1
|
Costa DVS, Thomasi B, Brito GAC, Gulbransen BD, Warren CA. The role of the enteric nervous system in the pathogenesis of Clostridioides difficile infection. Nat Rev Gastroenterol Hepatol 2025:10.1038/s41575-025-01071-x. [PMID: 40404838 DOI: 10.1038/s41575-025-01071-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/16/2025] [Indexed: 05/24/2025]
Abstract
Clostridioides difficile is the leading cause of antibiotic-associated diarrhoea worldwide. In the USA, C. difficile infection (CDI) is the eighth leading cause for hospital readmission and seventh for mortality among all gastrointestinal disorders. Gastrointestinal dysmotility and/or diarrhoea occurs after the acute phase of CDI, but persistent gastrointestinal dysfunction post-infection supports contributions of neuroplasticity in the enteric nervous system (ENS), which has a key role in regulating intestinal motility and secretion, in the natural course of CDI. Here, our goal is to provide an up-to-date summary of how the ENS and extrinsic innervation of the gut are affected by CDI and how ENS responses contribute to CDI pathogenesis and outcomes. Enteric neurons and glia are targets of C. difficile toxins in humans and in preclinical model, and changes to the ENS and extrinsic innervation contribute to intestinal inflammation, damage and secretory diarrhoea. These findings suggest possible bidirectional interaction between CDI and the ENS. More studies focusing on understanding how various neurotransmitters and mediators released by the ENS and extrinsic neurons modulate immune responses to CDI could provide insight into novel pharmacological approaches to balance the host response, improve the management of CDI and prevent gastrointestinal dysfunction post-infection.
Collapse
Affiliation(s)
- Deiziane V S Costa
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA, USA.
| | - Beatriz Thomasi
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Gerly A C Brito
- Department of Morphology, School of Medicine, Federal University of Ceará, Fortaleza, Brazil
| | - Brian D Gulbransen
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Cirle A Warren
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
2
|
Goepp M, Milburn JV, Zhang B, Dong Y, Tyrrell V, Zheng X, Marshall JM, Bolsega S, Basic M, Glendinning L, Ho GT, Satsangi J, Breyer RM, Narumiya S, McSorley HJ, Schwarze JKJ, Anderson CJ, Dockrell DH, Rossi AG, Bleich A, Lucas CD, O'Donnell VB, Mole D, Arends MJ, Zhou Y, Yao C. Age-related impairment of intestinal inflammation resolution through an eicosanoid-immune-microbiota axis. Cell Host Microbe 2025; 33:671-687.e6. [PMID: 40373750 DOI: 10.1016/j.chom.2025.04.014] [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: 07/09/2024] [Revised: 02/27/2025] [Accepted: 04/15/2025] [Indexed: 05/17/2025]
Abstract
Aging manifests a decline of immune function, induces microbiome dysbiosis, drives organ inflammation, and impedes the resolution of inflammation. However, the mechanisms underlying age-related intestinal inflammation remain poorly described. Here, we find that the resolution of T cell-initiated intestinal inflammation is impaired with aging. This impairment is mediated by disrupting the immune-microbiota interplay, controlled by intestinal eicosanoid metabolism. Pharmacologically inhibiting eicosanoid biosynthesis, blocking the prostaglandin E receptor subtype 4 (EP4), or genetically ablating EP4 diminishes age-related impairment of intestinal inflammation resolution. Mechanistically, mononuclear phagocyte-intrinsic eicosanoid-EP4 signaling impedes the resolution of intestinal inflammation through fostering gut microbial dysbiosis and, more importantly, interrupting segmented filamentous bacterial adhesion to the intestinal epithelium. Colonization with EP4-ablated mouse microbiota or segmented filamentous bacteria improves the resolution of intestinal inflammation. These findings reveal that eicosanoid-dependent immune-microbiota interactions impair inflammation resolution in the aged intestine, highlighting potential intervention strategies for improving age-related gut health.
Collapse
Affiliation(s)
- Marie Goepp
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Jemma V Milburn
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Birong Zhang
- Systems Immunity University Research Institute and Division of Infection and Immunity, Cardiff University, Cardiff CF14 4XN, UK
| | - Yijia Dong
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Victoria Tyrrell
- Systems Immunity University Research Institute and Division of Infection and Immunity, Cardiff University, Cardiff CF14 4XN, UK
| | - Xiaozhong Zheng
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Jennifer M Marshall
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Silvia Bolsega
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover 30625, Germany
| | - Marijana Basic
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover 30625, Germany
| | - Laura Glendinning
- The Roslin Institute & Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Edinburgh EH25 9RG, UK
| | - Gwo-Tzer Ho
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Jack Satsangi
- Translational Gastroenterology Unit, Nuffield Department of Medicine, The University of Oxford, Oxford OX3 9DU, UK
| | - Richard M Breyer
- Department of Veterans Affairs, Tennessee Valley Health Authority, and Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Shuh Narumiya
- Alliance Laboratory for Advanced Medical Research and Department of Drug Discovery Medicine, Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| | - Henry J McSorley
- Division of Cell Signaling and Immunology, School of Life Sciences, Wellcome Trust Building, The University of Dundee, Dundee DD1 4HN, UK
| | - Jürgen K J Schwarze
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Christopher J Anderson
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - David H Dockrell
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Adriano G Rossi
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - André Bleich
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover 30625, Germany
| | - Christopher D Lucas
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Valerie B O'Donnell
- Systems Immunity University Research Institute and Division of Infection and Immunity, Cardiff University, Cardiff CF14 4XN, UK
| | - Damian Mole
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Mark J Arends
- Edinburgh Pathology, Cancer Research UK Scotland Centre, Institute of Genetics & Cancer, The University of Edinburgh, Institute of Genetics & Cancer, Edinburgh EH4 2XR, UK
| | - You Zhou
- Systems Immunity University Research Institute and Division of Infection and Immunity, Cardiff University, Cardiff CF14 4XN, UK
| | - Chengcan Yao
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK.
| |
Collapse
|
3
|
Fernandez E, Wargo JA, Helmink BA. The Microbiome and Cancer: A Translational Science Review. JAMA 2025:2833859. [PMID: 40354071 DOI: 10.1001/jama.2025.2191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
Abstract
Importance Growing evidence suggests that microbes located within the gastrointestinal tract and other anatomical locations influence the development and progression of diseases such as cancer. Observations Clinical and preclinical evidence suggests that microbes in the gastrointestinal tract and other anatomical locations, such as the respiratory tract, may affect carcinogenesis, development of metastases, cancer treatment response, and cancer treatment-related adverse effects. Within tumors of patients with cancer, microbes may affect response to treatment, and therapies that reduce or eliminate these microbes may improve outcomes in patients with cancer. Modulating gastrointestinal tract (gut) microbes through fecal microbiota transplant and other strategies such as dietary intervention (eg, high-fiber diet intervention) has improved outcomes in small studies of patients treated with cancer immunotherapy. In contrast, disruption of the gut microbiota by receipt of broad-spectrum antibiotics prior to treatment with cancer immunotherapy has been associated with poorer overall survival and higher rates of adverse effects in patients treated with immune checkpoint blockade for solid tumors and also with chimeric antigen receptor T-cell therapy for hematologic malignancies. Conclusions and Relevance Microbes in the gut and other locations in the body may influence the development and progression of cancer and may affect the response to adverse effects from cancer therapy. Future therapies targeting microbes in the gut and other locations in the body could potentially improve outcomes in patients with cancer.
Collapse
Affiliation(s)
- Estefania Fernandez
- Department of Gynecologic Oncology & Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston
| | - Jennifer A Wargo
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston
| | - Beth A Helmink
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston
| |
Collapse
|
4
|
Coxon J, Linder E, Sweet C, Magness S, Green L. Replicating Host-Microbiome Interactions: Harnessing Organ-on-a-Chip and Organoid Technologies to Model Vaginal and Lung Physiology. Annu Rev Biomed Eng 2025; 27:403-423. [PMID: 39971348 DOI: 10.1146/annurev-bioeng-110122-122343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Organ-on-a-chip (OOC) and organoid technologies are at the forefront of developing sophisticated in vitro systems that replicate complex host-microbiome interactions, including those associated with vaginal health and lung infection. We explore how these technologies provide insights into host-microbiome and host-pathogen interactions and the associated immune responses. Integrating omics data and high-resolution imaging in analyzing these models enhances our understanding of host-microbiome interactions' temporal and spatial aspects, paving the way for new diagnostic and treatment strategies. This review underscores the potential of OOC and organoid technologies in elucidating the complexities of vaginal health and lung disease, which have received less attention than other organ systems in recent organoid and OCC studies. Yet, each system presents notable characteristics, rendering them ideal candidates for these designs. Additionally, this review describes the key factors associated with each organ system and how to choose the technology setup to replicate human physiology.
Collapse
Affiliation(s)
- Jade Coxon
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, Indiana, USA;
| | - Emily Linder
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA
| | - Caden Sweet
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Scott Magness
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina, USA
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina, Chapel Hill, North Carolina, USA
| | - Leopold Green
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, Indiana, USA;
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA
| |
Collapse
|
5
|
Hilligan KL, Darrah PA, Seder RA, Sher A. Deconvoluting the interplay of innate and adaptive immunity in BCG-induced nonspecific and TB-specific host resistance. J Exp Med 2025; 222:e20240496. [PMID: 40100096 PMCID: PMC11917170 DOI: 10.1084/jem.20240496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 01/23/2025] [Accepted: 02/27/2025] [Indexed: 03/20/2025] Open
Abstract
BCG is the oldest vaccine in continuous use. While current intradermal vaccination regimens confer limited protection outside the context of pediatric extrapulmonary tuberculosis (TB), promising new data indicate that when administered mucosally or intravenously at a higher dose, BCG can induce sterilizing immunity against pulmonary TB in nonhuman primates. BCG is also known to promote nonspecific host resistance against a variety of unrelated infections and is a standard immunotherapy for bladder cancer, suggesting that this innate immune function may contribute to its protective role against TB. Here, we propose that both the mycobacterial-specific and off-target effects of BCG depend on the interplay of adaptive and innate cells and the cytokines they produce, and that the elucidation of this interaction should be a major strategy in the development of more effective BCG-based vaccines and immunotherapies.
Collapse
Affiliation(s)
| | - Patricia A. Darrah
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Robert A. Seder
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Alan Sher
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
6
|
Papamentzelopoulou M, Pitiriga VC. Unlocking the Interactions Between the Whole-Body Microbiome and HPV Infection: A Literature Review. Pathogens 2025; 14:293. [PMID: 40137778 PMCID: PMC11945791 DOI: 10.3390/pathogens14030293] [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: 02/19/2025] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 03/29/2025] Open
Abstract
The human microbiome plays a vital role in maintaining human homeostasis, acting as a key regulator of host immunity and defense mechanisms. However, dysbiotic microbial communities may cause disruption of the symbiotic relationship between the host and the local microbiota, leading to the pathogenesis of various diseases, including viral infections and cancers. One of the most common infectious agents causing cancer is the human papilloma virus (HPV), which accounts for more than 90% of cervical cancers. In most cases, the host immune system is activated and clears HPV, whereas in some cases, the infection persists and can lead to precancerous lesions. Over the last two decades, the advent of next-generation sequencing (NGS) technology and bioinformatics has allowed a thorough and in-depth analysis of the microbial composition in various anatomical niches, allowing researchers to unveil the interactions and the underlying mechanisms through which the human microbiota could affect HPV infection establishment, persistence, and progression. Accordingly, the present narrative review aims to shed light on our understanding of the role of the human microbiome in the context of HPV infection and its progression, mainly to cervical cancer. Furthermore, we explore the mechanisms by which the composition and balance of microbial communities exert potential pathogenic or protective effects, leading to either HPV persistence and disease outcomes or clearance. Special interest is given to how the microbiome can modulate host immunity to HPV infection. Lastly, we summarize the latest findings on the therapeutic efficacy of probiotics and prebiotics in preventing and/or treating HPV infections and the potential of vaginal microbiota transplantation while highlighting the significance of personalized medicine approaches emerging from NGS-based microbiome profiling and artificial intelligence (AI) for the optimal management of HPV-related diseases.
