1
|
Zhao X, Zan J, Sun Z, Xue X, Ren H, Fu H, Si F, Jin X. Toxic effects of anionic polyacrylamide on the developmental stages of Oryzias melastigma embryos and larvae. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2025; 284:107402. [PMID: 40359789 DOI: 10.1016/j.aquatox.2025.107402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 04/22/2025] [Accepted: 05/05/2025] [Indexed: 05/15/2025]
Abstract
Anionic Polyacrylamide (APAM) is widely used in oil extraction processes, serving as an oil-repellent polymer and constituting a critical component of water-based drilling fluids. The environmental and ecological effects of APAM on fishery resources have attracted significant attention, yet its toxic mechanism in marine fish at early developmental stages remains poorly understood. The potential effects of APAM on marine medaka (Oryzias melastigma) embryos were investigated by exposing them to 0, 120, 240, 480, and 960 mg/L for 18 d. APAM exposure caused developmental toxicity in embryos, leading to reduced heart rates, delayed and decreased hatching, increased mortality and malformations. The activities of superoxide dismutase (SOD) and catalase (CAT) initially increased after 2 d of exposure but decreased after 8 and 18 days of prolonged stress, while malondialdehyde (MDA) concentration increased, causing lipid peroxidation and worsening oxidative damage. After 18 days of APAM exposure, low and medium concentrations increased the expression of cardiovascular genes GATA4 and NKX2.5, while high concentrations decreased NKX2.5, leading to heart defects like elongated hearts and pericardial cysts. Additionally, low concentrations significantly boosted nervous system genes SHHA and SYN2A, enhancing swimming behaviors, whereas high concentrations suppressed these genes, reducing swimming activity. In conclusion, this study demonstrated that APAM exposure causes developmental toxicity, oxidative stress, neurotoxicity, and disrupts early cardiac development in O. melastigma embryos, providing insight into its toxic effects on early marine fish development.
Collapse
Affiliation(s)
- Xinya Zhao
- Hebei Key Laboratory of Ocean Dynamics Resources and Environments, Hebei Normal University of Science & Technology, Qinhuangdao 066600, China; Hebei Key Laboratory of the Bohai Sea Fish Germplasm Resources Conservation and Utilization, Beidaihe Central Experiment Station, Chinese Academy of Fishery Sciences, Qinhuangdao 066100, China; National Agricultural Experimental Station for Fishery Resources and Environment Qinhuangdao, Qinhuangdao 066100, China; Bohai Sea Fishery Research Center, Chinese Academy of Fishery Science, Qinhuangdao 066100, China
| | - Jiangwei Zan
- Hebei Key Laboratory of the Bohai Sea Fish Germplasm Resources Conservation and Utilization, Beidaihe Central Experiment Station, Chinese Academy of Fishery Sciences, Qinhuangdao 066100, China; National Agricultural Experimental Station for Fishery Resources and Environment Qinhuangdao, Qinhuangdao 066100, China; Bohai Sea Fishery Research Center, Chinese Academy of Fishery Science, Qinhuangdao 066100, China
| | - Zhaohui Sun
- Hebei Key Laboratory of the Bohai Sea Fish Germplasm Resources Conservation and Utilization, Beidaihe Central Experiment Station, Chinese Academy of Fishery Sciences, Qinhuangdao 066100, China; National Agricultural Experimental Station for Fishery Resources and Environment Qinhuangdao, Qinhuangdao 066100, China; Bohai Sea Fishery Research Center, Chinese Academy of Fishery Science, Qinhuangdao 066100, China
| | - Xiangping Xue
- Hebei Key Laboratory of the Bohai Sea Fish Germplasm Resources Conservation and Utilization, Beidaihe Central Experiment Station, Chinese Academy of Fishery Sciences, Qinhuangdao 066100, China; National Agricultural Experimental Station for Fishery Resources and Environment Qinhuangdao, Qinhuangdao 066100, China; Bohai Sea Fishery Research Center, Chinese Academy of Fishery Science, Qinhuangdao 066100, China
| | - Hai Ren
- Hebei Key Laboratory of Ocean Dynamics Resources and Environments, Hebei Normal University of Science & Technology, Qinhuangdao 066600, China
| | - Huiru Fu
- Hebei Key Laboratory of Ocean Dynamics Resources and Environments, Hebei Normal University of Science & Technology, Qinhuangdao 066600, China
| | - Fei Si
- Hebei Key Laboratory of the Bohai Sea Fish Germplasm Resources Conservation and Utilization, Beidaihe Central Experiment Station, Chinese Academy of Fishery Sciences, Qinhuangdao 066100, China; National Agricultural Experimental Station for Fishery Resources and Environment Qinhuangdao, Qinhuangdao 066100, China; Bohai Sea Fishery Research Center, Chinese Academy of Fishery Science, Qinhuangdao 066100, China.
| | - Xiaomin Jin
- Hebei Key Laboratory of Ocean Dynamics Resources and Environments, Hebei Normal University of Science & Technology, Qinhuangdao 066600, China.
| |
Collapse
|
2
|
Cherrier M, Teo TH, Corrêa RO, Picard M, Couesnon A, Lebreton C, Carbone F, Masson C, Schnupf P, Cerf-Bensussan N, Gaboriau-Routhiau V. Hematopoietic MyD88 orchestrates the control of gut colonization by segmented filamentous bacteria. Mucosal Immunol 2025; 18:717-729. [PMID: 40090466 DOI: 10.1016/j.mucimm.2025.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 02/22/2025] [Accepted: 03/10/2025] [Indexed: 03/18/2025]
Abstract
Host-microbiota cooperation is critical for successful intestinal homeostasis. The commensal segmented filamentous bacteria (SFB) are crucial for orchestrating the post-natal maturation of the host gut immune system and establishing a healthy state of physiological inflammation, which largely depends on their intimate attachment to the ileal mucosa. However, the signaling pathways used by SFB to induce gut immune responses and how such responses ultimately control SFB colonization remain controversial. Using gnotobiotic approaches, we showed that SFB load is controlled by complex interactions involving the gut microbiota and the host immune system. Therefore, to clearly determine the role of host immune responses induced by SFB in directly controlling their growth, immunodeficient mice monocolonized with SFB were used. Here, we show that in the absence of a complex microbiota, the humoral immune response is dispensable to control SFB growth in the jejunum and ileum, shortly and later after colonization. In contrast, MyD88 signaling in myeloid cells is critical for licensing interleukin (IL)-22 production by type 3 innate lymphoid cells (ILC3) and CD4+ T cells, which ultimately limits SFB expansion. Thus, by revisiting the hierarchy of immune mechanisms that directly control SFB growth, our results emphasize the necessary and sufficient role of a hematopoietic MyD88/IL-22 axis.
