1
|
Sanaei E, de Roode JC. The role of host plants in driving pathogen susceptibility in insects through chemicals, immune responses and microbiota. Biol Rev Camb Philos Soc 2025; 100:1347-1364. [PMID: 39916634 DOI: 10.1111/brv.70003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 01/24/2025] [Accepted: 01/29/2025] [Indexed: 05/31/2025]
Abstract
In this comprehensive exploration, we delve into the pivotal role of host plants in shaping the intricate interactions between herbivorous insects and their pathogens. Recent decades have seen a surge in studies that demonstrate that host plants are crucial drivers of the interactions between insects and pathogens, providing novel insights into the direct and indirect interactions that shape tri-trophic interactions. These studies have built on a wide range of pathogens, from viruses to bacteria, and from protozoans to fungi. We summarise these studies, and discuss the mechanisms of plant-mediated insect resistance to infection, ranging from the toxicity of plant chemicals to pathogens to enhancement of anti-pathogen immune responses, and modulation of the insect's microbiome. Although we provide evidence for the roles of all these mechanisms, we also point out that the majority of existing studies are phenomenological, describing patterns without addressing the underlying mechanisms. To further our understanding of these tri-trophic interactions, we therefore urge researchers to design their studies to enable them specifically to distinguish the mechanisms by which plants affect insect susceptibility to pathogens.
Collapse
Affiliation(s)
- Ehsan Sanaei
- Biology Department, Emory University, 1510 Clifton Road, Atlanta, Georgia, 30322, USA
| | - Jacobus C de Roode
- Biology Department, Emory University, 1510 Clifton Road, Atlanta, Georgia, 30322, USA
| |
Collapse
|
2
|
Kodio A, Coulibaly D, Doumbo S, Konaté S, Koné AK, Dama S, Niangaly A, Tall ML, Konaté AM, L'Ollivier C, Levasseur A, Bittar F, Djimdé A, Doumbo OK, Raoult D, Thera MA, Ranque S. Gut microbiota influences Plasmodium falciparum malaria susceptibility. New Microbes New Infect 2025; 65:101586. [PMID: 40290900 PMCID: PMC12032372 DOI: 10.1016/j.nmni.2025.101586] [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: 08/25/2022] [Revised: 04/10/2025] [Accepted: 04/14/2025] [Indexed: 04/30/2025] Open
Abstract
Background The gut microbiota has recently been associated with malaria susceptibility/resistance in animal models and humans. This study aimed to assess its influence on malaria attack and Plasmodium parasitemia in children living in a malaria-endemic area of Mali. Methods Healthy children were enrolled in a 16-month cohort study in Bandiagara. Their gut bacteria and fungi community structures were characterized via 16S and ITS metabarcoding at enrolment. Clinicians monitored malaria attacks. Asymptomatic Plasmodium carriage was assessed by real-time polymerase chain reaction. Results Of the 300 children, 107 (36 %) had at least one malaria attack, and 82 (27 %) had at least one episode of asymptomatic Plasmodium parasitemia. The gut bacterial community structure, but not the fungal community, was associated with susceptibility/resistance to both malaria attacks and asymptomatic P. falciparum parasitemia. Higher gut bacteria richness was independently associated with susceptibility to both asymptomatic parasitemia episodes and malaria attacks. 17 bacteria, and 7 fungi were associated with susceptibility to malaria attacks, and 8 bacteria, and 3 fungi were associated with resistance. 15 bacteria and 13 fungi were associated with susceptibility to asymptomatic Plasmodium parasitemia episodes, and 19 bacteria and 3 fungi were associated with resistance. Conclusion Further studies are needed to confirm these findings, which point the way to strategies aimed at reducing the risk of malaria by modulating gut microbiota components in at-risk populations.
Collapse
Affiliation(s)
- Aly Kodio
- Aix Marseille Université, Service de Santé des Armées, RITMES, 19-21 Boulevard Jean Moulin, 13005, Marseille, France
- IHU Méditerranée Infection, 19-21 Bd Jean Moulin, 13005, Marseille, France
- Malaria Research and Training Centre-International Center for Excellence in Research (MRTC-ICER), Department of Epidemiology of Parasitic Diseases, Faculty of Medicine and Dentistry, Université des Sciences, des Techniques et des Technologies de Bamako, Point G, BP 1805, Bamako, Mali
| | - Drissa Coulibaly
- Malaria Research and Training Centre-International Center for Excellence in Research (MRTC-ICER), Department of Epidemiology of Parasitic Diseases, Faculty of Medicine and Dentistry, Université des Sciences, des Techniques et des Technologies de Bamako, Point G, BP 1805, Bamako, Mali
| | - Safiatou Doumbo
- Malaria Research and Training Centre-International Center for Excellence in Research (MRTC-ICER), Department of Epidemiology of Parasitic Diseases, Faculty of Medicine and Dentistry, Université des Sciences, des Techniques et des Technologies de Bamako, Point G, BP 1805, Bamako, Mali
| | - Salimata Konaté
- Malaria Research and Training Centre-International Center for Excellence in Research (MRTC-ICER), Department of Epidemiology of Parasitic Diseases, Faculty of Medicine and Dentistry, Université des Sciences, des Techniques et des Technologies de Bamako, Point G, BP 1805, Bamako, Mali
| | - Abdoulaye Kassoum Koné
- Malaria Research and Training Centre-International Center for Excellence in Research (MRTC-ICER), Department of Epidemiology of Parasitic Diseases, Faculty of Medicine and Dentistry, Université des Sciences, des Techniques et des Technologies de Bamako, Point G, BP 1805, Bamako, Mali
| | - Souleymane Dama
- Malaria Research and Training Centre-International Center for Excellence in Research (MRTC-ICER), Department of Epidemiology of Parasitic Diseases, Faculty of Medicine and Dentistry, Université des Sciences, des Techniques et des Technologies de Bamako, Point G, BP 1805, Bamako, Mali
| | - Amadou Niangaly
- Malaria Research and Training Centre-International Center for Excellence in Research (MRTC-ICER), Department of Epidemiology of Parasitic Diseases, Faculty of Medicine and Dentistry, Université des Sciences, des Techniques et des Technologies de Bamako, Point G, BP 1805, Bamako, Mali
| | - Mamadou Lamine Tall
- IHU Méditerranée Infection, 19-21 Bd Jean Moulin, 13005, Marseille, France
- Aix Marseille Université, Assistance Publique-Hôpitaux de Marseille, MEPHI: Microbes, Evolution, Phylogénie et Infection, 19-21 Boulevard Jean Moulin, 13005, Marseille, France
| | - Ahmed Mohamed Konaté
- Malaria Research and Training Centre-International Center for Excellence in Research (MRTC-ICER), Department of Epidemiology of Parasitic Diseases, Faculty of Medicine and Dentistry, Université des Sciences, des Techniques et des Technologies de Bamako, Point G, BP 1805, Bamako, Mali
| | - Coralie L'Ollivier
- Aix Marseille Université, Service de Santé des Armées, RITMES, 19-21 Boulevard Jean Moulin, 13005, Marseille, France
- IHU Méditerranée Infection, 19-21 Bd Jean Moulin, 13005, Marseille, France
| | - A. Levasseur
- IHU Méditerranée Infection, 19-21 Bd Jean Moulin, 13005, Marseille, France
- Aix Marseille Université, Assistance Publique-Hôpitaux de Marseille, MEPHI: Microbes, Evolution, Phylogénie et Infection, 19-21 Boulevard Jean Moulin, 13005, Marseille, France
| | - Fadi Bittar
- IHU Méditerranée Infection, 19-21 Bd Jean Moulin, 13005, Marseille, France
- Aix Marseille Université, Assistance Publique-Hôpitaux de Marseille, MEPHI: Microbes, Evolution, Phylogénie et Infection, 19-21 Boulevard Jean Moulin, 13005, Marseille, France
| | - Abdoulaye Djimdé
- Malaria Research and Training Centre-International Center for Excellence in Research (MRTC-ICER), Department of Epidemiology of Parasitic Diseases, Faculty of Medicine and Dentistry, Université des Sciences, des Techniques et des Technologies de Bamako, Point G, BP 1805, Bamako, Mali
| | - Ogobara K. Doumbo
- Malaria Research and Training Centre-International Center for Excellence in Research (MRTC-ICER), Department of Epidemiology of Parasitic Diseases, Faculty of Medicine and Dentistry, Université des Sciences, des Techniques et des Technologies de Bamako, Point G, BP 1805, Bamako, Mali
| | - Didier Raoult
- IHU Méditerranée Infection, 19-21 Bd Jean Moulin, 13005, Marseille, France
- Aix Marseille Université, Assistance Publique-Hôpitaux de Marseille, MEPHI: Microbes, Evolution, Phylogénie et Infection, 19-21 Boulevard Jean Moulin, 13005, Marseille, France
| | - Mahamadou Ali Thera
- Malaria Research and Training Centre-International Center for Excellence in Research (MRTC-ICER), Department of Epidemiology of Parasitic Diseases, Faculty of Medicine and Dentistry, Université des Sciences, des Techniques et des Technologies de Bamako, Point G, BP 1805, Bamako, Mali
| | - Stéphane Ranque
- Aix Marseille Université, Service de Santé des Armées, RITMES, 19-21 Boulevard Jean Moulin, 13005, Marseille, France
- IHU Méditerranée Infection, 19-21 Bd Jean Moulin, 13005, Marseille, France
| |
Collapse
|
3
|
Diallo D, Sun S, Somboro AM, Baya B, Koné A, Diarra B, Nantoumé M, Koloma I, Diakite M, Holl J, Maiga AI, Seydi M, Theron G, Hou L, Fodor A, Maiga M. Metabolic and immune consequences of antibiotic related microbiome alterations during first-line tuberculosis treatment in Bamako, Mali. Front Immunol 2025; 16:1561459. [PMID: 40438118 PMCID: PMC12116645 DOI: 10.3389/fimmu.2025.1561459] [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: 01/15/2025] [Accepted: 04/23/2025] [Indexed: 06/01/2025] Open
Abstract
Background Individuals with a history of tuberculosis (TB) treatment are at a higher risk of experiencing a recurrent episode of the disease. Previous cross-sectional studies identified a connection between dysbiosis (alterations) in the gut microbiota composition and the administration of first-line TB antibiotics. However, these studies have not successfully elucidated this dysbiosis's resulting metabolic and immune consequences. Methods In a longitudinal assessment, we studied the antituberculosis drug-related changes in the gut microbiota's composition and the resulting functional consequences. Sputum for TB culture, peripheral blood for metabolomics and cytokines analysis, and stool for shotgun metagenomics were collected from TB participants at Month-0, Month-2, Month-6 of treatment, and 9 Months after treatment (Month-15). Healthy controls were sampled at Month-0 and Month-6. Findings We found notable differences in gut microbiota between individuals with TB and healthy controls. While gut microbiota tended to resemble healthy controls at the end of TB treatment, significant differences for many taxa persisted up to Month-15. Concurrently, disturbances in plasma metabolites, including tryptophan, tricarboxylic acids, and cytokine levels were observed. Certain fatty acids associated with inflammation pathways negatively correlated with the abundance of several taxa. Conclusion We observed alterations in the gut microbiota composition and function during treatment and at Month-15. Numerous changes in bacterial taxa abundances and inflammation-linked metabolites did not reverse at Month-15. This study suggests potential influences of anti-TB drugs and the gut microbiome on the disease outcome, response to treatment, and resistance to future TB infections.