Collapse
Affiliation(s)
- Myrto Papamentzelopoulou
- Molecular Biology Unit, 1st Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Vassiliki C. Pitiriga
- Department of Microbiology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, 11527 Athens, Greece
| |
Collapse
|
7
|
McDonnell KJ. Operationalizing Team Science at the Academic Cancer Center Network to Unveil the Structure and Function of the Gut Microbiome. J Clin Med 2025; 14:2040. [PMID: 40142848 PMCID: PMC11943358 DOI: 10.3390/jcm14062040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 02/28/2025] [Accepted: 03/05/2025] [Indexed: 03/28/2025] Open
Abstract
Oncologists increasingly recognize the microbiome as an important facilitator of health as well as a contributor to disease, including, specifically, cancer. Our knowledge of the etiologies, mechanisms, and modulation of microbiome states that ameliorate or promote cancer continues to evolve. The progressive refinement and adoption of "omic" technologies (genomics, transcriptomics, proteomics, and metabolomics) and utilization of advanced computational methods accelerate this evolution. The academic cancer center network, with its immediate access to extensive, multidisciplinary expertise and scientific resources, has the potential to catalyze microbiome research. Here, we review our current understanding of the role of the gut microbiome in cancer prevention, predisposition, and response to therapy. We underscore the promise of operationalizing the academic cancer center network to uncover the structure and function of the gut microbiome; we highlight the unique microbiome-related expert resources available at the City of Hope of Comprehensive Cancer Center as an example of the potential of team science to achieve novel scientific and clinical discovery.
Collapse
Affiliation(s)
- Kevin J McDonnell
- Center for Precision Medicine, Department of Medical Oncology & Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| |
Collapse
|
8
|
Herrada AA, Rodríguez-Arriaza F, Olate-Briones A, Albornoz-Muñoz S, Faúndez-Acuña JY, Rojas-Henríquez V, Retamal-Quinteros L, Prado C, Escobedo N. Yerba Mate ( Ilex paraguariensis) Ameliorates Experimental Autoimmune Encephalomyelitis by Modulating Regulatory T Cell Function. Nutrients 2025; 17:897. [PMID: 40077767 PMCID: PMC11901674 DOI: 10.3390/nu17050897] [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: 01/27/2025] [Revised: 02/26/2025] [Accepted: 02/27/2025] [Indexed: 03/14/2025] Open
Abstract
Background/Objectives: In Latin America, yerba mate (YM) is a popular infusion processed from the leaves and stems of Ilex paraguariensis. YM has been shown to have anti-inflammatory properties in several studies, although the effect of YM on multiple sclerosis (MS) remains elusive. The purpose of this study was to examine the effect of YM on the development of MS, by using the experimental autoimmune encephalomyelitis (EAE) mouse model while also evaluating its effect over infiltration of immune cells into the central nervous system (CNS) and regulatory T cell (Treg) function. Methods: YM or vehicle were administrated to mice daily by oral gavage for seven days prior to EAE induction and during the entire course of the disease. EAE score was recorded daily, and immune cell infiltration into the CNS was measured by flow cytometry and immunofluorescence. Results: Our results showed that YM administration decreases EAE symptoms and immune cell infiltration into the CNS, along with reducing demyelination, compared to the vehicle treatment. Moreover, an increase in the Treg population, immune cells capable of generating tolerance and decreased inflammation, was observed in mice receiving YM, together with improved Treg suppressive capabilities after YM treatment in vitro. Conclusions: In summary, we showed that YM promotes an immunosuppressive environment by modulating Treg function, reducing EAE symptoms and immune cell infiltration into the brain, and suggesting that YM consumption could be a good cost-effective treatment for MS.
Collapse
Affiliation(s)
- Andrés A. Herrada
- Lymphatic Vasculature and Inflammation Research Laboratory, Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Talca 3467987, Chile; (F.R.-A.); (A.O.-B.); (S.A.-M.); (J.Y.F.-A.); (V.R.-H.); (L.R.-Q.)
| | - Francisca Rodríguez-Arriaza
- Lymphatic Vasculature and Inflammation Research Laboratory, Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Talca 3467987, Chile; (F.R.-A.); (A.O.-B.); (S.A.-M.); (J.Y.F.-A.); (V.R.-H.); (L.R.-Q.)
| | - Alexandra Olate-Briones
- Lymphatic Vasculature and Inflammation Research Laboratory, Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Talca 3467987, Chile; (F.R.-A.); (A.O.-B.); (S.A.-M.); (J.Y.F.-A.); (V.R.-H.); (L.R.-Q.)
| | - Sofía Albornoz-Muñoz
- Lymphatic Vasculature and Inflammation Research Laboratory, Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Talca 3467987, Chile; (F.R.-A.); (A.O.-B.); (S.A.-M.); (J.Y.F.-A.); (V.R.-H.); (L.R.-Q.)
| | - Jorge Y. Faúndez-Acuña
- Lymphatic Vasculature and Inflammation Research Laboratory, Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Talca 3467987, Chile; (F.R.-A.); (A.O.-B.); (S.A.-M.); (J.Y.F.-A.); (V.R.-H.); (L.R.-Q.)
| | - Victor Rojas-Henríquez
- Lymphatic Vasculature and Inflammation Research Laboratory, Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Talca 3467987, Chile; (F.R.-A.); (A.O.-B.); (S.A.-M.); (J.Y.F.-A.); (V.R.-H.); (L.R.-Q.)
| | - Ledaliz Retamal-Quinteros
- Lymphatic Vasculature and Inflammation Research Laboratory, Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Talca 3467987, Chile; (F.R.-A.); (A.O.-B.); (S.A.-M.); (J.Y.F.-A.); (V.R.-H.); (L.R.-Q.)
| | - Carolina Prado
- Laboratorio de Neuroinmunología, Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Avenida del Valle Norte #725, Huechuraba, Santiago 8580702, Chile;
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Providencia, Santiago 7510156, Chile
| | - Noelia Escobedo
- Lymphatic Vasculature and Inflammation Research Laboratory, Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Talca 3467987, Chile; (F.R.-A.); (A.O.-B.); (S.A.-M.); (J.Y.F.-A.); (V.R.-H.); (L.R.-Q.)
| |
Collapse
|
9
|
Piirainen V, König E, Husso A, Heinonen M, Iivanainen A, Pessa-Morikawa T, Niku M. Bacterial profiles of the oral, vaginal, and rectal mucosa and colostrum of periparturient sows. PLoS One 2025; 20:e0317513. [PMID: 39937738 PMCID: PMC11819496 DOI: 10.1371/journal.pone.0317513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 12/30/2024] [Indexed: 02/14/2025] Open
Abstract
The commensal microbiota influences the health, feeding efficiency, and reproductive performance of sows. The microbiota composition in the alimentary and genitourinary tracts and in colostrum/milk during pregnancy and lactation also impacts the microbiota and immune system, growth, and health of the piglets. Knowledge of the microbial compositions is important for evaluation of these effects and for discovering ways to improve the health and productivity of the sows. Oral, vaginal, and rectal mucosa and colostrum were sampled from 32 sows of variable parity in late pregnancy, and colostrum within 6 hours of delivery of the first piglet, on four commercial piglet-producing farms in Finland. Microbial compositions were analyzed by 16S rRNA gene amplicon sequencing. The most abundant genera of the oral microbiota were Rothia, Moraxella, and Streptococcus. The rectal microbiota was dominated by Clostridium sensu stricto 1. Streptococcus was the most abundant genus in the vagina and colostrum. Moderate differences in diversity and composition were observed between farms. The relative abundances of the genera Neisseria (MaAsLin 2 q = 0.002, ANCOMBC q = 0.005), Fusobacterium (MaAsLin 2 q = 0.008, ANCOMBC q = 0.04) and Bacteroides (MaAsLin 2 q < 0.005, ANCOMBC q = 0.06) were lower in oral samples and Romboutsia (MaAsLin 2 q = 0.07, ANCOMBC q = 0.05), Turicibacter (MaAsLin 2 q = 0.08, ANCOMBC q = 0.02) and Lachnospiraceae_UCG_007 (MaAsLin 2 q = 0.1, ANCOMBC q = 0.05) were higher in rectal samples of multiparous compared to primiparous sows. In vaginal samples there was a tendency of higher relative abundances of the genera Fusobacterium and Streptococcus in multiparous than primiparous sows. Among the differentially abundant taxa, F. necrophorum and F. nucleatum were identified in oral samples, F. gastrosuis and F. necrophorum in vaginal samples, and S. dysgalactiae in colostrum samples. This study provides a comprehensive overview of the mucosal and colostrum microbiota of periparturient sows during normal production conditions on Finnish commercial farms.
Collapse
Affiliation(s)
- Virpi Piirainen
- Department of Production Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Emilia König
- Department of Production Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Aleksi Husso
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Mari Heinonen
- Department of Production Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
- Research Centre for Animal Welfare, Department of Production Animal Medicine, University of Helsinki, Helsinki, Finland
| | - Antti Iivanainen
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Tiina Pessa-Morikawa
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Mikael Niku
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
10
|
Weiner HL. Immune mechanisms and shared immune targets in neurodegenerative diseases. Nat Rev Neurol 2025; 21:67-85. [PMID: 39681722 DOI: 10.1038/s41582-024-01046-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/15/2024] [Indexed: 12/18/2024]
Abstract
The immune system plays a major part in neurodegenerative diseases. In some, such as multiple sclerosis, it is the primary driver of the disease. In others, such as Alzheimer disease, amyotrophic lateral sclerosis and Parkinson disease, it has an amplifying role. Immunotherapeutic approaches that target the adaptive and innate immune systems are being explored for the treatment of almost all neurological diseases, and the targets and approaches are often common across diseases. Microglia are the primary immune cells in the brain that contribute to disease pathogenesis, and are consequently a common immune target for therapy. Other therapeutic approaches target components of the peripheral immune system, such as regulatory T cells and monocytes, which in turn act within the CNS. This Review considers in detail how microglia, monocytes and T cells contribute to the pathogenesis of multiple sclerosis, Alzheimer disease, amyotrophic lateral sclerosis and Parkinson disease, and their potential as shared therapeutic targets across these diseases. The microbiome is also highlighted as an emerging therapeutic target that indirectly modulates the immune system. Therapeutic approaches being developed to target immune function in neurodegenerative diseases are discussed, highlighting how immune-based approaches developed to treat one disease could be applicable to multiple other neurological diseases.
Collapse
Affiliation(s)
- Howard L Weiner
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
11
|
Ramírez JD, Castañeda S, Weatherhead J, Poveda C. Parasite-microbiota interactions: a pathway to innovative interventions for Chagas disease, leishmaniasis, and ascariasis. Future Microbiol 2025; 20:149-161. [PMID: 39574234 PMCID: PMC11792847 DOI: 10.1080/17460913.2024.2431417] [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: 08/19/2024] [Accepted: 11/15/2024] [Indexed: 02/02/2025] Open
Abstract
Parasitic infections are a major global health challenge, driven in part by complex interactions between parasites, host microbiota, and immune responses. Recent advances in microbiome research highlight the critical role of microbiota in influencing disease outcomes and treatment effectiveness. This review examines how changes in the microbiota impact parasite transmission, disease progression, and responses to treatment, focusing on key parasitic diseases such as Chagas disease, leishmaniasis, and ascariasis. The microbiota can either exacerbate or mitigate disease severity, depending on its composition, providing critical insights for novel therapeutic strategies. Emerging approaches discussed include the use of targeted probiotics, prebiotics, and microbiota-modulating drugs to influence parasite dynamics and enhance conventional therapies. The review also explores the potential of integrating microbiota knowledge into vaccine design and immunotherapy, aiming to develop vaccines that elicit stronger immune responses and identify new therapeutic targets. A multidisciplinary approach is essential for translating these findings into effective clinical solutions, with future research focusing on validating microbiota-based interventions in clinical settings. In conclusion, the interaction between microbiota and parasitic infections presents a promising avenue for innovative therapies, with the potential to significantly improve global health outcomes.