Collapse
Affiliation(s)
- Marie Cherrier
- Université Paris Cité, Imagine Institute, INSERM UMR1163, Laboratory of Intestinal Immunity, 75015 Paris, France
| | - Teck Hui Teo
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, F-75015 Paris, France; A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research, Singapore
| | - Renan Oliveira Corrêa
- Université Paris Cité, Imagine Institute, INSERM UMR1163, Laboratory of Intestinal Immunity, 75015 Paris, France
| | - Marion Picard
- Université Paris Cité, Imagine Institute, INSERM UMR1163, Laboratory of Intestinal Immunity, 75015 Paris, France
| | - Aurélie Couesnon
- Université Paris Cité, Imagine Institute, INSERM UMR1163, Laboratory of Intestinal Immunity, 75015 Paris, France
| | - Corinne Lebreton
- Université Paris Cité, Imagine Institute, INSERM UMR1163, Laboratory of Intestinal Immunity, 75015 Paris, France
| | - Francesco Carbone
- Université Paris Cité, Imagine Institute, INSERM UMR 1163, Labtech Single-Cell@Imagine, 75015 Paris, France
| | - Cécile Masson
- Université Paris Cité, Imagine Institute, Structure Fédérative de Recherche Necker, Bioinformatics Core Facility, 75015 Paris, France
| | - Pamela Schnupf
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, F-75015 Paris, France
| | - Nadine Cerf-Bensussan
- Université Paris Cité, Imagine Institute, INSERM UMR1163, Laboratory of Intestinal Immunity, 75015 Paris, France
| | - Valérie Gaboriau-Routhiau
- Université Paris Cité, Imagine Institute, INSERM UMR1163, Laboratory of Intestinal Immunity, 75015 Paris, France; Université Paris-Saclay, INRAe, AgroParisTech, Micalis Institute, 78350 Jouy-en-Josas, France.
| |
Collapse
|
3
|
Lisicka W, Earley ZM, Sifakis JJ, Erickson SA, Mattingly JR, Wu-Woods NJ, Krishnamurthy SR, Belkaid Y, Ismagilov RF, Cyster JG, Riesenfeld SJ, Bendelac A, Jabri B. Immunoglobulin A controls intestinal virus colonization to preserve immune homeostasis. Cell Host Microbe 2025; 33:498-511.e10. [PMID: 40154490 DOI: 10.1016/j.chom.2025.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 11/26/2024] [Accepted: 03/05/2025] [Indexed: 04/01/2025]
Abstract
Immunoglobulin A (IgA) is the predominant immunoglobulin isotype in mammals, primarily secreted at type I mucosal surfaces. Despite its abundance, the precise role of secretory IgA in the intestinal lumen, where it coats a diverse array of commensal microbiota, has remained elusive. Our study reveals that germinal center IgA responses are essential for preventing chronic colonization of the gut by specific viruses. In the absence of IgA, chronic viral colonization triggers an antigen-driven expansion of CD8αβ+ intraepithelial lymphocytes (IELs). Although these IELs are unable to clear the virus, they contribute to maintaining homeostasis by regulating its load and type I interferon responses. Consequently, IgA deficiency increases susceptibility to colitis in genetically susceptible hosts or following chemical induction but only in the presence of viral pathobionts requiring IgA for their clearance. These findings underscore the potential vulnerability of IgA-deficient individuals to immunopathology when exposed to selective viral pathobionts.
Collapse
Affiliation(s)
- Wioletta Lisicka
- Committee on Immunology, University of Chicago, Chicago, IL, USA; Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Zachary M Earley
- Committee on Immunology, University of Chicago, Chicago, IL, USA; Department of Medicine, University of Chicago, Chicago, IL, USA; Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
| | - Joseph J Sifakis
- Department of Chemistry, University of Chicago, Chicago, IL, USA
| | - Steven A Erickson
- Committee on Immunology, University of Chicago, Chicago, IL, USA; Department of Pathology, University of Chicago, Chicago, IL, USA
| | - Jonathan R Mattingly
- Committee on Immunology, University of Chicago, Chicago, IL, USA; Department of Pathology, University of Chicago, Chicago, IL, USA
| | - Natalie J Wu-Woods
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Siddharth R Krishnamurthy
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Yasmine Belkaid
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Rustem F Ismagilov
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, USA; Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Jason G Cyster
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
| | - Samantha J Riesenfeld
- Committee on Immunology, University of Chicago, Chicago, IL, USA; Department of Medicine, University of Chicago, Chicago, IL, USA; Institute for Biophysical Dynamics, University of Chicago, Chicago, IL, USA; Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Albert Bendelac
- Committee on Immunology, University of Chicago, Chicago, IL, USA; Department of Pathology, University of Chicago, Chicago, IL, USA
| | - Bana Jabri
- Committee on Immunology, University of Chicago, Chicago, IL, USA; Department of Medicine, University of Chicago, Chicago, IL, USA; Department of Pathology, University of Chicago, Chicago, IL, USA; Paris City University, Imagine Institute, Paris, France.
| |
Collapse
|
4
|
Wang M, Liu Y, Zhong L, Wu F, Wang J. Advancements in the investigation of gut microbiota-based strategies for stroke prevention and treatment. Front Immunol 2025; 16:1533343. [PMID: 40103814 PMCID: PMC11914130 DOI: 10.3389/fimmu.2025.1533343] [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: 11/23/2024] [Accepted: 02/11/2025] [Indexed: 03/20/2025] Open
Abstract
Stroke represents a predominant cause of mortality and disability on a global scale, impacting millions annually and exerting a considerable strain on healthcare systems. The incidence of stroke exhibits regional variability, with ischemic stroke accounting for the majority of occurrences. Post-stroke complications, such as cognitive impairment, motor dysfunction, and recurrent stroke, profoundly affect patients' quality of life. Recent advancements have elucidated the microbiota-gut-brain axis (MGBA), underscoring the complex interplay between gut health and brain function. Dysbiosis, characterized by an imbalance in gut microbiota, is significantly linked to an elevated risk of stroke and unfavorable outcomes. The MGBA plays a crucial role in modulating immune function, neurotransmitter levels, and metabolic byproducts, which may intensify neuroinflammation and impair cerebral health. This review elucidates the role of MGBA in stroke pathophysiology and explores potential gut-targeted therapeutic strategies to reduce stroke risk and promote recovery, including probiotics, prebiotics, pharmacological interventions, and dietary modifications. However, the current prevention and treatment strategies based on intestinal flora still face many problems, such as the large difference of individual intestinal flora, the stability of efficacy, and the long-term safety need to be considered. Further research needs to be strengthened to promote its better application in clinical practice.