Collapse
Affiliation(s)
- Dramane Diallo
- University Clinical Research Center (UCRC), Bamako, Mali
| | - Shan Sun
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, NC, United States
| | - Anou M. Somboro
- Antimicrobial Research Unit, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
- University of Sciences, Techniques, and Technologies of Bamako (USTTB), Bamako, Mali
| | - Bocar Baya
- University Clinical Research Center (UCRC), Bamako, Mali
| | - Amadou Koné
- University Clinical Research Center (UCRC), Bamako, Mali
- University of Sciences, Techniques, and Technologies of Bamako (USTTB), Bamako, Mali
| | - Bassirou Diarra
- University Clinical Research Center (UCRC), Bamako, Mali
- University of Sciences, Techniques, and Technologies of Bamako (USTTB), Bamako, Mali
| | | | - Isaac Koloma
- University Clinical Research Center (UCRC), Bamako, Mali
| | - Mahamadou Diakite
- University Clinical Research Center (UCRC), Bamako, Mali
- University of Sciences, Techniques, and Technologies of Bamako (USTTB), Bamako, Mali
| | - Jane Holl
- Department of Neurology and Center for Healthcare Delivery Science and Innovation, University of Chicago, Chicago, IL, United States
| | | | - Moussa Seydi
- Service des Maladies Infectieuses et Tropicales, Fann University Hospital Center, Dakar, Senegal
| | - Grant Theron
- Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis, Stellenbosch University, Cape Town, South Africa
| | - Lifang Hou
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Anthony Fodor
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, NC, United States
| | - Mamoudou Maiga
- University of Sciences, Techniques, and Technologies of Bamako (USTTB), Bamako, Mali
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
4
|
Wanelik KM, Begon M, Bradley JE, Fenn J, Jackson JA, Paterson S. Superspreaders have lower gut microbial alpha-diversity and distinct gut microbial composition in a natural rodent population. Anim Microbiome 2025; 7:42. [PMID: 40325481 PMCID: PMC12053855 DOI: 10.1186/s42523-025-00411-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Accepted: 04/14/2025] [Indexed: 05/07/2025] Open
Abstract
The microbiome is well known to drive variation in host states (e.g. behaviour, immunity) that would be expected to modulate the spread of infectious disease-but the role of microbiotal interactions in promoting superspreading is poorly understood. Superspreaders are individuals with a strongly disproportionate contribution to pathogen transmission, and come in two forms. Supershedders transmit infection to more individuals because they shed higher levels of pathogen. Supercontacters transmit infection to more individuals because they have larger numbers of social contacts. We explore associations between the gut microbiota and these two forms of superspreading in a wild rodent model-Bartonella spp. bacteraemia in the field vole (Microtus agrestis). We find evidence that individuals fall into distinct shedding and contacting clusters, and that higher-contacters have lower and more variable gut microbial alpha-diversity than lower-contacters. We also show evidence that both higher-shedders and higher-contacters have distinct gut microbial composition and identify OTUs that are differentially abundant in the gut microbiota of these two classes of individuals when compared to lower-shedders and lower-contacters respectively. We find that the Muribaculaceae are associated with differences in both shedding and contacting, and discuss potential mechanisms by which they may be acting on these host traits.
Collapse
Affiliation(s)
- Klara M Wanelik
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK.
- School of Biosciences, University of Surrey, Guildford, UK.
| | - Mike Begon
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | | | - Jonathan Fenn
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Joseph A Jackson
- School of Science, Engineering and Environment, University of Salford, Salford, UK
| | - Steve Paterson
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| |
Collapse
|
5
|
Prenen F, Pollenus E, Meers H, Knoops S, Sadler R, Deckers M, Mills EL, Van den Steen PE. Itaconate Has Limited Protective Effects in Experimental Malaria Models. Eur J Immunol 2025; 55:e202451595. [PMID: 40346757 DOI: 10.1002/eji.202451595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 04/24/2025] [Accepted: 04/25/2025] [Indexed: 05/12/2025]
Abstract
In severe malaria, dysregulated metabolism and excessive inflammatory responses contribute to fatal outcomes. Therapeutic strategies that address both metabolic and inflammatory balances are thus required. Itaconate, a metabolite produced by aconitate decarboxylase 1 (ACOD1), is a potent inhibitor of both inflammation and glycolysis with protective effects in various inflammatory diseases. Although elevated itaconate levels have been observed in Plasmodium-infected individuals, its role in malaria is still poorly understood, making further investigation essential for assessing its therapeutic potential. We investigated the role of itaconate in both severe and mild malaria using Plasmodium berghei NK65 (PbNK65) and Plasmodium chabaudi AS (PcAS) models, respectively. Using 13C-tracer metabolomics, we detected increased itaconate levels in various organs during infection and identified inflammatory monocytes as the source of this production. Nevertheless, ACOD1 knockout mice displayed no significant changes in phenotype after PbNK65 infection, and treatment of PbNK65-infected mice with 4-octyl itaconate did not affect disease severity either. However, in the PcAS model, ACOD1 deficiency worsened the disease, as indicated by increased weight loss, higher clinical scores, and elevated parasitemia. Therefore, in contrast to the findings in recent literature, our study shows that itaconate does not contribute to susceptibility, but rather provides limited protection to malaria.
Collapse
Affiliation(s)
- Fran Prenen
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Emilie Pollenus
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Hanne Meers
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Sofie Knoops
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Rebecca Sadler
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Margot Deckers
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
- Laboratory of Host-Pathogen Interactions, Department of Pathology, University of Utah, Salt Lake City, USA
| | - Evanna L Mills
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Immunology, Harvard Medical School, Boston, Massachusetts, USA
| | - Philippe E Van den Steen
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| |
Collapse
|
6
|
Rehermann B, Graham AL, Masopust D, Hamilton SE. Integrating natural commensals and pathogens into preclinical mouse models. Nat Rev Immunol 2025; 25:385-397. [PMID: 39562646 DOI: 10.1038/s41577-024-01108-3] [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: 10/18/2024] [Indexed: 11/21/2024]
Abstract
Fundamental discoveries in many aspects of mammalian physiology have been made using laboratory mice as research models. These studies have been facilitated by the genetic tractability and inbreeding of such mice, the large set of immunological reagents that are available, and the establishment of environmentally controlled, high-throughput facilities. Such facilities typically include barriers to keep the mouse colonies free of pathogens and the frequent re-derivation of the mice severely limits their commensal flora. Because humans have co-evolved with microorganisms and are exposed to a variety of pathogens, a growing community of researchers posits that preclinical disease research can be improved by studying mice in the context of the microbiota and pathogens that they would encounter in the natural world. Here, we provide a perspective of how these different approaches can be combined and integrated to improve existing mouse models to enhance our understanding of disease mechanisms and develop new therapies for humans. We also propose that the term 'mice with natural microbiota' is more appropriate for describing these models than existing terms such as 'dirty mice'.
Collapse
Affiliation(s)
- Barbara Rehermann
- Immunology Section, Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA.
| | - Andrea L Graham
- Department of Ecology & Evolutionary Biology, Princeton University, Princeton, NJ, USA
| | - David Masopust
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Sara E Hamilton
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
7
|
Calôba C, Sturtz AJ, Lyons TA, John L, Ramachandran A, Minns AM, Cannon AM, Whalley JP, Watts TH, Kaplan MH, Lindner SE, Vijay R. Systemic 4-1BB stimulation augments extrafollicular memory B cell formation and recall responses during Plasmodium infection. Cell Rep 2025; 44:115528. [PMID: 40215168 PMCID: PMC12079783 DOI: 10.1016/j.celrep.2025.115528] [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: 11/26/2024] [Revised: 02/21/2025] [Accepted: 03/15/2025] [Indexed: 04/26/2025] Open
Abstract
T-dependent germinal center (GC) output, comprising plasma cells and memory B cells (MBCs), is crucial for clearance of Plasmodium infection and protection against reinfection. In this study, we examine the effect of an agonistic antibody targeting 4-1BB (CD137) during experimental malaria. We show that exogenous 4-1BB stimulation, despite delaying the effector GC response, surprisingly enhances humoral memory recall and protection from reinfection. Single-cell RNA and assay for transposase-accessible chromatin (ATAC) sequencing of MBCs from mice receiving 4-1BB stimulation reveal populations with distinct transcriptional signatures and a chromatin landscape indicative of superior recall and proliferative potential. Importantly, our results indicate that the effects of 4-1BB stimulation are dependent on interleukin (IL)-9R signaling in B cells but independent of parasite load during primary infection. Our study proposes an immunomodulatory approach to enhance the quality of the MBC pool, providing superior protection during infection and vaccination, particularly in the context of malaria.