Collapse
Affiliation(s)
- Juan David Ramírez
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
- Molecular Microbiology Laboratory, Department of Pathology, Molecular and Cell-based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sergio Castañeda
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
| | - Jill Weatherhead
- Department of Pediatrics, Division of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
- Texas Children´s Hospital, Center for Vaccine Development, Houston, TX, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Cristina Poveda
- Department of Pediatrics, Division of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
- Texas Children´s Hospital, Center for Vaccine Development, Houston, TX, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
12
|
Brown EM, Nguyen PNU, Xavier RJ. Emerging biochemical, microbial and immunological evidence in the search for why HLA-B ∗27 confers risk for spondyloarthritis. Cell Chem Biol 2025; 32:12-24. [PMID: 39168118 PMCID: PMC11741937 DOI: 10.1016/j.chembiol.2024.07.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 05/25/2024] [Accepted: 07/22/2024] [Indexed: 08/23/2024]
Abstract
The strong association of the human leukocyte antigen B∗27 alleles (HLA-B∗27) with spondyloarthritis and related rheumatic conditions has long fascinated researchers, yet the precise mechanisms underlying its pathogenicity remain elusive. Here, we review how interplay between the microbiome, the immune system, and the enigmatic HLA-B∗27 could trigger spondyloarthritis, with a focus on whether HLA-B∗27 presents an arthritogenic peptide. We propose mechanisms by which the unique biochemical characteristics of the HLA-B∗27 protein structure, particularly its peptide binding groove, could dictate its propensity to induce pathological T cell responses. We further provide new insights into how TRBV9+ CD8+ T cells are implicated in the disease process, as well as how the immunometabolism of T cells modulates tissue-specific inflammatory responses in spondyloarthritis. Finally, we present testable models and suggest approaches to this problem in future studies given recent advances in computational biology, chemical biology, structural biology, and small-molecule therapeutics.
Collapse
Affiliation(s)
- Eric M Brown
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Center for Computational and Integrative Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | | | - Ramnik J Xavier
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Center for Computational and Integrative Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA.
| |
Collapse
|
13
|
Weagley J, Makimaa H, Cárdenas LAC, Romani A, Sullender M, Aggarwal S, Hogarty M, Rodgers R, Kennedy E, Foster L, Schriefer LA, Baldridge MT. Dynamics of Bacterial and Viral Transmission in Experimental Microbiota Transplantation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.15.633206. [PMID: 39868290 PMCID: PMC11761045 DOI: 10.1101/2025.01.15.633206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Mouse models are vital tools for discerning the relative contributions of host and microbial genetics to disease, often requiring the transplantation of microbiota between different mouse strains. Transfer methods include antibiotic treatment of recipients and colonization using either co-housing with donors or the transplantation of fecal or cecal donor material. However, the efficiency and dynamics of these methods in reconstituting recipients with donor microbes is not well understood. We thus directly compared co-housing, fecal transplantation, and cecal transplantation methods. Donor mice from Taconic Biosciences, possessing distinct microbial communities, served as the microbial source for recipient mice from Jackson Laboratories, which were treated with antibiotics to disrupt their native microbiota. We monitored microbial populations longitudinally over the course of antibiotics treatment and reconstitution using 16S rRNA gene sequencing, quantitative PCR, and shotgun sequencing of viral-like particles. As expected, antibiotic treatment rapidly depleted microbial biomass and diversity, with slow and incomplete natural recovery of the microbiota in non-transplanted control mice. While all transfer methods reconstituted recipient mice with donor microbiota, co-housing achieved this more rapidly for both bacterial and viral communities. This study provides valuable insights into microbial transfer methods, enhancing reproducibility and informing best practices for microbiota transplantation in mouse models.
Collapse
Affiliation(s)
- James Weagley
- Division of Infectious Diseases, Department of Medicine, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Heyde Makimaa
- Division of Infectious Diseases, Department of Medicine, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Luis Alberto Chica Cárdenas
- Division of Infectious Diseases, Department of Medicine, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Ana Romani
- Division of Infectious Diseases, Department of Medicine, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Meagan Sullender
- Division of Infectious Diseases, Department of Medicine, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Somya Aggarwal
- Division of Infectious Diseases, Department of Medicine, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Michael Hogarty
- Division of Infectious Diseases, Department of Medicine, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Rachel Rodgers
- Division of Infectious Diseases, Department of Medicine, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Elizabeth Kennedy
- Division of Infectious Diseases, Department of Medicine, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Lynne Foster
- Division of Infectious Diseases, Department of Medicine, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Lawrence A. Schriefer
- Division of Infectious Diseases, Department of Medicine, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Megan T. Baldridge
- Division of Infectious Diseases, Department of Medicine, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
14
|
Li D, Yang M, Ma Z, Che L, Feng B, Fang Z, Xu S, Zhuo Y, Li J, Wang J, Zhang Z, Wu Z, Lin T, Wu D, Lin Y. Glycerol Monolaurate Complex Improved Antioxidant, Anti-Inflammation, and Gut Microbiota Composition of Offspring in a Sow-Piglet Model. Vet Sci 2025; 12:24. [PMID: 39852899 PMCID: PMC11769162 DOI: 10.3390/vetsci12010024] [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: 11/18/2024] [Revised: 12/27/2024] [Accepted: 12/27/2024] [Indexed: 01/26/2025] Open
Abstract
This study aimed to investigate the effects of maternal glycerol monolaurate complex (GML) and antibiotic (acetylisovaleryltylosin tartrate, ATLL) supplementation during late gestation and lactation on the reproductive performance of sows and the growth performance of piglets. In total, 64 pregnant sows were randomly divided into control, antibiotic, 0.1% GML, and 0.2% GML groups. The GML shortened their delivery interval and farrowing duration. The ATLL increased the level of malondialdehyde (MDA) in sows and piglets and enhanced glutathione peroxidase (GSH-Px) in piglets, while reducing the tumor necrosis factor-α (TNF-α) level in sows. The GML tended to increase milk protein in the colostrum and decreased the TNF-α of sows at lactation. Meanwhile, 0.2% GML increased the serum total superoxide dismutase (T-SOD) activity and interleukin-6 level in weaned piglets and decreased the TNF-α level in sows and weaned piglets. Furthermore, ATLL decreased the microbial diversity of sows, and GML tended to increase the microbial diversity of sows and piglets. The ATLL group had an increased relative abundance of Bacteroidota in weaned piglets. The GML decreased the relative abundance of Peptostreptococcales-Tissierellales, Proteobacteria, and the harmful bacteria Romboutsia in sows. Compared with the ATLL group, the 0.2% GML reduced the relative abundance of Bacteroidota in weaned piglets. Interestingly, both ATLL and GML supplementation decreased the relative abundance of harmful bacteria Peptostreptococcaceae in sows. Correlation analysis also found positive effects of ATLL and GML in anti-inflammatory and antioxidant aspects. In conclusion, GML enhanced reproductive and growth performance by improving antioxidant and anti-inflammatory status and maintaining intestinal flora balance, making it a promising alternative to ATLL in future applications.
Collapse
Affiliation(s)
- Dan Li
- Key Laboratory of Animal Disease-Resistance Nutrition and Feed Science, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (D.L.); (Z.M.); (L.C.); (B.F.); (Z.F.); (S.X.); (Y.Z.); (J.L.); (T.L.); (D.W.)
| | - Min Yang
- Pet Nutrition and Health Research Center, Chengdu Agricultural College, Chengdu 611130, China;
| | - Zhao Ma
- Key Laboratory of Animal Disease-Resistance Nutrition and Feed Science, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (D.L.); (Z.M.); (L.C.); (B.F.); (Z.F.); (S.X.); (Y.Z.); (J.L.); (T.L.); (D.W.)
| | - Lianqiang Che
- Key Laboratory of Animal Disease-Resistance Nutrition and Feed Science, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (D.L.); (Z.M.); (L.C.); (B.F.); (Z.F.); (S.X.); (Y.Z.); (J.L.); (T.L.); (D.W.)
| | - Bin Feng
- Key Laboratory of Animal Disease-Resistance Nutrition and Feed Science, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (D.L.); (Z.M.); (L.C.); (B.F.); (Z.F.); (S.X.); (Y.Z.); (J.L.); (T.L.); (D.W.)
| | - Zhengfeng Fang
- Key Laboratory of Animal Disease-Resistance Nutrition and Feed Science, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (D.L.); (Z.M.); (L.C.); (B.F.); (Z.F.); (S.X.); (Y.Z.); (J.L.); (T.L.); (D.W.)
| | - Shengyu Xu
- Key Laboratory of Animal Disease-Resistance Nutrition and Feed Science, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (D.L.); (Z.M.); (L.C.); (B.F.); (Z.F.); (S.X.); (Y.Z.); (J.L.); (T.L.); (D.W.)
| | - Yong Zhuo
- Key Laboratory of Animal Disease-Resistance Nutrition and Feed Science, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (D.L.); (Z.M.); (L.C.); (B.F.); (Z.F.); (S.X.); (Y.Z.); (J.L.); (T.L.); (D.W.)
| | - Jian Li
- Key Laboratory of Animal Disease-Resistance Nutrition and Feed Science, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (D.L.); (Z.M.); (L.C.); (B.F.); (Z.F.); (S.X.); (Y.Z.); (J.L.); (T.L.); (D.W.)
| | - JiHhua Wang
- Calid Biotech (Wuhan) Co., Ltd., Wuhan 430073, China;
| | - Zhengfan Zhang
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China;
| | - Zehui Wu
- Sichuan Qiaozhu’er Breeding Co., Ltd., Neijiang 641100, China;
| | - Tao Lin
- Key Laboratory of Animal Disease-Resistance Nutrition and Feed Science, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (D.L.); (Z.M.); (L.C.); (B.F.); (Z.F.); (S.X.); (Y.Z.); (J.L.); (T.L.); (D.W.)
| | - De Wu
- Key Laboratory of Animal Disease-Resistance Nutrition and Feed Science, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (D.L.); (Z.M.); (L.C.); (B.F.); (Z.F.); (S.X.); (Y.Z.); (J.L.); (T.L.); (D.W.)
| | - Yan Lin
- Key Laboratory of Animal Disease-Resistance Nutrition and Feed Science, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (D.L.); (Z.M.); (L.C.); (B.F.); (Z.F.); (S.X.); (Y.Z.); (J.L.); (T.L.); (D.W.)
| |
Collapse
|
15
|
Jian J, Wei J. Ferroptosis: A New Pathway in the Interaction between Gut Microbiota and Multiple Sclerosis. FRONT BIOSCI-LANDMRK 2025; 30:26265. [PMID: 39862079 DOI: 10.31083/fbl26265] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 10/28/2024] [Accepted: 11/01/2024] [Indexed: 01/27/2025]
Abstract
Multiple sclerosis (MS) is a chronic autoimmune disorder marked by neuroinflammation, demyelination, and neuronal damage. Recent advancements highlight a novel interaction between iron-dependent cell death, known as ferroptosis, and gut microbiota, which may significantly influences the pathophysiology of MS. Ferroptosis, driven by lipid peroxidation and tightly linked to iron metabolism, is a pivotal contributor to the oxidative stress observed in MS. Concurrently, the gut microbiota, known to affect systemic immunity and neurological health, emerges as an important regulator of iron homeostasis and inflammatory responses, thereby influencing ferroptotic pathways. This review investigates how gut microbiota dysbiosis and ferroptosis impact MS, emphasizing their potential as therapeutic targets. Through an integrated examination of mechanistic pathways and clinical evidence, we discuss how targeting these interactions could lead to novel interventions that not only modulate disease progression but also offer personalized treatment strategies based on gut microbiota profiling. This synthesis aims at deepening insights into the microbial contributions to ferroptosis and their implications in MS, setting the stage for future research and therapeutic exploration.
