von Goetze
- Schwiete Laboratory for Microbiota and Inflammation, German Rheumatism Research Centre Berlin - A Leibniz Institute, Berlin, Germany
| | - Hyun-Dong Chang
- Schwiete Laboratory for Microbiota and Inflammation, German Rheumatism Research Centre Berlin - A Leibniz Institute, Berlin, Germany
| |
Collapse
|
50
|
Su X, Chen S, Liu J, Feng Y, Han E, Hao X, Liao M, Cai J, Zhang S, Niu J, He S, Huang S, Lo K, Zeng F. Composition of gut microbiota and non-alcoholic fatty liver disease: A systematic review and meta-analysis. Obes Rev 2024; 25:e13646. [PMID: 37813400 DOI: 10.1111/obr.13646] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 08/03/2023] [Accepted: 09/09/2023] [Indexed: 10/11/2023]
Abstract
The present systematic review and meta-analysis aimed to summarize the associations between gut microbiota composition and non-alcoholic fatty liver disease. To compare the differences between individuals with or without NAFLD, the standardized mean difference and 95% confidence interval were computed for each α-diversity index and relative abundance of gut microbes. The β-diversity indices were summarized in a qualitative manner. A total of 54 studies with 8894 participants were included. Overall, patients with NAFLD had moderate reduction in α-diversity indices including Shannon (SMD = -0.36, 95% CI = [-0.53, -0.19], p < 0.001) and Chao 1 (SMD = -0.42, 95% CI = [-0.68, -0.17], p = 0.001), but no significant differences were found for Simpson, observed species, phylogenetic diversity, richness, abundance-based coverage estimator, and evenness (p ranged from 0.081 to 0.953). Over 75% of the included studies reported significant differences in β-diversity. Although there was substantial interstudy heterogeneity, especially for analyses at the phylum, class, and family levels, the majority of the included studies showed alterations in the depletion of anti-inflammatory microbes (i.e., Ruminococcaceae and Coprococcus) and the enrichment of proinflammatory microbes (i.e., Fusobacterium and Escherichia) in patients with NAFLD. Perturbations in gut microbiota were associated with NAFLD, commonly reflected by a reduction in beneficial species and an increase in the pathogenic species.
Collapse
Affiliation(s)
- Xin Su
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Shiyun Chen
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Jiazi Liu
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Yonghui Feng
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Eerdun Han
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Xiaolei Hao
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Minqi Liao
- Helmholtz Zentrum München-German Research Center for Environmental Health, Institute of Epidemiology, Neuherberg, PR, Germany
| | - Jun Cai
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Shiwen Zhang
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Jianxiang Niu
- General Surgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Shihua He
- Department of Infectious Disease, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, China
| | - Shaofen Huang
- Shenzhen Qianhai Shekou Free Zone Hospital, Shenzhen, China
| | - Kenneth Lo
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
- Research Institute for Future Food, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
| | - Fangfang Zeng
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| |
Collapse
|