Collapse
Affiliation(s)
- Carolina Calôba
- Discipline of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA; School of Graduate and Postdoctoral Studies, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA; Center for Cancer Biology, Immunology and Infection, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Alexandria J Sturtz
- Division of Biology and Biomedical Sciences, Washington University, St. Louis, Missouri, USA
| | - Taylor A Lyons
- Center for Cancer Biology, Immunology and Infection, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Lijo John
- Discipline of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA; Deparment of Veterinary Biochemistry, Kerala Veterinary and Animal Sciences University, Thrissur, Kerala, India
| | - Akshaya Ramachandran
- Discipline of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA; School of Graduate and Postdoctoral Studies, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Allen M Minns
- Department of Biochemistry and Molecular Biology, Huck Center for Malaria Research, Pennsylvania State University, University Park, PA, USA
| | - Anthony M Cannon
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Justin P Whalley
- Discipline of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA; Center for Cancer Biology, Immunology and Infection, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Tania H Watts
- Department of Immunology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Mark H Kaplan
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Scott E Lindner
- Deparment of Veterinary Biochemistry, Kerala Veterinary and Animal Sciences University, Thrissur, Kerala, India
| | - Rahul Vijay
- Discipline of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA; Center for Cancer Biology, Immunology and Infection, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA.
| |
Collapse
|
8
|
Fusco EM, Bower L, Polidoro R, Minns AM, Lindner SE, Schmidt NW. Microbiome-mediated modulation of immune memory to P. yoelii affects the resistance to secondary cerebral malaria challenge. Immunohorizons 2025; 9:vlaf009. [PMID: 40193560 PMCID: PMC12086675 DOI: 10.1093/immhor/vlaf009] [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: 09/19/2024] [Accepted: 01/29/2025] [Indexed: 04/09/2025] Open
Abstract
Malaria is caused by protozoan parasites in the genus Plasmodium. Over time individuals slowly develop clinical immunity to malaria, but this process occurs at variable rates, and the mechanism of protection is not fully understood. We have recently demonstrated that in genetically identical C57BL/6N mice, gut microbiota composition dramatically impacts the quality of the humoral immune response to Plasmodium yoelii and subsequent protection against a lethal secondary challenge with Plasmodium berghei ANKA in C57BL/6N mice. Here, we utilize this genetically identical, gut microbiome-dependent model to investigate how the gut microbiota modulate immunological memory, hypothesizing that the gut microbiome impacts the formation and functionality of immune memory. In support of this hypothesis, P. yoelii hyperparasitemia-resistant C57BL/6N mice exhibit increased protection against P. berghei ANKA-induced experimental cerebral malaria (ECM) compared to P. yoelii hyperparasitemia-susceptible C57BL/6N mice. Despite differences in protection against ECM, P. yoelii-resistant and -susceptible mice accumulate similar numbers of memory B cells (MBCs) and memory T cells. Following challenge with P. berghei ANKA, P. yoelii-resistant mice generated more rapid germinal center reactions; however, P. yoelii-resistant and -susceptible mice had similar titers of P. yoelii- and P. berghei-specific antibodies. In contrast, P. yoelii-resistant mice had an increased number of regulatory T cells in response to secondary challenge with P. berghei ANKA, which may dampen the immune-mediated breakdown of the blood-brain barrier and susceptibility to P. berghei-induced ECM. These findings demonstrate the ability of the gut microbiome to shape immune memory and the potential to enhance resistance to severe malaria outcomes.
Collapse
Affiliation(s)
- Elizabeth M Fusco
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Layne Bower
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, United States
- Ryan White Center for Pediatric Infectious Diseases and Global Health, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Rafael Polidoro
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, United States
- Ryan White Center for Pediatric Infectious Diseases and Global Health, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Allen M Minns
- The Huck Institutes of Life Sciences, Pennsylvania State University, University Park, PA, United States
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA, United States
- The Huck Center for Malaria Research, University Park, PA, United States
| | - Scott E Lindner
- The Huck Institutes of Life Sciences, Pennsylvania State University, University Park, PA, United States
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA, United States
- The Huck Center for Malaria Research, University Park, PA, United States
| | - Nathan W Schmidt
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, United States
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, United States
- Ryan White Center for Pediatric Infectious Diseases and Global Health, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
9
|
Zhang B, Tian M, Qiu Y, Wu J, Cui C, Liu S, Hou J, Tian C, Wang L, Gao K, Jiang Z, Yang X. Glucuronolactone Restores the Intestinal Barrier and Redox Balance Partly Through the Nrf2/Akt/FOXO1 Pathway to Alleviate Weaning Stress-Induced Intestinal Dysfunction in Piglets. Antioxidants (Basel) 2025; 14:352. [PMID: 40227425 PMCID: PMC11939252 DOI: 10.3390/antiox14030352] [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: 02/06/2025] [Revised: 03/07/2025] [Accepted: 03/12/2025] [Indexed: 04/15/2025] Open
Abstract
(1) Background: Glucuronolactone (GLU) is a glucose metabolite with antioxidant activity. At present, the exact role of it in regulating the intestinal health of piglets under weaning stress is not clear. The purpose of this study is to investigate the effects of GLU on the growth performance and intestinal health of piglets under weaning stress and to explore potential mechanisms. (2) Methods: Twenty-four weaned piglets were randomly assigned into two groups, with one group receiving a basal diet and the other group receiving an experimental diet supplemented with 200 mg/kg of GLU. (3) Results: GLU increased the ADG, ADFI, and final body weight of piglets, while reducing the diarrhea rate. Mechanistically, GLU alleviates weaning stress-induced intestinal oxidative stress and inflammatory responses in piglets partly through activating the Nrf2-Akt signaling pathway to suppress the transcriptional activity of FOXO1, while also inhibiting the activation of the TLR4-MAPK signaling pathway to reduce the secretion of pro-inflammatory cytokines. Moreover, GLU increased the relative abundance of Lactobacillus reuteri in the ileum of piglets and improved the composition of the gut microbiota. (4) Conclusions: GLU reduced inflammation and oxidative stress through the Nrf2/Akt/FOXO1 signaling pathway and improved intestinal health, resulting in improved growth performance of the piglets.
Collapse
Affiliation(s)
- Beibei Zhang
- College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou 510640, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangzhou 510640, China
- Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, China
| | - Min Tian
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou 510640, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangzhou 510640, China
- Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, China
| | - Yueqin Qiu
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou 510640, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangzhou 510640, China
- Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, China
| | - Jing Wu
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou 510640, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangzhou 510640, China
- Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, China
| | - Chenbin Cui
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou 510640, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangzhou 510640, China
- Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, China
| | - Shilong Liu
- College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou 510640, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangzhou 510640, China
- Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, China
| | - Jing Hou
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou 510640, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangzhou 510640, China
- Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, China
| | - Chaoyang Tian
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou 510640, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangzhou 510640, China
- Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, China
| | - Li Wang
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou 510640, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangzhou 510640, China
- Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, China
| | - Kaiguo Gao
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou 510640, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangzhou 510640, China
- Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, China
| | - Zongyong Jiang
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou 510640, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangzhou 510640, China
- Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, China
| | - Xuefen Yang
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou 510640, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangzhou 510640, China
- Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, China
| |
Collapse
|
10
|
Ekregbesi P, Seibert B, Parish MA, Flores-Garcia Y, Creisher PS, Hoffmann JP, Liu J, Brayton C, Zavala F, Klein SL. Multi-System Dysregulation in Placental Malaria Contributes to Adverse Perinatal Outcomes in Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.15.633265. [PMID: 39868257 PMCID: PMC11761038 DOI: 10.1101/2025.01.15.633265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Sequestration of Plasmodium parasites in the placental vasculature causes increased morbidity and mortality in pregnant compared to non-pregnant patients in malaria-endemic regions. In this study, outbred pregnant CD1 mice with semi allogeneic fetuses were infected with transgenic Plasmodium berghei or mock-inoculated by mosquito bite at either embryonic day (E) 6 (first trimester-equivalent) or 10 (second trimester-equivalent) and compared with non-pregnant females. P. berghei-infected mosquitoes had greater biting avidity for E10 dams than uninfected mosquitoes, which was not apparent for E6 dams nor non-pregnant females. Infected E10 dams had greater numbers of parasites than E6 dams in the uterus and spleen, but not in the blood or liver. While parasites were found in placentas, no parasites were present in fetuses. Maternal infection at E6 caused greater maternal morbidity, with greater rates of fetal reabsorption and stillbirths than at E10. Infection at E10 caused adverse offspring outcomes, including growth restriction. To identify possible mechanisms of adverse offspring outcomes, E10 dams were euthanized during peak parasitemia (8 days post infection), and outcomes were compared with mock-infected dams. P. berghei caused significant systemic maternal immune activation with elevated circulating lymphocytes, eosinophils, and neutrophils and splenic cytokine concentrations. P. berghei infection at E10 increased corticosterone and decreased progesterone concentrations, which could contribute to adverse perinatal outcomes through immunomodulation. There were limited changes in the maternal fecal microbiome after P. berghei infection. Mosquito bite infection of outbred dams with P. berghei causes placental malaria and provides a novel, tractable model to investigate therapeutic treatments.