Collapse
Affiliation(s)
| | | | | | | | - Jinjin Wang
- Department of Gastroenterology, The First People’s Hospital of Xiaoshan District, Hangzhou, Zhejiang, China
| |
Collapse
|
5
|
Babu MA, Jyothi S R, Kaur I, Kumar S, Sharma N, Kumar MR, Rajput P, Ali H, Gupta G, Subramaniyan V, Wong LS, Kumarasamy V. The role of GATA4 in mesenchymal stem cell senescence: A new frontier in regenerative medicine. Regen Ther 2025; 28:214-226. [PMID: 39811069 PMCID: PMC11731776 DOI: 10.1016/j.reth.2024.11.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 11/21/2024] [Indexed: 01/16/2025] Open
Abstract
The Mesenchymal Stem Cell (MSC) is a multipotent progenitor cell with known differentiation potential towards various cell lineage, making it an appealing candidate for regenerative medicine. One major contributing factor to age-related MSC dysfunction is cellular senescence, which is the hallmark of relatively irreversible growth arrest and changes in functional properties. GATA4, a zinc-finger transcription factor, emerges as a critical regulator in MSC biology. Originally identified as a key regulator of heart development and specification, GATA4 has since been connected to several aspects of cellular processes, including stem cell proliferation and differentiation. Accumulating evidence suggests that the involvement of GATA4-nuclear signalizing in the process of MSC senescence-related traits may contribute to age-induced alterations in MSC behavior. GATA4 emerged as the central player in MSC senescence, interacting with several signaling pathways. Studies have shown that GATA4 expression is reduced with age in MSCs, which is associated with increased expression levels of senescence markers and impaired regenerative potential. At the mechanistic level, GATA4 regulates the expression of genes involved in cell cycle regulation, DNA repair, and oxidative stress response, thereby influencing the senescence phenotype in MSCs. The findings underscore the critical function of GATA4 in MSC homeostasis and suggest a promising new target to restore stem cell function during aging and disease. A better understanding of the molecular mechanisms that underlie GATA4 mediated modulation of MSC senescence would provide an opportunity to develop new therapies to revitalize old MSCs to increase their regenerative function for therapeutic purposes in regenerative medicine.
Collapse
Affiliation(s)
- M. Arockia Babu
- Institute of Pharmaceutical Research, GLA University, Mathura, UP, India
| | - Renuka Jyothi S
- Department of Biotechnology and Genetics, Jain (Deemed-to-be) University, Bengaluru, Karnataka, 560069, India
| | - Irwanjot Kaur
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan, 303012, India
| | - Sachin Kumar
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - Naveen Sharma
- Chandigarh Pharmacy College, Chandigarh Group of College, Jhanjeri, Mohali, 140307, Punjab, India
| | - M. Ravi Kumar
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh, 531162, India
| | - Pranchal Rajput
- School of Applied and Life Sciences, Division of Research and Innovation, Uttaranchal University, Dehradun, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India
| | - Gaurav Gupta
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
- Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Vetriselvan Subramaniyan
- Department of Medical Sciences, School of Medical and Life Sciences, Sunway University, Malaysia
| | - Ling Shing Wong
- Faculty of Health and Life Sciences, INTI International University, Nilai, 71800, Malaysia
| | - Vinoth Kumarasamy
- Department of Parasitology and Medical Entomology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, 56000, Cheras, Kuala Lumpur, Malaysia
| |
Collapse
|
6
|
Jena KK, Mambu J, Boehmer D, Sposito B, Millet V, de Sousa Casal J, Muendlein HI, Spreafico R, Fenouil R, Spinelli L, Wurbel S, Riquier C, Galland F, Naquet P, Chasson L, Elkins M, Mitsialis V, Ketelut-Carneiro N, Bugda Gwilt K, Thiagarajah JR, Ruan HB, Lin Z, Lien E, Shao F, Chou J, Poltorak A, Ordovas-Montanes J, Fitzgerald KA, Snapper SB, Broggi A, Zanoni I. Type III interferons induce pyroptosis in gut epithelial cells and impair mucosal repair. Cell 2024; 187:7533-7550.e23. [PMID: 39500322 DOI: 10.1016/j.cell.2024.10.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 06/25/2024] [Accepted: 10/07/2024] [Indexed: 11/30/2024]
Abstract
Tissue damage and repair are hallmarks of inflammation. Despite a wealth of information on the mechanisms that govern tissue damage, mechanistic insight into how inflammation affects repair is lacking. Here, we investigated how interferons influence tissue repair after damage to the intestinal mucosa. We found that type III, not type I or type II, interferons delay epithelial cell regeneration by inducing the upregulation of Z-DNA-binding protein 1 (ZBP1). Z-nucleic acids formed following intestinal damage are sensed by ZBP1, leading to caspase-8 activation and the cleavage of gasdermin C (GSDMC). Cleaved GSDMC drives epithelial cell death by pyroptosis and delays repair of the large or small intestine after colitis or irradiation, respectively. The type III interferon/ZBP1/caspase-8/GSDMC axis is also active in patients with inflammatory bowel disease (IBD). Our findings highlight the capacity of type III interferons to delay gut repair, which has implications for IBD patients or individuals exposed to radiation therapies.