Collapse
Affiliation(s)
- Junjie Jian
- The First College of Clinical Medical Science, China Three Gorges University, 443000 Yichang, Hubei, China
- Department of Neurology, Yichang Central People's Hospital, 443003 Yichang, Hubei, China
| | - Jun Wei
- The First College of Clinical Medical Science, China Three Gorges University, 443000 Yichang, Hubei, China
- Department of Neurology, Yichang Central People's Hospital, 443003 Yichang, Hubei, China
| |
Collapse
|
16
|
Sandys O, Stokkers PCF, Te Velde AA. DAMP-ing IBD: Extinguish the Fire and Prevent Smoldering. Dig Dis Sci 2025; 70:49-73. [PMID: 38963463 PMCID: PMC11761125 DOI: 10.1007/s10620-024-08523-5] [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: 02/21/2024] [Accepted: 06/04/2024] [Indexed: 07/05/2024]
Abstract
In inflammatory bowel diseases (IBD), the most promising therapies targeting cytokines or immune cell trafficking demonstrate around 40% efficacy. As IBD is a multifactorial inflammation of the intestinal tract, a single-target approach is unlikely to solve this problem, necessitating an alternative strategy that addresses its variability. One approach often overlooked by the pharmaceutically driven therapeutic options is to address the impact of environmental factors. This is somewhat surprising considering that IBD is increasingly viewed as a condition heavily influenced by such factors, including diet, stress, and environmental pollution-often referred to as the "Western lifestyle". In IBD, intestinal responses result from a complex interplay among the genetic background of the patient, molecules, cells, and the local inflammatory microenvironment where danger- and microbe-associated molecular patterns (D/MAMPs) provide an adjuvant-rich environment. Through activating DAMP receptors, this array of pro-inflammatory factors can stimulate, for example, the NLRP3 inflammasome-a major amplifier of the inflammatory response in IBD, and various immune cells via non-specific bystander activation of myeloid cells (e.g., macrophages) and lymphocytes (e.g., tissue-resident memory T cells). Current single-target biological treatment approaches can dampen the immune response, but without reducing exposure to environmental factors of IBD, e.g., by changing diet (reducing ultra-processed foods), the adjuvant-rich landscape is never resolved and continues to drive intestinal mucosal dysregulation. Thus, such treatment approaches are not enough to put out the inflammatory fire. The resultant smoldering, low-grade inflammation diminishes physiological resilience of the intestinal (micro)environment, perpetuating the state of chronic disease. Therefore, our hypothesis posits that successful interventions for IBD must address the complexity of the disease by simultaneously targeting all modifiable aspects: innate immunity cytokines and microbiota, adaptive immunity cells and cytokines, and factors that relate to the (micro)environment. Thus the disease can be comprehensively treated across the nano-, meso-, and microscales, rather than with a focus on single targets. A broader perspective on IBD treatment that also includes options to adapt the DAMPing (micro)environment is warranted.
Collapse
Affiliation(s)
- Oliver Sandys
- Tytgat Institute for Liver and Intestinal Research, AmsterdamUMC, AGEM, University of Amsterdam, Amsterdam, The Netherlands
| | - Pieter C F Stokkers
- Department of Gastroenterology and Hepatology, OLVG West, Amsterdam, The Netherlands
| | - Anje A Te Velde
- Tytgat Institute for Liver and Intestinal Research, AmsterdamUMC, AGEM, University of Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
17
|
Guha SK, Niyogi S. Microbial Dynamics in COVID-19: Unraveling the Impact of Human Microbiome on Disease Susceptibility and Therapeutic Strategies. Curr Microbiol 2024; 82:59. [PMID: 39720963 DOI: 10.1007/s00284-024-04041-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 12/12/2024] [Indexed: 12/26/2024]
Abstract
This review explores the bidirectional relationship between the human microbiome and SARS-CoV-2 infection, elucidating its implications for COVID-19 susceptibility, severity, and therapeutic strategies. Metagenomic analyses reveal notable alterations in microbiome composition associated with SARS-CoV-2 infection, impacting disease severity and clinical outcomes. Dysbiosis within the respiratory, gastrointestinal, oral, and skin microbiomes exacerbates COVID-19 pathology through immune dysregulation and inflammatory pathways. Understanding these microbial shifts is pivotal for devising targeted therapeutic interventions. Notably, co-infection of oral pathogens with SARS-CoV-2 worsens lung pathology, while gut microbiome dysbiosis influences viral susceptibility and severity. Potential therapeutic approaches targeting the microbiome include probiotics, antimicrobial agents, and immunomodulatory strategies. This review underscores the importance of elucidating host-microbiota interactions to advance precision medicine and public health initiatives in combating COVID-19 and other infectious diseases.
Collapse
Affiliation(s)
- Soumya Kanti Guha
- Department of Computer Application, Dinabandhu Andrews Institute of Technology and Management, BaishnabghataPatuli Township, Block-S, 1/406A, Near Satyajit Ray Park, Patuli, Kolkata, West Bengal, 700094, India
| | - Sougata Niyogi
- Department of Medical Laboratory Technology, Dinabandhu Andrews Institute of Technology and Management, BaishnabghataPatuli Township, Block-S, 1/406A, Near Satyajit Ray Park, Patuli, Kolkata, West Bengal, 700094, India.
| |
Collapse
|
18
|
Salvador-Martínez I, Murga-Moreno J, Nieto JC, Alsinet C, Enard D, Heyn H. Adaptation in human immune cells residing in tissues at the frontline of infections. Nat Commun 2024; 15:10329. [PMID: 39609395 PMCID: PMC11605006 DOI: 10.1038/s41467-024-54603-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 11/14/2024] [Indexed: 11/30/2024] Open
Abstract
Human immune cells are under constant evolutionary pressure, primarily through their role as first line of defence against pathogens. Most studies on immune adaptation are, however, based on protein-coding genes without considering their cellular context. Here, using data from the Human Cell Atlas, we infer the gene adaptation rate of the human immune landscape at cellular resolution. We find abundant cell types, like progenitor cells during development and adult cells in barrier tissues, to harbour significantly increased adaptation rates. We confirm the adaptation of tissue-resident T and NK cells in the adult lung located in compartments directly facing external challenges, such as respiratory pathogens. Analysing human iPSC-derived macrophages responding to various challenges, we find adaptation in early immune responses. Together, our study suggests host benefits to control pathogen spread at early stages of infection, providing a retrospect of forces that shaped the complexity, architecture, and function of the human body.
Collapse
Affiliation(s)
| | - Jesus Murga-Moreno
- Department of Ecology and Evolutionary Biology, University of Arizona, Tucson, AZ, USA
| | - Juan C Nieto
- CNAG, Centro Nacional de Análisis Genómico, Barcelona, Spain
| | - Clara Alsinet
- CNAG, Centro Nacional de Análisis Genómico, Barcelona, Spain
| | - David Enard
- Department of Ecology and Evolutionary Biology, University of Arizona, Tucson, AZ, USA.
| | - Holger Heyn
- CNAG, Centro Nacional de Análisis Genómico, Barcelona, Spain.
- Universitat de Barcelona (UB), Barcelona, Spain.
- ICREA, Barcelona, Spain.
| |
Collapse
|
19
|
Belnap N, Ramsey K, Carvalho ST, Nearman L, Haas H, Huentelman M, Lee K. Exploring the Frontier: The Human Microbiome's Role in Rare Childhood Neurological Diseases and Epilepsy. Brain Sci 2024; 14:1051. [PMID: 39595814 PMCID: PMC11592123 DOI: 10.3390/brainsci14111051] [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: 10/06/2024] [Revised: 10/19/2024] [Accepted: 10/19/2024] [Indexed: 11/28/2024] Open
Abstract
Emerging research into the human microbiome, an intricate ecosystem of microorganisms residing in and on our bodies, reveals that it plays a pivotal role in maintaining our health, highlighting the potential for microbiome-based interventions to prevent, diagnose, treat, and manage a myriad of diseases. The objective of this review is to highlight the importance of microbiome studies in enhancing our understanding of rare genetic epilepsy and related neurological disorders. Studies suggest that the gut microbiome, acting through the gut-brain axis, impacts the development and severity of epileptic conditions in children. Disruptions in microbial composition can affect neurotransmitter systems, inflammatory responses, and immune regulation, which are all critical factors in the pathogenesis of epilepsy. This growing body of evidence points to the potential of microbiome-targeted therapies, such as probiotics or dietary modifications, as innovative approaches to managing epilepsy. By harnessing the power of the microbiome, we stand to develop more effective and personalized treatment strategies for children affected by this disease and other rare neurological diseases.
Collapse
Affiliation(s)
- Newell Belnap
- Center for Rare Childhood Disorders, Translational Genomics Research Institute (TGen), Phoenix, AZ 85004, USA; (N.B.)
| | - Keri Ramsey
- Center for Rare Childhood Disorders, Translational Genomics Research Institute (TGen), Phoenix, AZ 85004, USA; (N.B.)
| | | | - Lexi Nearman
- Department of Biological Science, Northern Arizona University, Flagstaff, AZ 86011, USA
- TGen Integrated Microbiomics Center, Translational Genomics Research Institute (TGen), Flagstaff, AZ 86011, USA
| | - Hannah Haas
- Center for Rare Childhood Disorders, Translational Genomics Research Institute (TGen), Phoenix, AZ 85004, USA; (N.B.)
- Barrett, the Honors College, Arizona State University, Tempe, AZ 85281, USA
| | - Matt Huentelman
- Center for Rare Childhood Disorders, Translational Genomics Research Institute (TGen), Phoenix, AZ 85004, USA; (N.B.)
| | - Keehoon Lee
- TGen Integrated Microbiomics Center, Translational Genomics Research Institute (TGen), Flagstaff, AZ 86011, USA
| |
Collapse
|
20
|
Fritz SA, Bubeck Wardenburg J. A path forward for Staphylococcus aureus vaccine development. J Exp Med 2024; 221:e20240002. [PMID: 39150449 PMCID: PMC11329773 DOI: 10.1084/jem.20240002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/17/2024] Open
Abstract
The pursuit of a vaccine to quell Staphylococcus aureus disease has been unfruitful. In this Viewpoint, we explore the biological linkage between microbial niche acquisition and host immunity as a basis to guide future vaccine efforts.
Collapse
Affiliation(s)
- Stephanie A. Fritz
- Departments of Pediatrics and Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Juliane Bubeck Wardenburg
- Departments of Pediatrics and Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
21
|
Kim J, Sullivan O, Lee K, Jao J, Tamayo J, Madany AM, Wong B, Ashwood P, Ciernia AV. Repeated LPS induces training and tolerance of microglial responses across brain regions. J Neuroinflammation 2024; 21:233. [PMID: 39304952 PMCID: PMC11414187 DOI: 10.1186/s12974-024-03198-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 08/08/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND Neuroinflammation is involved in the pathogenesis of almost every central nervous system disorder. As the brain's innate immune cells, microglia fine tune their activity to a dynamic brain environment. Previous studies have shown that repeated bouts of peripheral inflammation can trigger long-term changes in microglial gene expression and function, a form of innate immune memory. METHODS AND RESULTS In this study, we used multiple low-dose lipopolysaccharide (LPS) injections in adult mice to study the acute cytokine, transcriptomic, and microglia morphological changes that contribute to the formation of immune memory in the frontal cortex, hippocampus, and striatum, as well as the long-term effects of these changes on behavior. Training and tolerance of gene expression was shared across regions, and we identified 3 unique clusters of DEGs (2xLPS-sensitive, 4xLPS-sensitive, LPS-decreased) enriched for different biological functions. 2xLPS-sensitive DEG promoters were enriched for binding sites for IRF and NFkB family transcription factors, two key regulators of innate immune memory. We quantified shifts in microglia morphological populations and found that while the proportion of ramified and rod-like microglia mostly remained consistent within brain regions and sexes with LPS treatment, there was a shift from ameboid towards hypertrophic morphological states across immune memory states and a dynamic emergence and resolution of events of microglia aligning end-to-end with repeated LPS. CONCLUSIONS Together, findings support the dynamic regulation of microglia during the formation of immune memories in the brain and support future work to exploit this model in brain disease contexts.