Collapse
Affiliation(s)
- Phebe Ekregbesi
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland USA
| | - Brittany Seibert
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland USA
| | - Maclaine A. Parish
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland USA
| | - Yevel Flores-Garcia
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland USA
| | - Patrick S. Creisher
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland USA
| | - Joseph P. Hoffmann
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland USA
| | - Jennifer Liu
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland USA
| | - Cory Brayton
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, Maryland USA
| | - Fidel Zavala
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland USA
| | - Sabra L. Klein
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland USA
| |
Collapse
|
11
|
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
|
12
|
Rollins RL, Griffin CD, Cowie RH. Snail coprophagy: the encounter filter, food preferences, and rat lungworm (Angiostrongylus cantonensis) prevalence. Parasite 2024; 31:76. [PMID: 39715443 DOI: 10.1051/parasite/2024075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 12/05/2024] [Indexed: 12/25/2024] Open
Abstract
Understanding the factors driving infection prevalence among host species is crucial for effective disease mitigation. Angiostrongylus cantonensis, the rat lungworm, causes neuroangiostrongyliasis and serves as an excellent model for studying infection dynamics across hosts. This study investigates the relative impact of encounter rates on A. cantonensis prevalence in snail hosts by assessing their coprophagic tendencies. Multiple-choice feeding assays were conducted with four snail species (Parmarion martensi, Laevicaulis alte, Lissachatina fulica, and Veronicella cubensis) differing in A. cantonensis prevalence. The snails were offered romaine lettuce, hibiscus flowers, papaya, and rat feces. The relative intake ratios (RIR) were calculated and used to evaluate 1) feces preference among the snail species, and 2) correlation between feces preference and A. cantonensis prevalence. We also compared preferences for feces from rats fed high-fat and balanced diets; no significant difference was observed. Feces made up the highest proportion of the diet of P. martensi (11.6%), followed by V. cubensis (7.8%), L. fulica (5.9%), and L. alte (5.1%). Additionally, P. martensi showed a significantly higher preference (RIR) than all other species. The correlation between feces preference and A. cantonensis prevalence among species was weakly positive. These findings suggest that the level of coprophagy influences encounter rates with A. cantonensis, contributing to variation in infection prevalence among snail species. However, other factors may also play a role, as preference and prevalence were only weakly correlated. Understanding these dynamics can inform strategies for managing the spread of A. cantonensis and mitigating its health impacts.
Collapse
Affiliation(s)
- Randi L Rollins
- Pacific Biosciences Research Center, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| | - Chasen D Griffin
- Pacific Biosciences Research Center, University of Hawaii at Manoa, Honolulu, Hawaii, USA - School of Life Sciences, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| | - Robert H Cowie
- Pacific Biosciences Research Center, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| |
Collapse
|
13
|
Ashrafi R, Sundberg LR, Hyvärinen P, Karvonen A. Heterogeneity of the rearing environment enhances diversity of microbial communities in intensive farming. Anim Microbiome 2024; 6:75. [PMID: 39707572 DOI: 10.1186/s42523-024-00359-8] [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: 08/28/2024] [Accepted: 11/24/2024] [Indexed: 12/23/2024] Open
Abstract
Heterogeneity of the rearing environment in farmed animals can improve welfare and stocking success by enhancing natural behaviours, reducing stress, and decreasing pathogen occurrence. Although microbial diversity is often associated with well-being, their direct and indirect effects on health of farmed animals remain underexplored. We examined the impact of structural heterogeneity of aquaculture tanks on microbial communities in tank biofilm and fish gut microbiome. Enrichment (stones and shelters) significantly promoted microbial diversity and community homogeneity in tank biofilm. However, diversity of gut microbiome did not depend on rearing treatment or microbial composition of the environment. Fish in enriched tanks exhibited greater compositional variation in gut microbiome than those in standard tanks. Tanks without enrichments had higher occurrence of potentially pathogenic bacterial families (Corynebacteriaceae and Staphylococcaceae), while enriched tanks had more beneficial gut microbes (Lactobacillus). Microbial diversity in tank biofilm was negatively associated with fish mortality during a natural epidemic of Flavobacterium columnare, suggesting a protective effect of diverse microbial communities. These findings support environmental enrichment in mitigating disease outbreaks through enhanced microbial diversity, providing important implications for disease control and sustainable health management in aquaculture.
Collapse
Affiliation(s)
- Roghaieh Ashrafi
- Department of Biological and Environmental Science, University of Jyväskylä, Jyväskylä, Finland.
| | - Lotta-Riina Sundberg
- Department of Biological and Environmental Science and Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland
| | - Pekka Hyvärinen
- Aquatic Population Dynamics, Natural Resources Institute Finland (Luke), Paltamo, Finland
| | - Anssi Karvonen
- Department of Biological and Environmental Science, University of Jyväskylä, Jyväskylä, Finland
| |
Collapse
|
14
|
Bamgbose T, Quadri A, Abdullahi IO, Inabo HI, Bello M, Kori LD, Anvikar AR, de la Fuente J, Piloto-Sardiñas E, Cabezas-Cruz A. Antiplasmodial Activity of Probiotic Limosilactobacillus fermentum YZ01 in Plasmodium berghei ANKA Infected BALB/c Mice. J Trop Med 2024; 2024:6697859. [PMID: 39703208 PMCID: PMC11658835 DOI: 10.1155/jotm/6697859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 11/30/2024] [Indexed: 12/21/2024] Open
Abstract
Malaria remains a significant global health challenge, with the deadliest infections caused by Plasmodium falciparum. In light of the escalating drug resistance and the limited effectiveness of available vaccines, innovative treatment approaches are urgently needed. This study explores the potential of the probiotic Limosilactobacillus fermentum YZ01, isolated from traditionally fermented kindirmo milk, to modify host responses to Plasmodium berghei ANKA infection. Twenty-five male BALB/c mice were grouped and administered various treatments, including probiotic-enriched yogurt alone or in combination with antibiotics. Parameters assessed included gut lactic acid bacteria (LAB) composition, parasitaemia progression, survival rates, and immune response dynamics over a 21-day postinfection period. The probiotic treatment significantly altered gut microbiota, evidenced by increased LAB counts and modulated immune responses, notably enhancing IgM and IL-4 production while reducing IFN-γ levels. Mice receiving prolonged probiotic treatment exhibited delayed parasitaemia onset, reduced mortality rates, and a more robust immune response compared to control groups. These outcomes suggest that probiotic intervention not only tempers the pathological effects of malaria but also enhances host resilience against infection. This study underscores the role of gut microbiota in infectious disease pathogenesis and supports probiotics as a promising adjunct therapy for malaria management.
Collapse
Affiliation(s)
- Timothy Bamgbose
- ICMR-National Institute of Malaria Research, Sector 8, Dwarka, New Delhi, India
- Department of Microbiology, Ahmadu Bello University, Samaru Zaria, Kaduna, Nigeria
| | - Afshana Quadri
- ICMR-National Institute of Malaria Research, Sector 8, Dwarka, New Delhi, India
| | - Isa O. Abdullahi
- Department of Microbiology, Ahmadu Bello University, Samaru Zaria, Kaduna, Nigeria
| | - Helen I. Inabo
- Department of Microbiology, Ahmadu Bello University, Samaru Zaria, Kaduna, Nigeria
| | - Mohammed Bello
- Department of Veterinary Public Health and Preventive Medicine, Ahmadu Bello University, Samaru Zaria, Kaduna, Nigeria
| | - Lokesh D. Kori
- ICMR-National Institute of Malaria Research, Sector 8, Dwarka, New Delhi, India
| | | | - José de la Fuente
- SaBio Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo s/n, Ciudad Real 13005, Spain
- Department of Veterinary Pathobiology, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater 74078, Oklahoma, USA
| | - Elianne Piloto-Sardiñas
- Direction of Animal Health, National Center for Animal and Plant Health, Carretera de Tapaste y Autopista Nacional, Apartado Postal 10, San José de las Lajas 32700, Mayabeque, Cuba
- ANSES, INRAE, Ecole Nationale Vétérinaire d'Alfort, UMR BIPAR, Laboratoire de Santé Animale, Maisons-Alfort F-94700, France
| | - Alejandro Cabezas-Cruz
- ANSES, INRAE, Ecole Nationale Vétérinaire d'Alfort, UMR BIPAR, Laboratoire de Santé Animale, Maisons-Alfort F-94700, France
| |
Collapse
|
15
|
Dias N, Dias M, Ribeiro A, Gomes N, Moraes A, Wesley M, Gonzaga C, Ramos DDAR, Braz S, Dallago B, de Carvalho JL, Hagström L, Nitz N, Hecht M. Network Analysis of Pathogenesis Markers in Murine Chagas Disease Under Antimicrobial Treatment. Microorganisms 2024; 12:2332. [PMID: 39597721 PMCID: PMC11596328 DOI: 10.3390/microorganisms12112332] [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: 10/08/2024] [Revised: 11/10/2024] [Accepted: 11/12/2024] [Indexed: 11/29/2024] Open
Abstract
Chagas disease (CD), a disease affecting millions globally, remains shrouded in scientific uncertainty, particularly regarding the role of the intestinal microbiota in disease progression. This study investigates the effects of antibiotic-induced microbiota depletion on parasite burden, immune responses, and clinical outcomes in BALB/c mice infected with either the Trypanosoma cruzi Colombiana or CL Brener strains. Mice were treated with a broad-spectrum antibiotic cocktail before infection, and parasite burden was quantified via qPCR at 30 and 100 days post-infection (dpi). Immune responses were analyzed using flow cytometry and ELISA, while histopathology was conducted on cardiac and intestinal tissues. Antibiotic treatment uncovered strain-specific correlations, with Colombiana infections affecting Bifidobacterium populations and CL Brener infections linked to Lactobacillus. Microbiota depletion initially reduced parasite burden in the heart and intestine, but an increase was observed in the chronic phase, except in the CL Brener-infected gut, where an early burden spike was followed by a decline. Antibiotic-induced bacterial shifts, such as reductions in Bacteroides and Bifidobacterium, promoted a more pro-inflammatory immune profile. These findings highlight the importance of microbiota and strain-specific factors in CD and suggest further research into microbiota manipulation as a potential therapeutic strategy.