Collapse
Affiliation(s)
- Kautilya K Jena
- Division of Immunology, Harvard Medical School and Boston Children's Hospital, Boston, MA 02115, USA
| | - Julien Mambu
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), 13288 Marseille Cedex, France
| | - Daniel Boehmer
- Division of Immunology, Harvard Medical School and Boston Children's Hospital, Boston, MA 02115, USA; Department of Medicine II, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Benedetta Sposito
- Division of Immunology, Harvard Medical School and Boston Children's Hospital, Boston, MA 02115, USA
| | - Virginie Millet
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), 13288 Marseille Cedex, France
| | - Joshua de Sousa Casal
- Division of Gastroenterology, Harvard Medical School and Boston Children's Hospital, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Program in Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Hayley I Muendlein
- Department of Immunology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Roberto Spreafico
- Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, CA, USA
| | - Romain Fenouil
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), 13288 Marseille Cedex, France
| | - Lionel Spinelli
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), 13288 Marseille Cedex, France
| | - Sarah Wurbel
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), 13288 Marseille Cedex, France
| | - Chloé Riquier
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), 13288 Marseille Cedex, France
| | - Franck Galland
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), 13288 Marseille Cedex, France
| | - Philippe Naquet
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), 13288 Marseille Cedex, France
| | - Lionel Chasson
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), 13288 Marseille Cedex, France
| | - Megan Elkins
- Division of Immunology, Harvard Medical School and Boston Children's Hospital, Boston, MA 02115, USA
| | - Vanessa Mitsialis
- Division of Gastroenterology, Harvard Medical School and Boston Children's Hospital, Boston, MA 02115, USA
| | - Natália Ketelut-Carneiro
- Program in Innate Immunity, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| | - Katlynn Bugda Gwilt
- Division of Gastroenterology, Harvard Medical School and Boston Children's Hospital, Boston, MA 02115, USA
| | - Jay R Thiagarajah
- Division of Gastroenterology, Harvard Medical School and Boston Children's Hospital, Boston, MA 02115, USA
| | - Hai-Bin Ruan
- Department of Integrative Biology and Physiology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Zhaoyu Lin
- State Key Laboratory of Pharmaceutical Biotechnology, Ministry of Education Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, National Resource Center for Mutant Mice of China, Nanjing Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing 210061, China
| | - Egil Lien
- Program in Innate Immunity, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA; Center for Molecular inflammation Research, Norwegian University of Science and Technology (NTNU), 7491 Trondheim, Norway
| | - Feng Shao
- National Institute of Biological Sciences, Beijing 102206, China
| | - Janet Chou
- Division of Immunology, Harvard Medical School and Boston Children's Hospital, Boston, MA 02115, USA
| | - Alexander Poltorak
- Department of Immunology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Jose Ordovas-Montanes
- Division of Gastroenterology, Harvard Medical School and Boston Children's Hospital, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Program in Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Katherine A Fitzgerald
- Program in Innate Immunity, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| | - Scott B Snapper
- Division of Gastroenterology, Harvard Medical School and Boston Children's Hospital, Boston, MA 02115, USA
| | - Achille Broggi
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), 13288 Marseille Cedex, France.
| | - Ivan Zanoni
- Division of Immunology, Harvard Medical School and Boston Children's Hospital, Boston, MA 02115, USA; Division of Gastroenterology, Harvard Medical School and Boston Children's Hospital, Boston, MA 02115, USA; Program in Immunology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
7
|
De Giovanni M, Inverso D, Iannacone M. Understanding local immunity to enable regionalized medicine. EMBO J 2024; 43:5788-5792. [PMID: 39385044 PMCID: PMC11612237 DOI: 10.1038/s44318-024-00255-6] [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: 07/31/2024] [Revised: 08/14/2024] [Accepted: 09/09/2024] [Indexed: 10/11/2024] Open
Abstract
This commentary of the Sparks of Science series from the Catalysts program discusses how dissection of local immunity paves the way for the selective targeting of specific niches in future therapeutics.
Collapse
Affiliation(s)
- Marco De Giovanni
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy.
- Vita-Salute San Raffaele University, Milan, Italy.
| | - Donato Inverso
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy.
- Vita-Salute San Raffaele University, Milan, Italy.
| | - Matteo Iannacone
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy.
- Vita-Salute San Raffaele University, Milan, Italy.
- Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
8
|
Bildstein T, Charbit-Henrion F, Azabdaftari A, Cerf-Bensussan N, Uhlig HH. Cellular and molecular basis of proximal small intestine disorders. Nat Rev Gastroenterol Hepatol 2024; 21:687-709. [PMID: 39117867 DOI: 10.1038/s41575-024-00962-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/28/2024] [Indexed: 08/10/2024]
Abstract
The proximal part of the small intestine, including duodenum and jejunum, is not only dedicated to nutrient digestion and absorption but is also a highly regulated immune site exposed to environmental factors. Host-protective responses against pathogens and tolerance to food antigens are essential functions in the small intestine. The cellular ecology and molecular pathways to maintain those functions are complex. Maladaptation is highlighted by common immune-mediated diseases such as coeliac disease, environmental enteric dysfunction or duodenal Crohn's disease. An expanding spectrum of more than 100 rare monogenic disorders inform on causative molecular mechanisms of nutrient absorption, epithelial homeostasis and barrier function, as well as inflammatory immune responses and immune regulation. Here, after summarizing the architectural and cellular traits that underlie the functions of the proximal intestine, we discuss how the integration of tissue immunopathology and molecular mechanisms can contribute towards our understanding of disease and guide diagnosis. We propose an integrated mechanism-based taxonomy and discuss the latest experimental approaches to gain new mechanistic insight into these disorders with large disease burden worldwide as well as implications for therapeutic interventions.
Collapse
Affiliation(s)
- Tania Bildstein
- Great Ormond Street Hospital for Children, Department of Paediatric Gastroenterology, London, UK
| | - Fabienne Charbit-Henrion
- Department of Genomic Medicine for Rare Diseases, Necker-Enfants Malades Hospital, APHP, University of Paris-Cité, Paris, France
- INSERM UMR1163, Intestinal Immunity, Institut Imagine, Paris, France
| | - Aline Azabdaftari
- Translational Gastroenterology Unit, Nuffield Department of Medicine, Oxford, UK
| | | | - Holm H Uhlig
- Translational Gastroenterology Unit, Nuffield Department of Medicine, Oxford, UK.
- Department of Paediatrics, University of Oxford, Oxford, UK.