Collapse
Affiliation(s)
- Jennifer Kim
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, Canada
- Djavad Mowafaghian Centre for Brain Health, Vancouver, Canada
| | - Olivia Sullivan
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, Canada
- Djavad Mowafaghian Centre for Brain Health, Vancouver, Canada
| | - Kristen Lee
- Djavad Mowafaghian Centre for Brain Health, Vancouver, Canada
| | - Justin Jao
- Djavad Mowafaghian Centre for Brain Health, Vancouver, Canada
| | - Juan Tamayo
- MIND Institute, University of California Davis, Davis, USA
| | | | - Brandon Wong
- Djavad Mowafaghian Centre for Brain Health, Vancouver, Canada
| | - Paul Ashwood
- MIND Institute, University of California Davis, Davis, USA
| | - Annie Vogel Ciernia
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, Canada.
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada.
- Djavad Mowafaghian Centre for Brain Health, Vancouver, Canada.
| |
Collapse
|
22
|
Karim ZA, Reese RA, Smith AN, Blackadar ME, Arora V, Moore NM, Johnson EA. Positive impact of nutrition in the prevention of peripheral vascular disease and severe acute respiratory syndrome coronavirus 2: review. Front Nutr 2024; 11:1418028. [PMID: 39364158 PMCID: PMC11448360 DOI: 10.3389/fnut.2024.1418028] [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: 04/15/2024] [Accepted: 08/27/2024] [Indexed: 10/05/2024] Open
Abstract
Recent research has shown that there is a link between the trend of cardiovascular disease (CVD), chronic symptoms of SARS-CoV-2 (COVID-19), and medical nutrition therapy. Making positive changes to an individual's lifestyle can help to reduce the symptoms that follow exposure to CVD and COVID-19. Sustainable nutrition and lifestyle changes can positively impact an individual's health. Studies have considered the risk factors associated with the disease, medical history, the link between nutrition and peripheral vascular disease (PVD), symptom management, and the interrelationship between nutrition, COVID-19, and PVD. One study has demonstrated that Western Dietary intake can boost the innate immune system while suppressing humoral response, causing chronic inflammation and poor host defense against viruses. However, further investigation is needed to confirm. Patients with PVD and COVID-19 have experienced a reduction in side effects when prescribed a regimen of medical nutrition therapy, heart-healthy diets, and adequate physical activity before and after symptoms of both diseases appear. This approach has proven to be a protective factor during the combination of both illnesses. Our findings indicate that balanced diet and lifestyle are essential in supporting an optimal immune system that can reduce the risk of virus load in individuals at risk of infection and symptoms from COVID-19 and PVD.
Collapse
Affiliation(s)
- Zubair A Karim
- Department of Nutrition and Dietetics, College of Allied Health Sciences, Augusta University, Augusta, GA, United States
| | - Rebecca A Reese
- Department of Nutrition and Dietetics, College of Allied Health Sciences, Augusta University, Augusta, GA, United States
| | - Adrianne N Smith
- Department of Nutrition and Dietetics, College of Allied Health Sciences, Augusta University, Augusta, GA, United States
| | - Madeline E Blackadar
- Department of Nutrition and Dietetics, College of Allied Health Sciences, Augusta University, Augusta, GA, United States
| | - Vishal Arora
- Department of Medicine: Cardiology, Wellstar MCG Health, Augusta University, Augusta, GA, United States
| | - Nicole M Moore
- Department of Nutrition and Dietetics, College of Allied Health Sciences, Augusta University, Augusta, GA, United States
| | - Emily A Johnson
- Department of Nutrition and Dietetics, College of Allied Health Sciences, Augusta University, Augusta, GA, United States
| |
Collapse
|
23
|
Caetano‐Silva ME, Shrestha A, Duff AF, Kontic D, Brewster PC, Kasperek MC, Lin C, Wainwright DA, Hernandez‐Saavedra D, Woods JA, Bailey MT, Buford TW, Allen JM. Aging amplifies a gut microbiota immunogenic signature linked to heightened inflammation. Aging Cell 2024; 23:e14190. [PMID: 38725282 PMCID: PMC11320341 DOI: 10.1111/acel.14190] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 04/20/2024] [Accepted: 04/22/2024] [Indexed: 08/15/2024] Open
Abstract
Aging is associated with low-grade inflammation that increases the risk of infection and disease, yet the underlying mechanisms remain unclear. Gut microbiota composition shifts with age, harboring microbes with varied immunogenic capacities. We hypothesized the gut microbiota acts as an active driver of low-grade inflammation during aging. Microbiome patterns in aged mice strongly associated with signs of bacterial-induced barrier disruption and immune infiltration, including marked increased levels of circulating lipopolysaccharide (LPS)-binding protein (LBP) and colonic calprotectin. Ex vivo immunogenicity assays revealed that both colonic contents and mucosa of aged mice harbored increased capacity to activate toll-like receptor 4 (TLR4) whereas TLR5 signaling was unchanged. We found patterns of elevated innate inflammatory signaling (colonic Il6, Tnf, and Tlr4) and endotoxemia (circulating LBP) in young germ-free mice after 4 weeks of colonization with intestinal contents from aged mice compared with young counterparts, thus providing a direct link between aging-induced shifts in microbiota immunogenicity and host inflammation. Additionally, we discovered that the gut microbiota of aged mice exhibited unique responses to a broad-spectrum antibiotic challenge (Abx), with sustained elevation in Escherichia (Proteobacteria) and altered TLR5 immunogenicity 7 days post-Abx cessation. Together, these data indicate that old age results in a gut microbiota that differentially acts on TLR signaling pathways of the innate immune system. We found that these age-associated microbiota immunogenic signatures are less resilient to challenge and strongly linked to host inflammatory status. Gut microbiota immunogenic signatures should be thus considered as critical factors in mediating chronic inflammatory diseases disproportionally impacting older populations.
Collapse
Affiliation(s)
- Maria Elisa Caetano‐Silva
- Department of Health and KinesiologyUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
- Division of Nutritional SciencesUniversity of Illinois at Urbana ChampaignUrbanaIllinoisUSA
| | - Akriti Shrestha
- Division of Nutritional SciencesUniversity of Illinois at Urbana ChampaignUrbanaIllinoisUSA
| | - Audrey F. Duff
- Center for Microbial PathogenesisNationwide Children's HospitalColumbusOhioUSA
| | - Danica Kontic
- Center for Microbial PathogenesisNationwide Children's HospitalColumbusOhioUSA
| | - Patricia C. Brewster
- Department of Health and KinesiologyUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
| | - Mikaela C. Kasperek
- Division of Nutritional SciencesUniversity of Illinois at Urbana ChampaignUrbanaIllinoisUSA
| | - Chia‐Hao Lin
- Department of Health and KinesiologyUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
| | - Derek A. Wainwright
- Departments of Cancer Biology and Neurological SurgeryLoyola University Chicago, Stritch School of MedicineMaywoodIllinoisUSA
| | - Diego Hernandez‐Saavedra
- Department of Health and KinesiologyUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
- Division of Nutritional SciencesUniversity of Illinois at Urbana ChampaignUrbanaIllinoisUSA
| | - Jeffrey A. Woods
- Department of Health and KinesiologyUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
- Division of Nutritional SciencesUniversity of Illinois at Urbana ChampaignUrbanaIllinoisUSA
| | - Michael T. Bailey
- Center for Microbial PathogenesisNationwide Children's HospitalColumbusOhioUSA
| | - Thomas W. Buford
- Division of Gerontology, Geriatrics and Palliative Care, Department of MedicineUniversity of Alabama at BirminghamBirminghamAlabamaUSA
- Birmingham/Atlanta VA GRECCBirmingham VA Medical CenterBirminghamAlabamaUSA
| | - Jacob M. Allen
- Department of Health and KinesiologyUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
- Division of Nutritional SciencesUniversity of Illinois at Urbana ChampaignUrbanaIllinoisUSA
| |
Collapse
|
24
|
Fuhri Snethlage CM, de Wit D, Wortelboer K, Rampanelli E, Hanssen NMJ, Nieuwdorp M. Can fecal microbiota transplantations modulate autoimmune responses in type 1 diabetes? Immunol Rev 2024; 325:46-63. [PMID: 38752578 DOI: 10.1111/imr.13345] [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] [Indexed: 08/22/2024]
Abstract
Type 1 diabetes (T1D) is a chronic autoimmune disease targeting insulin-producing pancreatic beta cells. T1D is a multifactorial disease incorporating genetic and environmental factors. In recent years, the advances in high-throughput sequencing have allowed researchers to elucidate the changes in the gut microbiota taxonomy and functional capacity that accompany T1D development. An increasing number of studies have shown a role of the gut microbiota in mediating immune responses in health and disease, including autoimmunity. Fecal microbiota transplantations (FMT) have been largely used in murine models to prove a causal role of the gut microbiome in disease progression and have been shown to be a safe and effective treatment in inflammatory human diseases. In this review, we summarize and discuss recent research regarding the gut microbiota-host interactions in T1D, the current advancement in therapies for T1D, and the usefulness of FMT studies to explore microbiota-host immunity encounters in murine models and to shape the course of human type 1 diabetes.
Collapse
Affiliation(s)
- Coco M Fuhri Snethlage
- Department of Internal and Vascular Medicine, Amsterdam University Medical Center, Location AMC, Amsterdam, The Netherlands
| | - Douwe de Wit
- Department of Internal and Vascular Medicine, Amsterdam University Medical Center, Location AMC, Amsterdam, The Netherlands
| | - Koen Wortelboer
- Department of Internal and Vascular Medicine, Amsterdam University Medical Center, Location AMC, Amsterdam, The Netherlands
| | - Elena Rampanelli
- Department of Internal and Vascular Medicine, Amsterdam University Medical Center, Location AMC, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity (AII), Amsterdam, The Netherlands
| | - Nordin M J Hanssen
- Department of Internal and Vascular Medicine, Amsterdam University Medical Center, Location AMC, Amsterdam, The Netherlands
- Amsterdam Diabeter Center, Amsterdam UMC, Amsterdam, The Netherlands
| | - Max Nieuwdorp
- Department of Internal and Vascular Medicine, Amsterdam University Medical Center, Location AMC, Amsterdam, The Netherlands
- Amsterdam Diabeter Center, Amsterdam UMC, Amsterdam, The Netherlands
| |
Collapse
|
25
|
Yamaguchi H, Hsu JM, Sun L, Wang SC, Hung MC. Advances and prospects of biomarkers for immune checkpoint inhibitors. Cell Rep Med 2024; 5:101621. [PMID: 38906149 PMCID: PMC11293349 DOI: 10.1016/j.xcrm.2024.101621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 04/22/2024] [Accepted: 05/29/2024] [Indexed: 06/23/2024]
Abstract
Immune checkpoint inhibitors (ICIs) activate anti-cancer immunity by blocking T cell checkpoint molecules such as programmed death 1 (PD-1) and cytotoxic T lymphocyte-associated protein 4 (CTLA-4). Although ICIs induce some durable responses in various cancer patients, they also have disadvantages, including low response rates, the potential for severe side effects, and high treatment costs. Therefore, selection of patients who can benefit from ICI treatment is critical, and identification of biomarkers is essential to improve the efficiency of ICIs. In this review, we provide updated information on established predictive biomarkers (tumor programmed death-ligand 1 [PD-L1] expression, DNA mismatch repair deficiency, microsatellite instability high, and tumor mutational burden) and potential biomarkers currently under investigation such as tumor-infiltrated and peripheral lymphocytes, gut microbiome, and signaling pathways related to DNA damage and antigen presentation. In particular, this review aims to summarize the current knowledge of biomarkers, discuss issues, and further explore future biomarkers.