Collapse
Affiliation(s)
- Nayra Dias
- Interdisciplinary Laboratory of Biosciences, Department of Pathology, Faculty of Medicine, University of Brasília, Brasília 70910-900, Brazil; (N.D.); (M.D.); (A.R.); (N.G.); (A.M.); (M.W.); (C.G.); (D.d.A.R.R.); (B.D.); (J.L.d.C.); (L.H.); (N.N.)
| | - Marina Dias
- Interdisciplinary Laboratory of Biosciences, Department of Pathology, Faculty of Medicine, University of Brasília, Brasília 70910-900, Brazil; (N.D.); (M.D.); (A.R.); (N.G.); (A.M.); (M.W.); (C.G.); (D.d.A.R.R.); (B.D.); (J.L.d.C.); (L.H.); (N.N.)
| | - Andressa Ribeiro
- Interdisciplinary Laboratory of Biosciences, Department of Pathology, Faculty of Medicine, University of Brasília, Brasília 70910-900, Brazil; (N.D.); (M.D.); (A.R.); (N.G.); (A.M.); (M.W.); (C.G.); (D.d.A.R.R.); (B.D.); (J.L.d.C.); (L.H.); (N.N.)
| | - Nélio Gomes
- Interdisciplinary Laboratory of Biosciences, Department of Pathology, Faculty of Medicine, University of Brasília, Brasília 70910-900, Brazil; (N.D.); (M.D.); (A.R.); (N.G.); (A.M.); (M.W.); (C.G.); (D.d.A.R.R.); (B.D.); (J.L.d.C.); (L.H.); (N.N.)
| | - Aline Moraes
- Interdisciplinary Laboratory of Biosciences, Department of Pathology, Faculty of Medicine, University of Brasília, Brasília 70910-900, Brazil; (N.D.); (M.D.); (A.R.); (N.G.); (A.M.); (M.W.); (C.G.); (D.d.A.R.R.); (B.D.); (J.L.d.C.); (L.H.); (N.N.)
| | - Moisés Wesley
- Interdisciplinary Laboratory of Biosciences, Department of Pathology, Faculty of Medicine, University of Brasília, Brasília 70910-900, Brazil; (N.D.); (M.D.); (A.R.); (N.G.); (A.M.); (M.W.); (C.G.); (D.d.A.R.R.); (B.D.); (J.L.d.C.); (L.H.); (N.N.)
| | - Carlito Gonzaga
- Interdisciplinary Laboratory of Biosciences, Department of Pathology, Faculty of Medicine, University of Brasília, Brasília 70910-900, Brazil; (N.D.); (M.D.); (A.R.); (N.G.); (A.M.); (M.W.); (C.G.); (D.d.A.R.R.); (B.D.); (J.L.d.C.); (L.H.); (N.N.)
| | - Doralina do Amaral Rabello Ramos
- Interdisciplinary Laboratory of Biosciences, Department of Pathology, Faculty of Medicine, University of Brasília, Brasília 70910-900, Brazil; (N.D.); (M.D.); (A.R.); (N.G.); (A.M.); (M.W.); (C.G.); (D.d.A.R.R.); (B.D.); (J.L.d.C.); (L.H.); (N.N.)
| | - Shélida Braz
- Institute of Exact and Technological Sciences, Federal University of Amazonas, Manaus 69000-000, Brazil;
| | - Bruno Dallago
- Interdisciplinary Laboratory of Biosciences, Department of Pathology, Faculty of Medicine, University of Brasília, Brasília 70910-900, Brazil; (N.D.); (M.D.); (A.R.); (N.G.); (A.M.); (M.W.); (C.G.); (D.d.A.R.R.); (B.D.); (J.L.d.C.); (L.H.); (N.N.)
| | - Juliana Lott de Carvalho
- Interdisciplinary Laboratory of Biosciences, Department of Pathology, Faculty of Medicine, University of Brasília, Brasília 70910-900, Brazil; (N.D.); (M.D.); (A.R.); (N.G.); (A.M.); (M.W.); (C.G.); (D.d.A.R.R.); (B.D.); (J.L.d.C.); (L.H.); (N.N.)
| | - Luciana Hagström
- Interdisciplinary Laboratory of Biosciences, Department of Pathology, Faculty of Medicine, University of Brasília, Brasília 70910-900, Brazil; (N.D.); (M.D.); (A.R.); (N.G.); (A.M.); (M.W.); (C.G.); (D.d.A.R.R.); (B.D.); (J.L.d.C.); (L.H.); (N.N.)
| | - Nadjar Nitz
- Interdisciplinary Laboratory of Biosciences, Department of Pathology, Faculty of Medicine, University of Brasília, Brasília 70910-900, Brazil; (N.D.); (M.D.); (A.R.); (N.G.); (A.M.); (M.W.); (C.G.); (D.d.A.R.R.); (B.D.); (J.L.d.C.); (L.H.); (N.N.)
| | - Mariana Hecht
- Interdisciplinary Laboratory of Biosciences, Department of Pathology, Faculty of Medicine, University of Brasília, Brasília 70910-900, Brazil; (N.D.); (M.D.); (A.R.); (N.G.); (A.M.); (M.W.); (C.G.); (D.d.A.R.R.); (B.D.); (J.L.d.C.); (L.H.); (N.N.)
| |
Collapse
|
16
|
Van Den Ham KM, Bower LK, Li S, Lorenzi H, Doumbo S, Doumtabe D, Kayentao K, Ongoiba A, Traore B, Crompton PD, Schmidt NW. The gut microbiome is associated with susceptibility to febrile malaria in Malian children. Nat Commun 2024; 15:9525. [PMID: 39500866 PMCID: PMC11538534 DOI: 10.1038/s41467-024-52953-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 09/23/2024] [Indexed: 11/08/2024] Open
Abstract
Malaria is a major public health problem, but many of the factors underlying the pathogenesis of this disease are not well understood, including protection from the development of febrile symptoms, which is observed in individuals residing in areas with moderate-to-high transmission by early adolescence. Here, we demonstrate that susceptibility to febrile malaria following Plasmodium falciparum infection is associated with the composition of the gut microbiome prior to the malaria season in 10-year-old Malian children, but not in younger children. Gnotobiotic mice colonized with the fecal samples of malaria-susceptible children were shown to have a significantly higher parasite burden following Plasmodium infection compared to gnotobiotic mice colonized with the fecal samples of malaria-resistant children. The fecal microbiome of the susceptible children was determined to be enriched for bacteria associated with inflammation, mucin degradation and gut permeability, and to have increased levels of nitric oxide-derived DNA adducts and lower levels of mucus phospholipids compared to the resistant children. Overall, these results indicate that the composition of the gut microbiome is associated with the prospective risk of febrile malaria in Malian children and suggest that modulation of the gut microbiome could decrease malaria morbidity in endemic areas.
Collapse
Affiliation(s)
- Kristin M Van Den Ham
- Ryan White Center for Pediatric Infectious Diseases and Global Health, Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Layne K Bower
- Ryan White Center for Pediatric Infectious Diseases and Global Health, Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Shanping Li
- Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Hernan Lorenzi
- Infectious Diseases Group, J. Craig Venter Institute, Bethesda, MD, USA
| | - Safiatou Doumbo
- Mali International Center of Excellence in Research; Malaria Research and Training Center, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Didier Doumtabe
- Mali International Center of Excellence in Research; Malaria Research and Training Center, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Kassoum Kayentao
- Mali International Center of Excellence in Research; Malaria Research and Training Center, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Aissata Ongoiba
- Mali International Center of Excellence in Research; Malaria Research and Training Center, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Boubacar Traore
- Mali International Center of Excellence in Research; Malaria Research and Training Center, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Peter D Crompton
- Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Nathan W Schmidt
- Ryan White Center for Pediatric Infectious Diseases and Global Health, Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
17
|
Kumar T, Maitra S, Rai R, Priyanka, Maitra S, Tirkey NN, Kumari R. The dichotomy between probiotic lactic acid bacteria and Plasmodium: A promising therapeutic avenue. Acta Trop 2024; 257:107284. [PMID: 38857820 DOI: 10.1016/j.actatropica.2024.107284] [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: 03/21/2024] [Revised: 06/02/2024] [Accepted: 06/07/2024] [Indexed: 06/12/2024]
Abstract
Our understanding of gut microbial populations and their immense influence on host immunity, health, and diseases has increased deeply in recent years. Numerous reports have identified the role of mosquito and mammalian gut microbiota in the modulation of host susceptibility to Plasmodium infection. Artemisinin resistance in malaria-endemic regions necessitates the development of new, safer, and more affordable treatments to supplement existing therapies. In this review, we compiled a colossal amount of data from numerous studies that have assessed the roles played by gut microbial communities in Plasmodium infection, progression, transmission, and severity. Most interestingly, our study points to the overwhelming evidence from experimental studies in mural malaria to human trials, suggesting that the presence of lactic acid bacteria in the gut microbiota of mammalian hosts provides a great degree of protection against malaria. Therefore, our study provides a compelling narrative for probiotic administration as an adjunct therapy for combatting malaria.