- National Institute for Health and Care Research (NIHR) Oxford Biomedical Research Centre, Oxford, UK.
| |
Collapse
|
9
|
Gong Z, Xue Q, Luo Y, Yu B, Hua B, Liu Z. The interplay between the microbiota and opioid in the treatment of neuropathic pain. Front Microbiol 2024; 15:1390046. [PMID: 38919504 PMCID: PMC11197152 DOI: 10.3389/fmicb.2024.1390046] [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: 02/22/2024] [Accepted: 05/24/2024] [Indexed: 06/27/2024] Open
Abstract
Neuropathic pain (NP) is characterized by its complex and multifactorial nature and limited responses to opioid therapy; NP is associated with risks of drug resistance, addiction, difficulty in treatment cessation, and psychological disorders. Emerging research on gut microbiota and their metabolites has demonstrated their effectiveness in alleviating NP and augmenting opioid-based pain management, concurrently mitigating the adverse effects of opioids. This review addresses the following key points: (1) the current advances in gut microbiota research and the challenges in using opioids to treat NP, (2) the reciprocal effects and benefits of gut microbiota on NP, and (3) the interaction between opioids with gut microbiota, as well as the benefits of gut microbiota in opioid-based treatment of NP. Through various intricate mechanisms, gut microbiota influences the onset and progression of NP, ultimately enhancing the efficacy of opioids in the management of NP. These insights pave the way for further pragmatic clinical research, ultimately enhancing the efficacy of opioid-based pain management.
Collapse
Affiliation(s)
- Zexiong Gong
- Department of Anesthesiology, Health Science Center, Shenzhen Second People's Hospital/The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Qingsheng Xue
- Department of Anesthesiology, School of Medicine, Ruijin Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Yan Luo
- Department of Anesthesiology, School of Medicine, Ruijin Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Buwei Yu
- Department of Anesthesiology, School of Medicine, Ruijin Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Bo Hua
- Department of Anesthesiology, Health Science Center, Shenzhen Second People's Hospital/The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Zhiheng Liu
- Department of Anesthesiology, Health Science Center, Shenzhen Second People's Hospital/The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| |
Collapse
|
10
|
Cullum E, Perez-Betancourt Y, Shi M, Gkika E, Schneewind O, Missiakas D, Golovkina T. Deficiency in non-classical major histocompatibility class II-like molecule, H2-O confers protection against Staphylococcus aureus in mice. PLoS Pathog 2024; 20:e1012306. [PMID: 38843309 PMCID: PMC11185455 DOI: 10.1371/journal.ppat.1012306] [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: 01/23/2024] [Revised: 06/18/2024] [Accepted: 05/29/2024] [Indexed: 06/19/2024] Open
Abstract
Staphylococcus aureus is a human-adapted pathogen that replicates by asymptomatically colonizing its host. S. aureus is also the causative agent of purulent skin and soft tissue infections as well as bloodstream infections that result in the metastatic seeding of abscess lesions in all organ tissues. Prolonged colonization, infection, disease relapse, and recurrence point to the versatile capacity of S. aureus to bypass innate and adaptive immune defenses as well as the notion that some hosts fail to generate protective immune responses. Here, we find a genetic trait that provides protection against this pathogen. Mice lacking functional H2-O, the equivalent of human HLA-DO, inoculated with a mouse-adapted strain of S. aureus, efficiently decolonize the pathogen. Further, these decolonized animals resist subsequent bloodstream challenge with methicillin-resistant S. aureus. A genetic approach demonstrates that T-cell dependent B cell responses are required to control S. aureus colonization and infection in H2-O-deficient mice. Reduced bacterial burdens in these animals correlate with increased titers and enhanced phagocytic activity of S. aureus-specific antibodies. H2-O negatively regulates the loading of high affinity peptides on major histocompatibility class II (MHC-II) molecules. Thus, we hypothesize that immune responses against S. aureus are derepressed in mice lacking H2-O because more high affinity peptides are presented by MHC-II. We speculate that loss-of-function HLA-DO alleles may similarly control S. aureus replication in humans.
Collapse
Affiliation(s)
- Emily Cullum
- Department of Microbiology, University of Chicago, Chicago, Illinois, United States of America
- Committee on Immunology, University of Chicago, Chicago, Illinois, United States of America
| | - Yunys Perez-Betancourt
- Department of Microbiology, University of Chicago, Chicago, Illinois, United States of America
- Howard T. Ricketts Laboratory, University of Chicago, Chicago, Illinois, United States of America
| | - Miaomiao Shi
- Department of Microbiology, University of Chicago, Chicago, Illinois, United States of America
- Howard T. Ricketts Laboratory, University of Chicago, Chicago, Illinois, United States of America
| | - Eirinaios Gkika
- Department of Microbiology, University of Chicago, Chicago, Illinois, United States of America
| | - Olaf Schneewind
- Department of Microbiology, University of Chicago, Chicago, Illinois, United States of America
| | - Dominique Missiakas
- Department of Microbiology, University of Chicago, Chicago, Illinois, United States of America
- Howard T. Ricketts Laboratory, University of Chicago, Chicago, Illinois, United States of America
| | - Tatyana Golovkina
- Department of Microbiology, University of Chicago, Chicago, Illinois, United States of America
- Committee on Immunology, University of Chicago, Chicago, Illinois, United States of America
- Committee on Microbiology, University of Chicago, Chicago, Illinois, United States of America
- Committee on Genetics, Genomics and System Biology, University of Chicago, Chicago, Illinois, United States of America
| |
Collapse
|
11
|
Abadie V, Han AS, Jabri B, Sollid LM. New Insights on Genes, Gluten, and Immunopathogenesis of Celiac Disease. Gastroenterology 2024; 167:4-22. [PMID: 38670280 PMCID: PMC11283582 DOI: 10.1053/j.gastro.2024.03.042] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 03/10/2024] [Accepted: 03/11/2024] [Indexed: 04/28/2024]
Abstract
Celiac disease (CeD) is a gluten-induced enteropathy that develops in genetically susceptible individuals upon consumption of cereal gluten proteins. It is a unique and complex immune disorder to study as the driving antigen is known and the tissue targeted by the immune reaction can be interrogated. This review integrates findings gained from genetic, biochemical, and immunologic studies, which together have revealed mechanisms of gluten peptide modification and HLA binding, thereby enabling a maladapted anti-gluten immune response. Observations in human samples combined with experimental mouse models have revealed that the gluten-induced immune response involves CD4+ T cells, cytotoxic CD8+ T cells, and B cells; their cross-talks are critical for the tissue-damaging response. The emergence of high-throughput technologies is increasing our understanding of the phenotype, location, and presumably function of the gluten-specific cells, which are all required to identify novel therapeutic targets and strategies for CeD.