Collapse
Affiliation(s)
- Hirohito Yamaguchi
- Graduate Institute of Cell Biology, China Medical University, Taichung City 406040, Taiwan; Graduate Institute of Biomedical Sciences and Institute of Biochemistry and Molecular Biology, China Medical University, Taichung City 406040, Taiwan; Cancer Biology and Precision Therapeutics Center and Research Center for Cancer Biology, China Medical University, Taichung City 40402, Taiwan
| | - Jung-Mao Hsu
- Graduate Institute of Biomedical Sciences and Institute of Biochemistry and Molecular Biology, China Medical University, Taichung City 406040, Taiwan; Cancer Biology and Precision Therapeutics Center and Research Center for Cancer Biology, China Medical University, Taichung City 40402, Taiwan
| | - Linlin Sun
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Shao-Chun Wang
- Graduate Institute of Biomedical Sciences and Institute of Biochemistry and Molecular Biology, China Medical University, Taichung City 406040, Taiwan; Cancer Biology and Precision Therapeutics Center and Research Center for Cancer Biology, China Medical University, Taichung City 40402, Taiwan; Center for Molecular Medicine, China Medical University Hospital, Taichung City 40402, Taiwan
| | - Mien-Chie Hung
- Graduate Institute of Biomedical Sciences and Institute of Biochemistry and Molecular Biology, China Medical University, Taichung City 406040, Taiwan; Cancer Biology and Precision Therapeutics Center and Research Center for Cancer Biology, China Medical University, Taichung City 40402, Taiwan; Center for Molecular Medicine, China Medical University Hospital, Taichung City 40402, Taiwan.
| |
Collapse
|
26
|
Wang J, Yang B, Chandra J, Ivanov A, Brown JM, Florian R. Preventing fibrosis in IBD: update on immune pathways and clinical strategies. Expert Rev Clin Immunol 2024; 20:727-734. [PMID: 38475672 PMCID: PMC11180587 DOI: 10.1080/1744666x.2024.2330604] [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: 11/25/2023] [Accepted: 03/11/2024] [Indexed: 03/14/2024]
Abstract
INTRODUCTION Intestinal fibrosis is a common and serious complication of inflammatory bowel diseases (IBD) driving stricture formation in Crohn's disease patients and leading to submucosal damage in ulcerative colitis. Recent studies provided novel insights into the role of immune and nonimmune components in the pathogenesis of intestinal fibrosis. Those new findings may accelerate the development of anti-fibrotic treatment in IBD patients. AREAS COVERED This review is designed to cover the recent progress in mechanistic research and therapeutic developments on intestinal fibrosis in IBD patients, including new cell clusters, cytokines, proteins, microbiota, creeping fat, and anti-fibrotic therapies. EXPERT OPINION Due to the previously existing major obstacle of missing consensus on stricture definitions and the absence of clinical trial endpoints, testing of drugs with an anti-fibrotic mechanism is just starting in stricturing Crohn's disease (CD). A biomarker to stratify CD patients at diagnosis without any complications into at-risk populations for future strictures would be highly desirable. Further investigations are needed to identify novel mechanisms of fibrogenesis in the intestine that are targetable and ideally gut specific.
Collapse
Affiliation(s)
- Jie Wang
- Xinxiang Key Laboratory of Inflammation and Immunology, School of Medical Technology, Xinxiang Medical University, Xinxiang 453003, Henan Province, China
| | - Bo Yang
- Xinxiang Key Laboratory of Inflammation and Immunology, School of Medical Technology, Xinxiang Medical University, Xinxiang 453003, Henan Province, China
| | - Jyotsna Chandra
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Andrei Ivanov
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - J. Mark Brown
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Rieder Florian
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
- Department of Gastroenterology, Hepatology and Nutrition, Digestive Diseases Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
- Program for Global Translational Inflammatory Bowel Diseases, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| |
Collapse
|
27
|
Algavi YM, Borenstein E. Relative dispersion ratios following fecal microbiota transplant elucidate principles governing microbial migration dynamics. Nat Commun 2024; 15:4447. [PMID: 38789466 PMCID: PMC11126695 DOI: 10.1038/s41467-024-48717-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: 09/12/2023] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
Microorganisms frequently migrate from one ecosystem to another. Yet, despite the potential importance of this process in modulating the environment and the microbial ecosystem, our understanding of the fundamental forces that govern microbial dispersion is still lacking. Moreover, while theoretical models and in-vitro experiments have highlighted the contribution of species interactions to community assembly, identifying such interactions in vivo, specifically in communities as complex as the human gut, remains challenging. To address this gap, here we introduce a robust and rigorous computational framework, termed Relative Dispersion Ratio (RDR) analysis, and leverage data from well-characterized fecal microbiota transplant trials, to rigorously pinpoint dependencies between taxa during the colonization of human gastrointestinal tract. Our analysis identifies numerous pairwise dependencies between co-colonizing microbes during migration between gastrointestinal environments. We further demonstrate that identified dependencies agree with previously reported findings from in-vitro experiments and population-wide distribution patterns. Finally, we explore metabolic dependencies between these taxa and characterize the functional properties that facilitate effective dispersion. Collectively, our findings provide insights into the principles and determinants of community dynamics following ecological translocation, informing potential opportunities for precise community design.
Collapse
Affiliation(s)
- Yadid M Algavi
- Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Elhanan Borenstein
- Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv, Israel.
- The Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv, Israel.
- Santa Fe Institute, Santa Fe, NM, USA.
| |
Collapse
|
28
|
Soni J, Pandey R. Single cell genomics based insights into the impact of cell-type specific microbial internalization on disease severity. Front Immunol 2024; 15:1401320. [PMID: 38835769 PMCID: PMC11148356 DOI: 10.3389/fimmu.2024.1401320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 04/19/2024] [Indexed: 06/06/2024] Open
Abstract
Host-microbe interactions are complex and ever-changing, especially during infections, which can significantly impact human physiology in both health and disease by influencing metabolic and immune functions. Infections caused by pathogens such as bacteria, viruses, fungi, and parasites are the leading cause of global mortality. Microbes have evolved various immune evasion strategies to survive within their hosts, which presents a multifaceted challenge for detection. Intracellular microbes, in particular, target specific cell types for survival and replication and are influenced by factors such as functional roles, nutrient availability, immune evasion, and replication opportunities. Identifying intracellular microbes can be difficult because of the limitations of traditional culture-based methods. However, advancements in integrated host microbiome single-cell genomics and transcriptomics provide a promising basis for personalized treatment strategies. Understanding host-microbiota interactions at the cellular level may elucidate disease mechanisms and microbial pathogenesis, leading to targeted therapies. This article focuses on how intracellular microbes reside in specific cell types, modulating functions through persistence strategies to evade host immunity and prolong colonization. An improved understanding of the persistent intracellular microbe-induced differential disease outcomes can enhance diagnostics, therapeutics, and preventive measures.
Collapse
Affiliation(s)
- Jyoti Soni
- Division of Immunology and Infectious Disease Biology, INtegrative GENomics of HOst PathogEn (INGEN-HOPE) Laboratory, Council of Scientific & Industrial Research-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Rajesh Pandey
- Division of Immunology and Infectious Disease Biology, INtegrative GENomics of HOst PathogEn (INGEN-HOPE) Laboratory, Council of Scientific & Industrial Research-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
29
|
Murdaca G, Tagliafico L, Page E, Paladin F, Gangemi S. Gender Differences in the Interplay between Vitamin D and Microbiota in Allergic and Autoimmune Diseases. Biomedicines 2024; 12:1023. [PMID: 38790985 PMCID: PMC11117902 DOI: 10.3390/biomedicines12051023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 04/22/2024] [Accepted: 05/01/2024] [Indexed: 05/26/2024] Open
Abstract
The synergic role of vitamin D and the intestinal microbiota in the regulation of the immune system has been thoroughly described in the literature. Vitamin D deficiency and intestinal dysbiosis have shown a pathogenetic role in the development of numerous immune-mediated and allergic diseases. The physiological processes underlying aging and sex have proven to be capable of having a negative influence both on vitamin D values and the biodiversity of the microbiome. This leads to a global increase in levels of systemic inflammatory markers, with potential implications for all immune-mediated diseases and allergic conditions. Our review aims to collect and analyze the relationship between vitamin D and the intestinal microbiome with the immune system and the diseases associated with it, emphasizing the effect mediated by sexual hormones and aging.
Collapse
Affiliation(s)
- Giuseppe Murdaca
- Department of Internal Medicine, University of Genova, 16132 Genova, Italy (E.P.)
- Allergology and Clinical Immunology Unit, San Bartolomeo Hospital, 19038 Sarzana, Italy
| | - Luca Tagliafico
- Department of Internal Medicine, University of Genova, 16132 Genova, Italy (E.P.)
- Ospedale Policlinico San Martino IRCCS, Largo Rosanna Benzi 10, 16132 Genova, Italy
| | - Elena Page
- Department of Internal Medicine, University of Genova, 16132 Genova, Italy (E.P.)
- Ospedale Policlinico San Martino IRCCS, Largo Rosanna Benzi 10, 16132 Genova, Italy
| | - Francesca Paladin
- Elderly and Disabeld Department, San Paolo Hospital, 17100 Savona, Italy
| | - Sebastiano Gangemi
- School and Operative Unit of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy
| |
Collapse
|
30
|
Karaman I, Pathak A, Bayik D, Watson DC. Harnessing Bacterial Extracellular Vesicle Immune Effects for Cancer Therapy. Pathog Immun 2024; 9:56-90. [PMID: 38690563 PMCID: PMC11060327 DOI: 10.20411/pai.v9i1.657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 03/28/2024] [Indexed: 05/02/2024] Open
Abstract
There are a growing number of studies linking the composition of the human microbiome to disease states and treatment responses, especially in the context of cancer. This has raised significant interest in developing microbes and microbial products as cancer immunotherapeutics that mimic or recapitulate the beneficial effects of host-microbe interactions. Bacterial extracellular vesicles (bEVs) are nano-sized, membrane-bound particles secreted by essentially all bacteria species and contain a diverse bioactive cargo of the producing cell. They have a fundamental role in facilitating interactions among cells of the same species, different microbial species, and even with multicellular host organisms in the context of colonization (microbiome) and infection. The interaction of bEVs with the immune system has been studied extensively in the context of infection and suggests that bEV effects depend largely on the producing species. They thus provide functional diversity, while also being nonreplicative, having inherent cell-targeting qualities, and potentially overcoming natural barriers. These characteristics make them highly appealing for development as cancer immunotherapeutics. Both natively secreted and engineered bEVs are now being investigated for their application as immunotherapeutics, vaccines, drug delivery vehicles, and combinations of the above, with promising early results. This suggests that both the intrinsic immunomodulatory properties of bEVs and their ability to be modified could be harnessed for the development of next-generation microbe-inspired therapies. Nonetheless, there remain major outstanding questions regarding how the observed preclinical effectiveness will translate from murine models to primates, and humans in particular. Moreover, research into the pharmacology, toxicology, and mass manufacturing of this potential novel therapeutic platform is still at early stages. In this review, we highlight the breadth of bEV interactions with host cells, focusing on immunologic effects as the main mechanism of action of bEVs currently in preclinical development. We review the literature on ongoing efforts to develop natively secreted and engineered bEVs from a variety of bacterial species for cancer therapy and finally discuss efforts to overcome outstanding challenges that remain for clinical translation.