Collapse
Affiliation(s)
- Tarkeshwar Kumar
- Department of Zoology, Panch Pargana Kisan College, Ranchi University, Ranchi, Jharkhand, 835204, India.
| | - Satarupa Maitra
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Richa Rai
- Department of Zoology, Allahabad University, Prayagraj, Uttar Pradesh, India
| | - Priyanka
- Department of Zoology, Allahabad University, Prayagraj, Uttar Pradesh, India
| | - Satwat Maitra
- Noida International Institute of Medical Sciences, Greater Noida, Uttar Pradesh, India
| | | | - Rajesh Kumari
- Department of Zoology, Allahabad University, Prayagraj, Uttar Pradesh, India
| |
Collapse
|
18
|
Manuel CA, Pugazhenthi U, Fink MK, Habenicht LM, Fong DL, Leszczynski JK, Schurr MJ. A Clinical Scoring System and Impact of Athymic Nude Mouse Age on Corynebacterium bovis Clinical Disease. Comp Med 2024; 74:246-254. [PMID: 38704217 PMCID: PMC11373686 DOI: 10.30802/aalas-cm-23-000088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/31/2024] [Accepted: 04/16/2024] [Indexed: 05/06/2024]
Abstract
Corynebacterium bovis infection in biomedical research is synonymous with skin hyperkeratosis of athymic nude mice. This clinical sign can be obvious and is the namesake for 'scaly skin disease.' Other clinical signs that accompany scaly skin, including early presentation, duration, and rate of resolution, are less well known. The goal of this study was to characterize the clinical signs of C. bovis infection in nude mice under experimental conditions and develop a quantifiable scoring system. For the development, prospective trial, and application of this clinical scoring system, 93 naïve Hsd:Athymic Nude mice were used, of which 81 were exposed to soiled bedding from clinically ill C. bovis -infected NSG mice. The emergence of clinical signs was monitored and scored daily for 14 d. We identified 3 categories of clinical signs including skin hyperemia, skin hyperkeratosis, and surrogate indicators of overall health. Each of these defined categories appeared consistently and progressed and regressed temporally. We subsequently used this scoring system to determine if the age of Hsd:Athymic Nude mice (6 compared with 10 wk) at time of infection affects clinical severity. Our findings demonstrate that 6-wk-old mice demonstrate more severe clinical signs. Ten-week-old mice showed less skin hyperemia and no skin hyperkeratosis and were less affected by the infection based on surrogates of overall health. Here we show the utility of this novel scoring system and the impact of nude mouse age at the time of infection on C. bovis clinical disease.
Collapse
Affiliation(s)
- Christopher A Manuel
- Office of Laboratory Animal Resources, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- University of Colorado Cancer Center, Aurora, Colorado
| | - Umarani Pugazhenthi
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado; and
| | - Michael K Fink
- Office of Laboratory Animal Resources, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Lauren M Habenicht
- Office of Laboratory Animal Resources, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Derek L Fong
- Office of Laboratory Animal Resources, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Jori K Leszczynski
- Office of Laboratory Animal Resources, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Michael J Schurr
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|
19
|
Reed JS, Nayan R, Deckers M, Evavold BD, Lamb TJ. Influenza-induced alveolar macrophages protect against death by malaria-associated acute lung injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.12.601219. [PMID: 39026845 PMCID: PMC11257580 DOI: 10.1101/2024.07.12.601219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Lower respiratory tract infections are common in malaria-endemic areas, and there is some evidence that co-infections between various bacteria/viruses and Plasmodium may affect disease prognosis. In this study, we report the novel finding that co-infection with influenza/A/X31 protects mice from death by Plasmodium berghei NK65-Edinburgh, a model of severe malarial pulmonary leak which underpins malaria-associated acute lung injury (MA-ALI) and malaria-associated acute respiratory distress (MA-ARDS). Co-infected mice exhibit equivalent parasitemia as mice with malaria only, suggesting that the survival phenotype is due to differences in immune kinetics. We demonstrated that the pulmonary leak typical of Pb E is attenuated in co-infected mice without alteration in CD8 T cell activation and recruitment to the lung. Upon further examination of the immune response to influenza/A/X31 we identified a population of arginase 1-expressing alveolar macrophages that traffic to the lungs early during infection. In vitro these macrophages inhibit CD8 T cell activation and proliferation better than non-arginase expressing cells. Removal of arginase-1 expressing alveolar macrophages in vivo via administration of the antimetabolite gemcitabine removed the protective effects of influenza/A/X31co-infection on MA-ALI. This study opens a route to better understanding of how to modulate the immunopathology observed in pulmonary leak associated with severe malaria, which must be achieved to rationally design therapeutic interventions for MA-ARDS / MA-ALI.
Collapse
|
20
|
Sun Y, Liu P, Guo W, Guo J, Chen J, Xue X, Duan C, Wang Z, Yan X. Study on the alleviative effect of Lactobacillus plantarum on Eimeria falciformis infection. Infect Immun 2024; 92:e0013024. [PMID: 38842306 PMCID: PMC11324035 DOI: 10.1128/iai.00130-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 05/16/2024] [Indexed: 06/07/2024] Open
Abstract
Coccidia of the genus Eimeria are specialized intracellular parasitic protozoa that cause severe coccidiosis when they infect their hosts. Animals infected with Eimeria develop clinical symptoms, such as anorexia, diarrhea, and hematochezia, which can even cause death. Although the current preferred regimen for the treatment of coccidiosis is antibiotics, this treatment strategy is limited by the ban on antibiotics and the growing problem of drug resistance. Therefore, the exploration of alternative methods for controlling coccidiosis has attracted much attention. Lactobacillus plantarum has been shown to have many beneficial effects. In this study, L. plantarum M2 was used as a research object to investigate the effect of L. plantarum on intestinal inflammation induced by infection with Eimeria falciformis in mice by detecting indicators, such as oocyst output, serum cytokines, and the intestinal microbiota. Compared with that in the infection group, the percent weight loss of the mice that were administered with L. plantarum M2 was significantly reduced (P < 0.05). Supplemented L. plantarum M2 and probiotics combined with diclazuril can reduce the total oocyst output significantly (P < 0.05, P < 0.001). L. plantarum M2 had outstanding performance in maintaining intestinal barrier function, and the levels of the mucin MUC1 and the tight junction protein E-cadherin were significantly elevated (P < 0.01, P < 0.05). Studies have shown that probiotic supplementation can alleviate adverse reactions after infection and significantly improve intestinal barrier function. In addition, probiotics combined with diclazuril could optimize the partial efficacy of diclazuril, which not only enhanced the effect of antibiotics but also alleviated their adverse effects. This study expands the application of probiotics, provides new ideas for alternative strategies for coccidia control, and suggests a basis for related research on lactobacilli antagonizing intracellular pathogen infection.IMPORTANCECoccidia of the genus Eimeria are specialized intracellular parasitic protozoa, and the current preferred regimen for the treatment of coccidiosis is antibiotics. However, due to antibiotic bans and drug resistance, the exploration of alternative methods for controlling coccidiosis has attracted much attention. In this work, we focused on Lactobacillus plantarum M2 and found that probiotic supplementation can alleviate adverse reactions after infection and improve intestinal barrier function. This study proposes the possibility of using lactic acid bacteria to control coccidiosis, and its potential mechanism needs further exploration.
Collapse
Affiliation(s)
- Yufei Sun
- College of Food
Science and Engineering, Inner Mongolia Agricultural
University, Hohhot,
Inner Mongolia, China
| | - Pufang Liu
- College of Food
Science and Engineering, Inner Mongolia Agricultural
University, Hohhot,
Inner Mongolia, China
| | - Wenhui Guo
- College of Food
Science and Engineering, Inner Mongolia Agricultural
University, Hohhot,
Inner Mongolia, China
| | - Jun Guo
- College of Food
Science and Engineering, Inner Mongolia Agricultural
University, Hohhot,
Inner Mongolia, China
| | - Jia Chen
- College of Food
Science and Engineering, Inner Mongolia Agricultural
University, Hohhot,
Inner Mongolia, China
| | - Xinyu Xue
- College of Food
Science and Engineering, Inner Mongolia Agricultural
University, Hohhot,
Inner Mongolia, China
| | - Chao Duan
- College of Food
Science and Engineering, Inner Mongolia Agricultural
University, Hohhot,
Inner Mongolia, China
| | - Zixuan Wang
- College of Food
Science and Engineering, Inner Mongolia Agricultural
University, Hohhot,
Inner Mongolia, China
| | - Xinlei Yan
- College of Food
Science and Engineering, Inner Mongolia Agricultural
University, Hohhot,
Inner Mongolia, China
| |
Collapse
|
21
|
Yuan J, Yu Y, Li S, Zhang X, Zhang C, Li R, Hu J, Si S, Zhang C, Xiang J, Li F. Shrimp shapes a resistance trait against vibriosis by memorizing the colonization resistance of intestinal microbiota. PLoS Pathog 2024; 20:e1012321. [PMID: 38990823 PMCID: PMC11239079 DOI: 10.1371/journal.ppat.1012321] [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: 02/05/2024] [Accepted: 06/06/2024] [Indexed: 07/13/2024] Open
Abstract
Vibriosis is one of the most serious diseases that commonly occurs in aquatic animals, thus, shaping a steady inherited resistance trait in organisms has received the highest priority in aquaculture. Whereas, the mechanisms underlying the development of such a resistance trait are mostly elusive. In this study, we constructed vibriosis-resistant and susceptible families of the Pacific white shrimp Litopenaeus vannamei after four generations of artificial selection. Microbiome sequencing indicated that shrimp can successfully develop a colonization resistance trait against Vibrio infections. This trait was characterized by a microbial community structure with specific enrichment of a single probiotic species (namely Shewanella algae), and notably, its formation was inheritable and might be memorized by host epigenetic remodeling. Regardless of the infection status, a group of genes was specifically activated in the resistant family through disruption of complete methylation. Specifically, hypo-methylation and hyper-expression of genes related to lactate dehydrogenase (LDH) and iron homeostasis might provide rich sources of specific carbon (lactate) and ions for the colonization of S. algae, which directly results in the reduction of Vibrio load in shrimp. Lactate feeding increased the survival of shrimp, while knockdown of LDH gene decreased the survival when shrimp was infected by Vibrio pathogens. In addition, treatment of shrimp with the methyltransferase inhibitor 5-azacytidine resulted in upregulations of LDH and some protein processing genes, significant enrichment of S. algae, and simultaneous reduction of Vibrio in shrimp. Our results suggest that the colonization resistance can be memorized as epigenetic information by the host, which has played a pivotal role in vibriosis resistance. The findings of this study will aid in disease control and the selection of superior lines of shrimp with high disease resistance.