Collapse
Affiliation(s)
- Valérie Abadie
- Department of Medicine, University of Chicago, Chicago, Illinois; Section of Gastroenterology, Nutrition and Hepatology, University of Chicago, Chicago, Illinois; Committee on Immunology, University of Chicago, Chicago, Illinois.
| | - Arnold S Han
- Columbia Center for Translational Immunology, Columbia University, New York, New York; Department of Microbiology and Immunology, Columbia University, New York, New York; Department of Medicine, Digestive and Liver Diseases, Columbia University, New York, New York
| | - Bana Jabri
- Department of Medicine, University of Chicago, Chicago, Illinois; Section of Gastroenterology, Nutrition and Hepatology, University of Chicago, Chicago, Illinois; Committee on Immunology, University of Chicago, Chicago, Illinois; Department of Pathology, University of Chicago, Chicago, Illinois; Department of Pediatrics, University of Chicago, Chicago, Illinois
| | - Ludvig M Sollid
- Norwegian Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Immunology, Oslo University Hospital-Rikshospitalet, Oslo, Norway
| |
Collapse
|
12
|
Akinsuyi OS, Xhumari J, Ojeda A, Roesch LFW. Gut permeability among Astronauts during Space missions. LIFE SCIENCES IN SPACE RESEARCH 2024; 41:171-180. [PMID: 38670644 DOI: 10.1016/j.lssr.2024.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 02/02/2024] [Accepted: 03/13/2024] [Indexed: 04/28/2024]
Abstract
The space environment poses substantial challenges to human physiology, including potential disruptions in gastrointestinal health. Gut permeability has only recently become widely acknowledged for its potential to cause adverse effects on a systemic level, rendering it a critical factor to investigate in the context of spaceflight. Here, we propose that astronauts experience the onset of leaky gut during space missions supported by transcriptomic and metagenomic analysis of human and murine samples. A genetic map contributing to intestinal permeability was constructed from a systematic review of current literature. This was referenced against our re-analysis of three independent transcriptomic datasets which revealed significant changes in gene expression patterns associated with the gut barrier. Specifically, in astronauts during flight, we observed a substantial reduction in the expression genes that are crucial for intestinal barrier function, goblet cell development, gut microbiota modulation, and immune responses. Among rodent spaceflight studies, differential expression of cytokines, chemokines, and genes which regulate mucin production and post-translational modifications suggest a similar dysfunction of intestinal permeability. Metagenomic analysis of feces from two murine studies revealed a notable reduction probiotic, short chain fatty acid-producing bacteria and an increase in the Gram-negative pathogens, including Citrobacter rodentium, Enterobacter cloacea, Klebsiella aerogenes, and Proteus hauseri which promote LPS circulation, a recipe for barrier disruption and systemic inflammatory activation. These findings emphasize the critical need to understand the underlying mechanisms and develop interventions to maintain gastrointestinal health in space.
Collapse
Affiliation(s)
- Oluwamayowa S Akinsuyi
- Institute of Food and Agricultural Sciences, Department of Microbiology and Cell Science, University of Florida, Gainesville 32611, FL, USA
| | - Jessica Xhumari
- Institute of Food and Agricultural Sciences, Department of Microbiology and Cell Science, University of Florida, Gainesville 32611, FL, USA
| | - Amanda Ojeda
- Institute of Food and Agricultural Sciences, Department of Microbiology and Cell Science, University of Florida, Gainesville 32611, FL, USA
| | - Luiz F W Roesch
- Institute of Food and Agricultural Sciences, Department of Microbiology and Cell Science, University of Florida, Gainesville 32611, FL, USA.
| |
Collapse
|
13
|
Wang CX, Wang B, Sun JJ, Xiao CY, Ma H, Jia FY, Li HH. Circulating retinol and 25(OH)D contents and their association with symptoms in children with chronic tic disorders. Eur Child Adolesc Psychiatry 2024; 33:1017-1028. [PMID: 37166521 PMCID: PMC11032271 DOI: 10.1007/s00787-023-02226-4] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 05/04/2023] [Indexed: 05/12/2023]
Abstract
The present study measured serum levels of vitamin A (VA) and vitamin D (VD) in children with chronic tic disorders (CTD) and investigated their potential association with CTD and comorbidity of attention deficit hyperactivity disorder (ADHD) and the association of their co-insufficiencies or deficiencies with CTD symptoms. A total of 176 children (131 boys and 45 girls, median age of 9 years) with CTD were recruited as the CTD group. During the same period, 154 healthy children were selected as the healthy control (HC) cohort. Circulating retinol and 25-hydroxyvitamin D (25[OH]D) levels were measured for all participants using high-performance liquid chromatography (HPLC) and tandem mass spectrometry. The Yale Global Tic Severity Scale (YGTSS) was employed for the assessment of tic status and CTD impairment. The Swanson, Nolan, and Pelham Rating Scale (SNAP-IV) and the Children's Yale-Brown Obsessive-Compulsive Scale (CY-BOCS) were used to evaluate comorbidity symptoms. CTD pediatric participants exhibited markedly diminished circulating retinol and 25(OH)D levels compared to HCs. Moreover, VA and VD deficiencies and their co-insufficiencies/deficiencies were more prevalent in CTD participants than HCs. Circulating 25(OH)D levels were inversely proportional to the YGTSS motor tic scores. YGTSS scores in CTD children with only VA or VD insufficiency or deficiency or with VA and VD co-insufficiency/deficiency did not differ from those in CTD children with normal VA and VD. CTD children with comorbid ADHD displayed reduced circulating retinol and 25(OH)D concentrations and elevated prevalence of VD deficiency compared to CTD participants without comorbid ADHD. Lower serum retinol content was intricately linked to the presence of elevated CTD and comorbid ADHD. VA and VD deficiencies and their co-insufficiencies/deficiencies were markedly enhanced in CTD pediatric participants compared to HCs. Lower VA concentration was linked to the presence of enhanced CTD and comorbid ADHD. Therefore, children with CTD, especially with comorbid ADHD, may be at a higher risk of VA or VD deficiency, which may prompt the clinicians to consider whether blood tests for VA and VD in CTD children would be helpful for clinical care.