Collapse
Affiliation(s)
- Irem Karaman
- Bahcesehir University School of Medicine, Istanbul, Turkey
| | - Asmita Pathak
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Florida
| | - Defne Bayik
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Florida
| | - Dionysios C. Watson
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Florida
| |
Collapse
|
31
|
Qin X, Fang Z, Zhang J, Zhao W, Zheng N, Wang X. Regulatory effect of Ganoderma lucidum and its active components on gut flora in diseases. Front Microbiol 2024; 15:1362479. [PMID: 38572237 PMCID: PMC10990249 DOI: 10.3389/fmicb.2024.1362479] [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: 12/28/2023] [Accepted: 02/26/2024] [Indexed: 04/05/2024] Open
Abstract
Driven by the good developmental potential and favorable environment at this stage, Ganoderma lucidum is recognized as a precious large fungus with medicinal and nutritional health care values. Among them, polysaccharides, triterpenoids, oligosaccharides, trace elements, etc. are important bioactive components in G. lucidum. These bioactive components will have an impact on gut flora, thus alleviating diseases such as hyperglycemia, hyperlipidemia and obesity caused by gut flora disorder. While numerous studies have demonstrated the ability of G. lucidum and its active components to regulate gut flora, a systematic review of this mechanism is currently lacking. The purpose of this paper is to summarize the regulatory effects of G. lucidum and its active components on gut flora in cardiovascular, gastrointestinal and renal metabolic diseases, and summarize the research progress of G. lucidum active components in improving related diseases by regulating gut flora. Additionally, review delves into the principle by which G. lucidum and its active components can treat or assist treat diseases by regulating gut flora. The research progress of G. lucidum in intestinal tract and its potential in medicine, health food and clinical application were fully explored for researchers.
Collapse
Affiliation(s)
- Xinjie Qin
- College of Food Engineering, Jilin Engineering Normal University, Changchun, China
- Jilin Province Key Field of Social Sciences (Food Industry) Research Base, Changchun, China
| | - Zinan Fang
- College of Food Engineering, Jilin Engineering Normal University, Changchun, China
- Jilin Province Key Field of Social Sciences (Food Industry) Research Base, Changchun, China
| | - Jinkang Zhang
- College of Food Engineering, Jilin Engineering Normal University, Changchun, China
- Jilin Province Key Field of Social Sciences (Food Industry) Research Base, Changchun, China
| | - Wenbo Zhao
- College of Food Engineering, Jilin Engineering Normal University, Changchun, China
- Jilin Province Key Field of Social Sciences (Food Industry) Research Base, Changchun, China
| | - Ni Zheng
- College of Food Engineering, Jilin Engineering Normal University, Changchun, China
- Jilin Province Key Field of Social Sciences (Food Industry) Research Base, Changchun, China
| | - Xiaoe Wang
- College of Food Engineering, Jilin Engineering Normal University, Changchun, China
- Jilin Province Key Field of Social Sciences (Food Industry) Research Base, Changchun, China
| |
Collapse
|
32
|
Schäfer PSL, Dimitrov D, Villablanca EJ, Saez-Rodriguez J. Integrating single-cell multi-omics and prior biological knowledge for a functional characterization of the immune system. Nat Immunol 2024; 25:405-417. [PMID: 38413722 DOI: 10.1038/s41590-024-01768-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 01/16/2024] [Indexed: 02/29/2024]
Abstract
The immune system comprises diverse specialized cell types that cooperate to defend the host against a wide range of pathogenic threats. Recent advancements in single-cell and spatial multi-omics technologies provide rich information about the molecular state of immune cells. Here, we review how the integration of single-cell and spatial multi-omics data with prior knowledge-gathered from decades of detailed biochemical studies-allows us to obtain functional insights, focusing on gene regulatory processes and cell-cell interactions. We present diverse applications in immunology and critically assess underlying assumptions and limitations. Finally, we offer a perspective on the ongoing technological and algorithmic developments that promise to get us closer to a systemic mechanistic understanding of the immune system.
Collapse
Affiliation(s)
- Philipp Sven Lars Schäfer
- Institute for Computational Bioscience, Faculty of Medicine and Heidelberg University Hospital, Heidelberg University, Heidelberg, Germany
| | - Daniel Dimitrov
- Institute for Computational Bioscience, Faculty of Medicine and Heidelberg University Hospital, Heidelberg University, Heidelberg, Germany
| | - Eduardo J Villablanca
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
- Center of Molecular Medicine, Stockholm, Sweden
| | - Julio Saez-Rodriguez
- Institute for Computational Bioscience, Faculty of Medicine and Heidelberg University Hospital, Heidelberg University, Heidelberg, Germany.
| |
Collapse
|
33
|
Kamel M, Aleya S, Alsubih M, Aleya L. Microbiome Dynamics: A Paradigm Shift in Combatting Infectious Diseases. J Pers Med 2024; 14:217. [PMID: 38392650 PMCID: PMC10890469 DOI: 10.3390/jpm14020217] [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: 12/26/2023] [Revised: 02/15/2024] [Accepted: 02/16/2024] [Indexed: 02/24/2024] Open
Abstract
Infectious diseases have long posed a significant threat to global health and require constant innovation in treatment approaches. However, recent groundbreaking research has shed light on a previously overlooked player in the pathogenesis of disease-the human microbiome. This review article addresses the intricate relationship between the microbiome and infectious diseases and unravels its role as a crucial mediator of host-pathogen interactions. We explore the remarkable potential of harnessing this dynamic ecosystem to develop innovative treatment strategies that could revolutionize the management of infectious diseases. By exploring the latest advances and emerging trends, this review aims to provide a new perspective on combating infectious diseases by targeting the microbiome.
Collapse
Affiliation(s)
- Mohamed Kamel
- Department of Medicine and Infectious Diseases, Faculty of Veterinary Medicine, Cairo University, Giza 11221, Egypt
| | - Sami Aleya
- Faculty of Medecine, Université de Bourgogne Franche-Comté, Hauts-du-Chazal, 25030 Besançon, France;
| | - Majed Alsubih
- Department of Civil Engineering, King Khalid University, Guraiger, Abha 62529, Saudi Arabia;
| | - Lotfi Aleya
- Laboratoire de Chrono-Environnement, Université de Bourgogne Franche-Comté, UMR CNRS 6249, La Bouloie, 25030 Besançon, France;
| |
Collapse
|
34
|
Li Z, Wang Q, Huang X, Wu Y, Shan D. Microbiome's role in musculoskeletal health through the gut-bone axis insights. Gut Microbes 2024; 16:2410478. [PMID: 39387683 PMCID: PMC11469435 DOI: 10.1080/19490976.2024.2410478] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 08/06/2024] [Accepted: 09/24/2024] [Indexed: 10/15/2024] Open
Abstract
The interplay between the human microbiome and the musculoskeletal system represents a burgeoning field of research with profound implications for understanding and treating musculoskeletal disorders. This review articulates the pivotal role of the microbiome in modulating bone health, highlighting the gut-bone axis as a critical nexus for potential therapeutic intervention. Through a meticulous analysis of recent clinical research, we underscore the microbiome's influence on osteoporosis, sarcopenia, osteoarthritis, and rheumatoid arthritis, delineating both the direct and indirect mechanisms by which microbiota could impact musculoskeletal integrity and function. Our investigation reveals novel insights into the microbiota's contribution to bone density regulation, hormone production, immune modulation, and nutrient absorption, laying the groundwork for innovative microbiome-based strategies in musculoskeletal disease management. Significantly, we identify the challenges hindering the translation of research into clinical practice, including the limitations of current microbial sequencing techniques and the need for standardized methodologies in microbiome studies. Furthermore, we highlight promising directions for future research, particularly in the realm of personalized medicine, where the microbiome's variability offers unique opportunities for tailored treatment approaches. This review sets a new agenda for leveraging gut microbiota in the diagnosis, prevention, and treatment of musculoskeletal conditions, marking a pivotal step toward integrating microbiome science into clinical musculoskeletal care.
Collapse
Affiliation(s)
- Zhengrui Li
- School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qi Wang
- Jiangsu University, Zhenjiang, China
| | - Xufeng Huang
- Faculty of Dentistry, University of Debrecen, Debrecen, Hungary
| | - Yinteng Wu
- Department of Orthopedic and Trauma Surgery, The First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Dan Shan
- Faculty of Health and Medicine, Lancaster University, Lancaster, UK
- Department of Biobehavioral Sciences, Columbia University, New York, NY, USA
| |
Collapse
|
35
|
Krause FF, Mangold KI, Ruppert AL, Leister H, Hellhund-Zingel A, Lopez Krol A, Pesek J, Watzer B, Winterberg S, Raifer H, Binder K, Kinscherf R, Walker A, Nockher WA, Taudte RV, Bertrams W, Schmeck B, Kühl AA, Siegmund B, Romero R, Luu M, Göttig S, Bekeredjian-Ding I, Steinhoff U, Schütz B, Visekruna A. Clostridium sporogenes-derived metabolites protect mice against colonic inflammation. Gut Microbes 2024; 16:2412669. [PMID: 39397690 PMCID: PMC11485882 DOI: 10.1080/19490976.2024.2412669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 09/17/2024] [Accepted: 09/30/2024] [Indexed: 10/15/2024] Open
Abstract
Gut microbiota-derived metabolites play a pivotal role in the maintenance of intestinal immune homeostasis. Here, we demonstrate that the human commensal Clostridium sporogenes possesses a specific metabolic fingerprint, consisting predominantly of the tryptophan catabolite indole-3-propionic acid (IPA), the branched-chain acids (BCFAs) isobutyrate and isovalerate and the short-chain fatty acids (SCFAs) acetate and propionate. Mono-colonization of germ-free mice with C. sporogenes (CS mice) affected colonic mucosal immune cell phenotypes, including up-regulation of Il22 gene expression, and increased abundance of transcriptionally active colonic tuft cells and Foxp3+ regulatory T cells (Tregs). In DSS-induced colitis, conventional mice suffered severe inflammation accompanied by loss of colonic crypts. These symptoms were absent in CS mice. In conventional, but not CS mice, bulk RNAseq analysis of the colon revealed an increase in inflammatory and Th17-related gene signatures. C. sporogenes-derived IPA reduced IL-17A protein expression by suppressing mTOR activity and by altering ribosome-related pathways in Th17 cells. Additionally, BCFAs and SCFAs generated by C. sporogenes enhanced the activity of Tregs and increased the production of IL-22, which led to protection from colitis. Collectively, we identified C. sporogenes as a therapeutically relevant probiotic bacterium that might be employed in patients with inflammatory bowel disease (IBD).