Collapse
Affiliation(s)
- Jianbo Yuan
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Chinese Academy of Sciences, Wuhan, China
| | - Yang Yu
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Chinese Academy of Sciences, Wuhan, China
| | - Shihao Li
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Chinese Academy of Sciences, Wuhan, China
| | - Xiaojun Zhang
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Chinese Academy of Sciences, Wuhan, China
| | - Chuntao Zhang
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Roujing Li
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
| | - Jie Hu
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
| | - Shuqing Si
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Chengyi Zhang
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jianhai Xiang
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Chinese Academy of Sciences, Wuhan, China
| | - Fuhua Li
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Chinese Academy of Sciences, Wuhan, China
| |
Collapse
|
22
|
Schmidt N, Van Den Ham K, Bower L, Li S, Lorenzi H, Doumbo S, Doumtabe D, Kayentao K, Ongoiba A, Traore B, Crompton P. Susceptibility to febrile malaria is associated with an inflammatory gut microbiome. RESEARCH SQUARE 2024:rs.3.rs-3974068. [PMID: 38645126 PMCID: PMC11030534 DOI: 10.21203/rs.3.rs-3974068/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Malaria is a major public health problem, but many of the factors underlying the pathogenesis of this disease are not well understood. Here, we demonstrate in Malian children that susceptibility to febrile malaria following infection with Plasmodium falciparum is associated with the composition of the gut microbiome prior to the malaria season. Gnotobiotic mice colonized with the fecal samples of malaria-susceptible children had a significantly higher parasite burden following Plasmodium infection compared to gnotobiotic mice colonized with the fecal samples of malaria-resistant children. The fecal microbiome of the susceptible children was enriched for bacteria associated with inflammation, mucin degradation, gut permeability and inflammatory bowel disorders (e.g., Ruminococcus gauvreauii, Ruminococcus torques, Dorea formicigenerans, Dorea longicatena, Lachnoclostridium phocaeense and Lachnoclostridium sp. YL32). However, the susceptible children also had a greater abundance of bacteria known to produce anti-inflammatory short-chain fatty acids and those associated with favorable prognosis and remission following dysbiotic intestinal events (e.g., Anaerobutyricum hallii, Blautia producta and Sellimonas intestinalis). Metabolomics analysis of the human fecal samples corroborated the existence of inflammatory and recovery-associated features within the gut microbiome of the susceptible children. There was an enrichment of nitric oxide-derived DNA adducts (deoxyinosine and deoxyuridine) and long-chain fatty acids, the absorption of which has been shown to be inhibited by inflamed intestinal epithelial cells, and a decrease in the abundance of mucus phospholipids. Nevertheless, there were also increased levels of pseudouridine and hypoxanthine, which have been shown to be regulated in response to cellular stress and to promote recovery following injury or hypoxia. Overall, these results indicate that the gut microbiome may contribute malaria pathogenesis and suggest that therapies targeting intestinal inflammation could decrease malaria susceptibility.
Collapse
|
23
|
Van Den Ham KM, Little MR, Bednarski OJ, Fusco EM, Mandal RK, Mitra R, Li S, Doumbo S, Doumtabe D, Kayentao K, Ongoiba A, Traore B, Crompton PD, Schmidt NW. Creation of a non-Western humanized gnotobiotic mouse model through the transplantation of rural African fecal microbiota. Microbiol Spectr 2023; 11:e0155423. [PMID: 37819130 PMCID: PMC10714993 DOI: 10.1128/spectrum.01554-23] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 09/05/2023] [Indexed: 10/13/2023] Open
Abstract
IMPORTANCE There is increasing evidence that microbes residing within the intestines (gut microbiota) play important roles in the well-being of humans. Yet, there are considerable challenges in determining the specific role of gut microbiota in human diseases owing to the complexity of diverse internal and environmental factors that can contribute to diseases. Mice devoid of all microorganisms (germ-free mice) can be colonized with human stool samples to examine the specific contribution of the gut microbiota to a disease. These approaches have been primarily focused on stool samples obtained from individuals in Western countries. Thus, there is limited understanding as to whether the same methods used to colonize germ-free mice with stool from Western individuals would apply to the colonization of germ-free mice with stool from non-Western individuals. Here, we report the results from colonizing germ-free mice with stool samples of Malian children.
Collapse
Affiliation(s)
- Kristin M. Van Den Ham
- Department of Pediatrics, Ryan White Center for Pediatric Infectious Diseases and Global Health, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Morgan R. Little
- Department of Pediatrics, Ryan White Center for Pediatric Infectious Diseases and Global Health, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Olivia J. Bednarski
- Department of Pediatrics, Ryan White Center for Pediatric Infectious Diseases and Global Health, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Elizabeth M. Fusco
- Department of Pediatrics, Ryan White Center for Pediatric Infectious Diseases and Global Health, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Rabindra K. Mandal
- Department of Pediatrics, Ryan White Center for Pediatric Infectious Diseases and Global Health, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Riten Mitra
- Department of Bioinformatics and Biostatistics, University of Louisville, Louisville, Kentucky, USA
| | - Shanping Li
- Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Safiatou Doumbo
- Mali International Center of Excellence in Research, Malaria Research and Training Center, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Didier Doumtabe
- Mali International Center of Excellence in Research, Malaria Research and Training Center, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Kassoum Kayentao
- Mali International Center of Excellence in Research, Malaria Research and Training Center, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Aissata Ongoiba
- Mali International Center of Excellence in Research, Malaria Research and Training Center, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Boubacar Traore
- Mali International Center of Excellence in Research, Malaria Research and Training Center, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Peter D. Crompton
- Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Nathan W. Schmidt
- Department of Pediatrics, Ryan White Center for Pediatric Infectious Diseases and Global Health, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
24
|
Golenser J, Hunt NH, Birman I, Jaffe CL, Zech J, Mäder K, Gold D. Applicability of Redirecting Artemisinins for New Targets. GLOBAL CHALLENGES (HOBOKEN, NJ) 2023; 7:2300030. [PMID: 38094863 PMCID: PMC10714028 DOI: 10.1002/gch2.202300030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 08/03/2023] [Indexed: 10/16/2024]
Abstract
Employing new therapeutic indications for drugs that are already approved for human use has obvious advantages, including reduced costs and timelines, because some routine steps of drug development and regulation are not required. This work concentrates on the redirection of artemisinins (ARTS) that already are approved for clinical use, or investigated, for malaria treatment. Several mechanisms of action are suggested for ARTS, among which only a few have been successfully examined in vivo, mainly the induction of oxidant stress and anti-inflammatory effects. Despite these seemingly contradictory effects, ARTS are proposed for repurposing in treatment of inflammatory disorders and diverse types of diseases caused by viral, bacterial, fungal, and parasitic infections. When pathogens are treated the expected outcome is diminution of the causative agents and/or their inflammatory damage. In general, repurposing ARTS is successful in only a very few cases, specifically when a valid mechanism can be targeted using an additional therapeutic agent and appropriate drug delivery. Investigation of repurposing should include optimization of drug combinations followed by examination in relevant cell lines, organoids, and animal models, before moving to clinical trials.
Collapse
Affiliation(s)
- Jacob Golenser
- Department of Microbiology and Molecular GeneticsKuvin Center for the Study of Infectious and Tropical DiseasesThe Hebrew University – Hadassah Medical CenterJerusalemIsrael
| | - Nicholas H. Hunt
- School of Medical SciencesUniversity of SydneySydney2050Australia
| | - Ida Birman
- Department of Microbiology and Molecular GeneticsKuvin Center for the Study of Infectious and Tropical DiseasesThe Hebrew University – Hadassah Medical CenterJerusalemIsrael
| | - Charles L. Jaffe
- Department of Microbiology and Molecular GeneticsKuvin Center for the Study of Infectious and Tropical DiseasesThe Hebrew University – Hadassah Medical CenterJerusalemIsrael
| | - Johanna Zech
- Institute of PharmacyMartin Luther University Halle‐Wittenberg06108HalleGermany
| | - Karsten Mäder
- Institute of PharmacyMartin Luther University Halle‐Wittenberg06108HalleGermany
| | - Daniel Gold
- Department of Clinical Microbiology and ImmunologyFaculty of MedicineTel Aviv UniversityTel AvivIsrael
| |
Collapse
|
25
|
Navine AK, Paxton KL, Paxton EH, Hart PJ, Foster JT, McInerney N, Fleischer RC, Videvall E. Microbiomes associated with avian malaria survival differ between susceptible Hawaiian honeycreepers and sympatric malaria-resistant introduced birds. Mol Ecol 2023; 32:6659-6670. [PMID: 36281504 DOI: 10.1111/mec.16743] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 10/11/2022] [Accepted: 10/14/2022] [Indexed: 11/30/2022]
Abstract
Of the estimated 55 Hawaiian honeycreepers (subfamily Carduelinae) only 17 species remain, nine of which the International Union for Conservation of Nature considers endangered. Among the most pressing threats to honeycreeper survival is avian malaria, caused by the introduced blood parasite Plasmodium relictum, which is increasing in distribution in Hawai'i as a result of climate change. Preventing further honeycreeper decline will require innovative conservation strategies that confront malaria from multiple angles. Research on mammals has revealed strong connections between gut microbiome composition and malaria susceptibility, illuminating a potential novel approach to malaria control through the manipulation of gut microbiota. One honeycreeper species, Hawai'i 'amakihi (Chlorodrepanis virens), persists in areas of high malaria prevalence, indicating they have acquired some level of immunity. To investigate if avian host-specific microbes may be associated with malaria survival, we characterized cloacal microbiomes and malaria infection for 174 'amakihi and 172 malaria-resistant warbling white-eyes (Zosterops japonicus) from Hawai'i Island using 16S rRNA gene metabarcoding and quantitative polymerase chain reaction. Neither microbial alpha nor beta diversity covaried with infection, but 149 microbes showed positive associations with malaria survivors. Among these were Escherichia and Lactobacillus spp., which appear to mitigate malaria severity in mammalian hosts, revealing promising candidates for future probiotic research for augmenting malaria immunity in sensitive endangered species.