Collapse
Affiliation(s)
- Cheng-Xin Wang
- Department of Developmental and Behavioral Pediatrics, The First Hospital of Jilin University, Changchun, 130021, Jilin, China
- Department of Laboratory Medicine, The First Hospital of Jilin University, Changchun, 130021, China
| | - Bing Wang
- Department of Developmental and Behavioral Pediatrics, The First Hospital of Jilin University, Changchun, 130021, Jilin, China
| | - Jian-Jian Sun
- Department of Developmental and Behavioral Pediatrics, The First Hospital of Jilin University, Changchun, 130021, Jilin, China
| | - Chun-Ying Xiao
- Department of Developmental and Behavioral Pediatrics, The First Hospital of Jilin University, Changchun, 130021, Jilin, China
| | - Huan Ma
- Department of Developmental and Behavioral Pediatrics, The First Hospital of Jilin University, Changchun, 130021, Jilin, China
| | - Fei-Yong Jia
- Department of Developmental and Behavioral Pediatrics, The First Hospital of Jilin University, Changchun, 130021, Jilin, China
| | - Hong-Hua Li
- Department of Developmental and Behavioral Pediatrics, The First Hospital of Jilin University, Changchun, 130021, Jilin, China.
- School of Public Health, Jilin University, Changchun, Jilin, China.
| |
Collapse
|
14
|
Gaudino SJ, Singh A, Huang H, Padiadpu J, Jean-Pierre M, Kempen C, Bahadur T, Shiomitsu K, Blumberg R, Shroyer KR, Beyaz S, Shulzhenko N, Morgun A, Kumar P. Intestinal IL-22RA1 signaling regulates intrinsic and systemic lipid and glucose metabolism to alleviate obesity-associated disorders. Nat Commun 2024; 15:1597. [PMID: 38383607 PMCID: PMC10881576 DOI: 10.1038/s41467-024-45568-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 01/26/2024] [Indexed: 02/23/2024] Open
Abstract
IL-22 is critical for ameliorating obesity-induced metabolic disorders. However, it is unknown where IL-22 acts to mediate these outcomes. Here we examine the importance of tissue-specific IL-22RA1 signaling in mediating long-term high fat diet (HFD) driven metabolic disorders. To do so, we generated intestinal epithelium-, liver-, and white adipose tissue (WAT)-specific Il22ra1 knockout and littermate control mice. Intestinal epithelium- and liver-specific IL-22RA1 signaling upregulated systemic glucose metabolism. Intestinal IL-22RA1 signaling also mediated liver and WAT metabolism in a microbiota-dependent manner. We identified an association between Oscillibacter and elevated WAT inflammation, likely induced by Mmp12 expressing macrophages. Mechanistically, transcription of intestinal lipid metabolism genes is regulated by IL-22 and potentially IL-22-induced IL-18. Lastly, we show that Paneth cell-specific IL-22RA1 signaling, in part, mediates systemic glucose metabolism after HFD. Overall, these results elucidate a key role of intestinal epithelium-specific IL-22RA1 signaling in regulating intestinal metabolism and alleviating systemic obesity-associated disorders.
Collapse
Affiliation(s)
- Stephen J Gaudino
- Department of Microbiology and Immunology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Ankita Singh
- Department of Microbiology and Immunology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Huakang Huang
- Department of Microbiology and Immunology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Jyothi Padiadpu
- College of Pharmacy, Oregon State University, Corvallis, OR, USA
| | - Makheni Jean-Pierre
- Department of Microbiology and Immunology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Cody Kempen
- Department of Microbiology and Immunology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Tej Bahadur
- Department of Microbiology and Immunology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Kiyoshi Shiomitsu
- Department of Microbiology and Immunology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Richard Blumberg
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Kenneth R Shroyer
- Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Semir Beyaz
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA
| | - Natalia Shulzhenko
- Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR, USA
| | - Andrey Morgun
- College of Pharmacy, Oregon State University, Corvallis, OR, USA
| | - Pawan Kumar
- Department of Microbiology and Immunology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA.
| |
Collapse
|
15
|
Wu-Woods NJ, Barlow JT, Trigodet F, Shaw DG, Romano AE, Jabri B, Eren AM, Ismagilov RF. Microbial-enrichment method enables high-throughput metagenomic characterization from host-rich samples. Nat Methods 2023; 20:1672-1682. [PMID: 37828152 PMCID: PMC10885704 DOI: 10.1038/s41592-023-02025-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 08/27/2023] [Indexed: 10/14/2023]
Abstract
Host-microbe interactions have been linked to health and disease states through the use of microbial taxonomic profiling, mostly via 16S ribosomal RNA gene sequencing. However, many mechanistic insights remain elusive, in part because studying the genomes of microbes associated with mammalian tissue is difficult due to the high ratio of host to microbial DNA in such samples. Here we describe a microbial-enrichment method (MEM), which we demonstrate on a wide range of sample types, including saliva, stool, intestinal scrapings, and intestinal mucosal biopsies. MEM enabled high-throughput characterization of microbial metagenomes from human intestinal biopsies by reducing host DNA more than 1,000-fold with minimal microbial community changes (roughly 90% of taxa had no significant differences between MEM-treated and untreated control groups). Shotgun sequencing of MEM-treated human intestinal biopsies enabled characterization of both high- and low-abundance microbial taxa, pathways and genes longitudinally along the gastrointestinal tract. We report the construction of metagenome-assembled genomes directly from human intestinal biopsies for bacteria and archaea at relative abundances as low as 1%. Analysis of metagenome-assembled genomes reveals distinct subpopulation structures between the small and large intestine for some taxa. MEM opens a path for the microbiome field to acquire deeper insights into host-microbe interactions by enabling in-depth characterization of host-tissue-associated microbial communities.