Collapse
Affiliation(s)
- Felix F. Krause
- Institute for Medical Microbiology and Hygiene, Philipps-University, Marburg, Germany
| | - Kira I. Mangold
- Institute for Medical Microbiology and Hygiene, Philipps-University, Marburg, Germany
| | - Anna-Lena Ruppert
- Institute of Anatomy and Cell Biology, Philipps-University, Marburg, Germany
| | - Hanna Leister
- Institute for Medical Microbiology and Hygiene, Philipps-University, Marburg, Germany
| | - Anne Hellhund-Zingel
- Institute for Medical Microbiology and Hygiene, Philipps-University, Marburg, Germany
| | - Aleksandra Lopez Krol
- Institute for Medical Microbiology and Hygiene, Philipps-University, Marburg, Germany
| | - Jelena Pesek
- Core Facility for Metabolomics, Department of Medicine, Philipps-University, Marburg, Germany
| | - Bernhard Watzer
- Core Facility for Metabolomics, Department of Medicine, Philipps-University, Marburg, Germany
| | - Sarah Winterberg
- Institute of Anatomy and Cell Biology, Philipps-University, Marburg, Germany
| | - Hartmann Raifer
- Flow Cytometry Core Facility, Philipps-University, Marburg, Germany
| | - Kai Binder
- Institute for Medical Microbiology and Hygiene, Philipps-University, Marburg, Germany
| | - Ralf Kinscherf
- Institute of Anatomy and Cell Biology, Philipps-University, Marburg, Germany
| | - Alesia Walker
- Research Unit Analytical BioGeoChemistry, Helmholtz Zentrum, München, Germany
| | - Wolfgang A. Nockher
- Core Facility for Metabolomics, Department of Medicine, Philipps-University, Marburg, Germany
| | - R. Verena Taudte
- Core Facility for Metabolomics, Department of Medicine, Philipps-University, Marburg, Germany
| | - Wilhelm Bertrams
- Institute for Lung Research, Philipps-University, Marburg, Germany
| | - Bernd Schmeck
- Institute for Lung Research, Philipps-University, Marburg, Germany
- Department for Respiratory and Critical Care Medicine, Philipps-University, Marburg, Germany
- Member of the German Center for Lung Research (DZL/UGMLC, ) and German Center for Infectious Disease Research (DZIF), Marburg, Germany
| | - Anja A. Kühl
- iPATH.Berlin, Core Unit of Charité-Universitätsmedizin Berlin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Britta Siegmund
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Rossana Romero
- Institute for Medical Microbiology and Hygiene, Philipps-University, Marburg, Germany
| | - Maik Luu
- Lehrstuhl für Zelluläre Immuntherapie, Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Stephan Göttig
- Institute of Medical Microbiology and Infection Control, Goethe University Frankfurt, University Hospital, Frankfurt am Main, Germany
| | | | - Ulrich Steinhoff
- Institute for Medical Microbiology and Hygiene, Philipps-University, Marburg, Germany
| | - Burkhard Schütz
- Institute of Anatomy and Cell Biology, Philipps-University, Marburg, Germany
| | - Alexander Visekruna
- Institute for Medical Microbiology and Hygiene, Philipps-University, Marburg, Germany
| |
Collapse
|
36
|
Martínez-Ugalde E, Ávila-Akerberg V, González Martínez TM, Rebollar EA. Gene functions of the Ambystoma altamirani skin microbiome vary across space and time but potential antifungal genes are widespread and prevalent. Microb Genom 2024; 10:001181. [PMID: 38240649 PMCID: PMC10868611 DOI: 10.1099/mgen.0.001181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 01/02/2024] [Indexed: 01/23/2024] Open
Abstract
Amphibian skin microbiomes can play a critical role in host survival against emerging diseases by protecting their host against pathogens. While a plethora of biotic and abiotic factors have been shown to influence the taxonomic diversity of amphibian skin microbiomes it remains unclear whether functional genomic diversity varies in response to temporal and environmental factors. Here we applied a metagenomic approach to evaluate whether seasonality, distinct elevations/sites, and pathogen presence influenced the functional genomic diversity of the A. altamirani skin microbiome. We obtained a gene catalogue of 92 107 nonredundant annotated genes and a set of 50 unique metagenome assembled genomes (MAGs). Our analysis showed that genes linked to general and potential antifungal traits significantly differed across seasons and sampling locations at different elevations. Moreover, we found that the functional genomic diversity of A. altamirani skin microbiome differed between B. dendrobatidis infected and not infected axolotls only during winter, suggesting an interaction between seasonality and pathogen infection. In addition, we identified the presence of genes and biosynthetic gene clusters (BGCs) linked to potential antifungal functions such as biofilm formation, quorum sensing, secretion systems, secondary metabolite biosynthesis, and chitin degradation. Interestingly genes linked to these potential antifungal traits were mainly identified in Burkholderiales and Chitinophagales MAGs. Overall, our results identified functional traits linked to potential antifungal functions in the A. altamirani skin microbiome regardless of variation in the functional diversity across seasons, elevations/sites, and pathogen presence. Our findings suggest that potential antifungal traits found in Burkholderiales and Chitinophagales taxa could be related to the capacity of A. altamirani to survive in the presence of Bd, although further experimental analyses are required to test this hypothesis.
Collapse
Affiliation(s)
| | - Víctor Ávila-Akerberg
- Instituto de Ciencias Agropecuarias y Rurales, Universidad Autónoma del Estado de México, Toluca, Mexico
| | | | - Eria A. Rebollar
- Centro de Ciencias Genómicas, Universidad Nacional Autónoma de México, Cuernavaca, Mexico
| |
Collapse
|
37
|
Tuor M, LeibundGut-Landmann S. The skin mycobiome and intermicrobial interactions in the cutaneous niche. Curr Opin Microbiol 2023; 76:102381. [PMID: 37703811 DOI: 10.1016/j.mib.2023.102381] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/18/2023] [Accepted: 08/18/2023] [Indexed: 09/15/2023]
Abstract
Mammalian microbiomes have coevolved with their host to establish a stable homeostatic relationship. Multifaceted commensal-host and commensal-commensal interactions contribute to the maintenance of the equilibrium with an impact on diverse host physiological processes. Despite constant exposure to physical and chemical insults from the environment, the skin harbors a surprisingly stable microbiome. The fungal compartment of the skin microbiome, the skin mycobiome, is unique in that it is dominated by a single fungus, Malassezia. The lack in diversity suggests that the skin may provide a unique niche for this fungal genus and that Malassezia may efficiently outcompete other fungi from the skin. This opinion article examines aspects in support of this hypothesis, discusses how changes in niche conditions associate with skin mycobiome dysregulation, and highlights an emerging example of Malassezia being displaced from the skin by the emerging fungal pathogen C. auris, thereby generating a predisposing situation for fatal-invasive infection.
Collapse
Affiliation(s)
- Meret Tuor
- Section of Immunology, Vetsuisse Faculty and Institute of Experimental Immunology, University of Zurich, Switzerland
| | - Salomé LeibundGut-Landmann
- Section of Immunology, Vetsuisse Faculty and Institute of Experimental Immunology, University of Zurich, Switzerland.
| |
Collapse
|
38
|
Bang S, Shin YH, Ma X, Park SM, Graham DB, Xavier RJ, Clardy J. A Cardiolipin from Muribaculum intestinale Induces Antigen-Specific Cytokine Responses. J Am Chem Soc 2023; 145:23422-23426. [PMID: 37871232 PMCID: PMC10623554 DOI: 10.1021/jacs.3c09734] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/07/2023] [Accepted: 10/18/2023] [Indexed: 10/25/2023]
Abstract
An systematic phenotypic screen of the mouse gut microbiome for metabolites with an immunomodulatory effect identified Muribaculum intestinale as one of only two members with an oversized effect on T-cell populations. Here we report the identification and characterization of a lipid, MiCL-1, as the responsible metabolite. MiCL-1 is an 18:1-16:0 cardiolipin, whose close relatives are found on concave lipid surfaces of both mammals and bacteria. MiCL-1 was synthesized to confirm the structural analysis and functionally characterized in cell-based assays. It has a highly restrictive structure-activity profile, as its chain-switched analog fails to induce responses in any of our assays. MiCL-1 robustly induces the production of pro-inflammatory cytokines like TNF-α, IL-6, and IL-23, but has no detectable effect on the anti-inflammatory cytokine IL-10. As is the case with other recently discovered immunomodulatory lipids, MiCL-1 requires functional TLR2 and TLR1 but not TLR6 in cell-based assays.
Collapse
Affiliation(s)
- Sunghee Bang
- Department
of Biological Chemistry and Molecular Pharmacology, Harvard Medical School and Blavatnik Institute, Boston, Massachusetts 02115, United States
| | - Yern-Hyerk Shin
- Department
of Biological Chemistry and Molecular Pharmacology, Harvard Medical School and Blavatnik Institute, Boston, Massachusetts 02115, United States
| | - Xiao Ma
- Department
of Biological Chemistry and Molecular Pharmacology, Harvard Medical School and Blavatnik Institute, Boston, Massachusetts 02115, United States
- Laboratory
of Systems Pharmacology, Harvard Medical
School and Blavatnik Institute, Boston, Massachusetts 02115, United States
| | - Sung-Moo Park
- Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
- Department
of Molecular Biology, Massachusetts General
Hospital, Boston, Massachusetts 02114, United States
- Center
for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
| | - Daniel B. Graham
- Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
- Department
of Molecular Biology, Massachusetts General
Hospital, Boston, Massachusetts 02114, United States
- Center
for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
| | - Ramnik J. Xavier
- Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
- Department
of Molecular Biology, Massachusetts General
Hospital, Boston, Massachusetts 02114, United States
- Center
for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
| | - Jon Clardy
- Department
of Biological Chemistry and Molecular Pharmacology, Harvard Medical School and Blavatnik Institute, Boston, Massachusetts 02115, United States
| |
Collapse
|
39
|
Rouleau N, Levin M. The Multiple Realizability of Sentience in Living Systems and Beyond. eNeuro 2023; 10:ENEURO.0375-23.2023. [PMID: 37963652 PMCID: PMC10646883 DOI: 10.1523/eneuro.0375-23.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 10/23/2023] [Indexed: 11/16/2023] Open
Affiliation(s)
- Nicolas Rouleau
- Department of Health Sciences, Wilfrid Laurier University, Waterloo, Ontario N2L 3C5, Canada
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155
- Allen Discovery Center at, Tufts University, Medford, MA 02155
| | - Michael Levin
- Allen Discovery Center at, Tufts University, Medford, MA 02155
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02215
| |
Collapse
|
40
|
Nagakubo D, Kaibori Y. Oral Microbiota: The Influences and Interactions of Saliva, IgA, and Dietary Factors in Health and Disease. Microorganisms 2023; 11:2307. [PMID: 37764151 PMCID: PMC10535076 DOI: 10.3390/microorganisms11092307] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/31/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Recent advances in metagenomic analyses have made it easier to analyze microbiota. The microbiota, a symbiotic community of microorganisms including bacteria, archaea, fungi, and viruses within a specific environment in tissues such as the digestive tract and skin, has a complex relationship with the host. Recent studies have revealed that microbiota composition and balance particularly affect the health of the host and the onset of disease. Influences such as diet, food preferences, and sanitation play crucial roles in microbiota composition. The oral cavity is where the digestive tract directly communicates with the outside. Stable temperature and humidity provide optimal growth environments for many bacteria. However, the oral cavity is a unique environment that is susceptible to pH changes, salinity, food nutrients, and external pathogens. Recent studies have emphasized the importance of the oral microbiota, as changes in bacterial composition and balance could contribute to the development of systemic diseases. This review focuses on saliva, IgA, and fermented foods because they play critical roles in maintaining the oral bacterial environment by regulating its composition and balance. More attention should be paid to the oral microbiota and its regulatory factors in oral and systemic health.
Collapse
Affiliation(s)
- Daisuke Nagakubo
- Division of Health and Hygienic Sciences, Faculty of Pharmaceutical Sciences, Himeji Dokkyo University, 7-2-1 Kamiohno, Himeji 670-8524, Hyogo, Japan
| | - Yuichiro Kaibori
- Division of Health and Hygienic Sciences, Faculty of Pharmaceutical Sciences, Himeji Dokkyo University, 7-2-1 Kamiohno, Himeji 670-8524, Hyogo, Japan
- Laboratory of Analytics for Biomolecules, Faculty of Pharmaceutical Science, Setsunan University, 45-1 Nagaotoge-cho, Hirakata-shi 573-0101, Osaka, Japan;
| |
Collapse
|
41
|
Kargbo RB. Microbiome: The Next Frontier in Psychedelic Renaissance. J Xenobiot 2023; 13:386-401. [PMID: 37606422 PMCID: PMC10443327 DOI: 10.3390/jox13030025] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 07/11/2023] [Accepted: 07/21/2023] [Indexed: 08/23/2023] Open
Abstract
The psychedelic renaissance has reignited interest in the therapeutic potential of psychedelics for mental health and well-being. An emerging area of interest is the potential modulation of psychedelic effects by the gut microbiome-the ecosystem of microorganisms in our digestive tract. This review explores the intersection of the gut microbiome and psychedelic therapy, underlining potential implications for personalized medicine and mental health. We delve into the current understanding of the gut-brain axis, its influence on mood, cognition, and behavior, and how the microbiome may affect the metabolism and bioavailability of psychedelic substances. We also discuss the role of microbiome variations in shaping individual responses to psychedelics, along with potential risks and benefits. Moreover, we consider the prospect of microbiome-targeted interventions as a fresh approach to boost or modulate psychedelic therapy's effectiveness. By integrating insights from the fields of psychopharmacology, microbiology, and neuroscience, our objective is to advance knowledge about the intricate relationship between the microbiome and psychedelic substances, thereby paving the way for novel strategies to optimize mental health outcomes amid the ongoing psychedelic renaissance.
Collapse
Affiliation(s)
- Robert B Kargbo
- Usona Institute, 2800 Woods Hollow Rd., Madison, WI 53711-5300, USA
| |
Collapse
|