Collapse
Affiliation(s)
- Natalie J Wu-Woods
- Biology and Bioengineering, California Institute of Technology, Pasadena, CA, USA
| | - Jacob T Barlow
- Biology and Bioengineering, California Institute of Technology, Pasadena, CA, USA
| | - Florian Trigodet
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Dustin G Shaw
- Department of Medicine, The University of Chicago, Chicago, IL, USA
- Committee on Immunology, The University of Chicago, Chicago, IL, USA
- Department of Pathology, The University of Chicago, Chicago, IL, USA
| | - Anna E Romano
- Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, USA
| | - Bana Jabri
- Department of Medicine, The University of Chicago, Chicago, IL, USA
- Committee on Immunology, The University of Chicago, Chicago, IL, USA
- Department of Pathology, The University of Chicago, Chicago, IL, USA
| | - A Murat Eren
- Bay Paul Center, Marine Biological Laboratory, Woods Hole, MA, USA
- Institute for Chemistry and Biology of the Marine Environment, University of Oldenburg, Oldenburg, Germany
- Alfred-Wegener-Institute for Marine and Polar Research, Bremerhaven, Germany
- Helmholtz Institute for Functional Marine Biodiversity, Oldenburg, Germany
| | - Rustem F Ismagilov
- Biology and Bioengineering, California Institute of Technology, Pasadena, CA, USA.
- Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, USA.
| |
Collapse
|
16
|
Calderon RM, Golczak M, Paik J, Blaner WS. Dietary Vitamin A Affects the Function of Incretin-Producing Enteroendocrine Cells in Male Mice Fed a High-Fat Diet. J Nutr 2023; 153:2901-2914. [PMID: 37648113 PMCID: PMC10613727 DOI: 10.1016/j.tjnut.2023.08.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 08/12/2023] [Accepted: 08/24/2023] [Indexed: 09/01/2023] Open
Abstract
BACKGROUND Retinol-binding protein 2 (RBP2) is an intracellular carrier for vitamin A in the absorptive enterocytes. Mice lacking RBP2 (Rbp2-/-) display an unexpected phenotype of obesity, glucose intolerance, and elevated glucose-dependent insulinotropic polypeptide (GIP) levels. GIP and glucagon-like peptide 1 (GLP-1) are incretin hormones secreted by enteroendocrine cells (EECs). We recently demonstrated the presence of RBP2 and other retinoid-related proteins in EECs. OBJECTIVES Given RBP2's role in intracellular retinoid trafficking, we aimed to evaluate whether dietary vitamin A affects incretin-secreting cell function and gene expression. METHODS Male Rbp2-/- mice and sex- and age-matched controls (n = 6-9) were fed a high-fat diet (HFD) for 18 wk containing normal (VAN, 4000 IU/kg of diet) or low (VAL, 25% of normal) vitamin A concentrations. Body weight was recorded biweekly. Plasma GIP and GLP-1 levels were obtained fasting and 30 min after an oral fat gavage at week 16. Glucose tolerance tests were also performed. Mice were killed at week 18, and blood and tissue samples were obtained. RESULTS Rbp2-/- mice displayed greater weight gain on the VAN compared with the VAL diet from week 7 of the intervention (P ≤ 0.01). Stimulated GIP levels were elevated in Rbp2-/- mice compared with their controls fed the VAN diet (P = 0.02), whereas their GIP response was lower when fed the VAL diet (P = 0.03). Although no differences in GLP-1 levels were observed in the VAN diet group, a lower GLP-1 response was seen in Rbp2-/- mice fed the VAL diet (P = 0.02). Changes in incretin gene expression and that of other genes associated with EEC lineage and function were consistent with these observations. Circulating and hepatic retinoid levels revealed no systemic vitamin A deficiency across dietary groups. CONCLUSIONS Our data support a role for RBP2 and dietary vitamin A in incretin secretion and gene expression in mice fed a HFD.
Collapse
Affiliation(s)
- Rossana M Calderon
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, United States.
| | - Marcin Golczak
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH, United States; Cleveland Center for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Jisun Paik
- Department of Comparative Medicine, University of Washington, Seattle, WA, United States
| | - William S Blaner
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, United States
| |
Collapse
|
17
|
Howley K, Berthelette A, Ceglia S, Kang J, Reboldi A. Embryonic type 3 innate lymphoid cells sense maternal dietary cholesterol to control local Peyer's patch development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.19.533339. [PMID: 36993524 PMCID: PMC10055282 DOI: 10.1101/2023.03.19.533339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
Lymphoid tissue inducer (LTi) cells develop during intrauterine life and rely on developmental programs to initiate the organogenesis of secondary lymphoid organs (SLOs). This evolutionary conserved process endows the fetus with the ability to orchestrate the immune response after birth and to react to the triggers present in the environment. While it is established that LTi function can be shaped by maternal-derived cues and is critical to prepare the neonate with a functional scaffold to mount immune response, the cellular mechanisms that control anatomically distinct SLO organogenesis remain unclear. We discovered that LTi cells forming Peyer's patches, gut-specific SLOs, require the coordinated action of two migratory G protein coupled receptors (GPCR) GPR183 and CCR6. These two GPCRs are uniformly expressed on LTi cells across SLOs, but their deficiency specifically impacts Peyer's patch formation, even when restricted to fetal window. The unique CCR6 ligand is CCL20, while the ligand for GPR183 is the cholesterol metabolite 7α,25-Dihydroxycholesterol (7α,25-HC), whose production is controlled by the enzyme cholesterol 25-hydroxylase (CH25H). We identified a fetal stromal cell subset that expresses CH25H and attracts LTi cells in the nascent Peyer's patch anlagen. GPR183 ligand concentration can be modulated by the cholesterol content in the maternal diet and impacts LTi cell maturation in vitro and in vivo, highlighting a link between maternal nutrients and intestinal SLO organogenesis. Our findings revealed that in the fetal intestine, cholesterol metabolite sensing by GPR183 in LTi cells for Peyer's patch formation is dominant in the duodenum, the site of cholesterol absorption in the adult. This anatomic requirement suggests that embryonic, long-lived non-hematopoietic cells might exploit adult metabolic functions to ensure highly specialized SLO development in utero.
Collapse
Affiliation(s)
- Kelsey Howley
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA
| | - Alyssa Berthelette
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA
| | - Simona Ceglia
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA
| | - Joonsoo Kang
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA
| | - Andrea Reboldi
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA
| |
Collapse
|