Collapse
Affiliation(s)
- Amanda K Navine
- Biology Department, University of Hawai'i at Hilo, Hilo, Hawaii, USA
- Center for Conservation Genomics, Smithsonian Conservation Biology Institute, National Zoological Park, Washington, District of Columbia, USA
| | - Kristina L Paxton
- Hawai'i Cooperative Studies Unit, University of Hawai'i at Hilo, Hawai'i National Park, Hawaii, USA
| | - Eben H Paxton
- U.S. Geological Survey, Pacific Island Ecosystems Research Center, Hawai'i National Park, Hawaii, USA
| | - Patrick J Hart
- Biology Department, University of Hawai'i at Hilo, Hilo, Hawaii, USA
| | - Jeffrey T Foster
- Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, Arizona, USA
| | - Nancy McInerney
- Center for Conservation Genomics, Smithsonian Conservation Biology Institute, National Zoological Park, Washington, District of Columbia, USA
| | - Robert C Fleischer
- Center for Conservation Genomics, Smithsonian Conservation Biology Institute, National Zoological Park, Washington, District of Columbia, USA
| | - Elin Videvall
- Center for Conservation Genomics, Smithsonian Conservation Biology Institute, National Zoological Park, Washington, District of Columbia, USA
- Department of Ecology and Evolutionary Biology, Brown University, Providence, Rhode Island, USA
- Institute at Brown for Environment and Society, Brown University, Providence, Rhode Island, USA
- Department of Ecology and Genetics, Uppsala University, Uppsala, Sweden
| |
Collapse
|
26
|
Shi H, Yu X, Cheng G. Impact of the microbiome on mosquito-borne diseases. Protein Cell 2023; 14:743-761. [PMID: 37186167 PMCID: PMC10599646 DOI: 10.1093/procel/pwad021] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 04/10/2023] [Indexed: 05/17/2023] Open
Abstract
Mosquito-borne diseases present a significant threat to human health, with the possibility of outbreaks of new mosquito-borne diseases always looming. Unfortunately, current measures to combat these diseases such as vaccines and drugs are often either unavailable or ineffective. However, recent studies on microbiomes may reveal promising strategies to fight these diseases. In this review, we examine recent advances in our understanding of the effects of both the mosquito and vertebrate microbiomes on mosquito-borne diseases. We argue that the mosquito microbiome can have direct and indirect impacts on the transmission of these diseases, with mosquito symbiotic microorganisms, particularly Wolbachia bacteria, showing potential for controlling mosquito-borne diseases. Moreover, the skin microbiome of vertebrates plays a significant role in mosquito preferences, while the gut microbiome has an impact on the progression of mosquito-borne diseases in humans. As researchers continue to explore the role of microbiomes in mosquito-borne diseases, we highlight some promising future directions for this field. Ultimately, a better understanding of the interplay between mosquitoes, their hosts, pathogens, and the microbiomes of mosquitoes and hosts may hold the key to preventing and controlling mosquito-borne diseases.
Collapse
Affiliation(s)
- Huicheng Shi
- Tsinghua University-Peking University Joint Center for Life Sciences, School of Medicine, Tsinghua University, Beijing 100084, China
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen 518000, China
| | - Xi Yu
- Tsinghua University-Peking University Joint Center for Life Sciences, School of Medicine, Tsinghua University, Beijing 100084, China
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen 518000, China
| | - Gong Cheng
- Tsinghua University-Peking University Joint Center for Life Sciences, School of Medicine, Tsinghua University, Beijing 100084, China
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen 518000, China
- Department of Parasitology, School of Basic Medical Sciences, Central South University, Changsha 410013, China
| |
Collapse
|
27
|
Tatsinkou LLT, Fossi BT, Sotoing GT, Mambou HMAY, Ivo PEA, Achidi EA. Prophylactic effects of probiotic bacterium Latilactobacillus sakei on haematological parameters and cytokine profile of mice infected with Plasmodium berghei ANKA during early malaria infection. Life Sci 2023; 331:122056. [PMID: 37652156 DOI: 10.1016/j.lfs.2023.122056] [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: 03/29/2023] [Revised: 08/26/2023] [Accepted: 08/28/2023] [Indexed: 09/02/2023]
Abstract
Malaria is a deadly parasitic disease caused a by protozoan parasite of the genus plasmodium. The challenges facing by chemotherapy and vector control couple with the lack of vaccine against malaria necessitate an urgent need for the development of alternative treatment regimens to combat this disease. One possible antimalarial treatment regimen is the use of probiotic bacteria as dietary supplements. Traditionally fermented milk is a rich source of probiotic bacteria that up to date, very few studies have been carried out on their immunoprotective effects against early malaria infection in mice. This study sought to assess the prophylactic activities of a probiotic bacterium Latilactobacillus sakei on malaria and inflammation in Plasmodium berghei infected mice. The probiotic bacterium was isolated from the Fulani's traditionally fermented milk and identified using the sequencing of the 16S r RNA gene. The repository activity of L. sakei on malaria was assessed using the method described by Peters with slight modification. Eighty-four BALB/c mice were randomly divided into two sets of seven groups of six mice each. One set received orally different doses of L. sakei Chloroquine and Sulfadoxine/Pyrimethamine for seven days before infection while the other set received for fourteen days before infection with 0.1 mL of 107Plasmodium berghei. Parasitaemia density, haematological parameters and inflammatory cytokines profile were evaluated. Data were presented as Mean ± SEM and analysed using SPSS version 20.0. The results of this study revealed that L. sakei significantly (p < 0.05) reduced in dose dependent manner parasite load, body weight loss and reduction of body temperature in all the treated mice when compare to untreated mice. Leukocytopenia, thrombocytosis and inflammation were also found to be significantly (p < 0.05) prevented in treated mice as compared to untreated mice. This study suggested that L sakei possesses immunomodulation and protective effects on early malaria infection in Plasmodium berghei mice.
Collapse
Affiliation(s)
| | - Bertrand Tatsinkou Fossi
- Department of Microbiology and Parasitology, Faculty of Science, University of Buea, PO Box 63, Buea, Cameroon.
| | - Germain Taiwe Sotoing
- Department of Zoology and Animal Physiology, Faculty of Science, University of Buea, PO Box 63, Buea, Cameroon
| | | | | | - Eric Akum Achidi
- Department of Biochemistry and Molecular Biology, Faculty of Science, University of Buea, PO Box 63, Buea, Cameroon
| |
Collapse
|
28
|
Mandal RK, Mandal A, Denny JE, Namazii R, John CC, Schmidt NW. Gut Bacteroides act in a microbial consortium to cause susceptibility to severe malaria. Nat Commun 2023; 14:6465. [PMID: 37833304 PMCID: PMC10575898 DOI: 10.1038/s41467-023-42235-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 10/03/2023] [Indexed: 10/15/2023] Open
Abstract
Malaria is caused by Plasmodium species and remains a significant cause of morbidity and mortality globally. Gut bacteria can influence the severity of malaria, but the contribution of specific bacteria to the risk of severe malaria is unknown. Here, multiomics approaches demonstrate that specific species of Bacteroides are causally linked to the risk of severe malaria. Plasmodium yoelii hyperparasitemia-resistant mice gavaged with murine-isolated Bacteroides fragilis develop P. yoelii hyperparasitemia. Moreover, Bacteroides are significantly more abundant in Ugandan children with severe malarial anemia than with asymptomatic P. falciparum infection. Human isolates of Bacteroides caccae, Bacteroides uniformis, and Bacteroides ovatus were able to cause susceptibility to severe malaria in mice. While monocolonization of germ-free mice with Bacteroides alone is insufficient to cause susceptibility to hyperparasitemia, meta-analysis across multiple studies support a main role for Bacteroides in susceptibility to severe malaria. Approaches that target gut Bacteroides present an opportunity to prevent severe malaria and associated deaths.
Collapse
Affiliation(s)
- Rabindra K Mandal
- Ryan White Center for Pediatric Infectious Diseases and Global Health, Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Anita Mandal
- Ryan White Center for Pediatric Infectious Diseases and Global Health, Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Joshua E Denny
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, USA
| | - Ruth Namazii
- Department of Paediatrics and Child Health, Makerere University, Kampala, Uganda
| | - Chandy C John
- Ryan White Center for Pediatric Infectious Diseases and Global Health, Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Nathan W Schmidt
- Ryan White Center for Pediatric Infectious Diseases and Global Health, Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA.
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, USA.
| |
Collapse
|
29
|
Sriboonvorakul N, Chotivanich K, Silachamroon U, Phumratanaprapin W, Adams JH, Dondorp AM, Leopold SJ. Intestinal injury and the gut microbiota in patients with Plasmodium falciparum malaria. PLoS Pathog 2023; 19:e1011661. [PMID: 37856470 PMCID: PMC10586672 DOI: 10.1371/journal.ppat.1011661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2023] Open
Abstract
The pathophysiology of severe falciparum malaria involves a complex interaction between the host, parasite, and gut microbes. In this review, we focus on understanding parasite-induced intestinal injury and changes in the human intestinal microbiota composition in patients with Plasmodium falciparum malaria. During the blood stage of P. falciparum infection, infected red blood cells adhere to the vascular endothelium, leading to widespread microcirculatory obstruction in critical tissues, including the splanchnic vasculature. This process may cause intestinal injury and gut leakage. Epidemiological studies indicate higher rates of concurrent bacteraemia in severe malaria cases. Furthermore, severe malaria patients exhibit alterations in the composition and diversity of the intestinal microbiota, although the exact contribution to pathophysiology remains unclear. Mouse studies have demonstrated that the gut microbiota composition can impact susceptibility to Plasmodium infections. In patients with severe malaria, the microbiota shows an enrichment of pathobionts, including pathogens that are known to cause concomitant bloodstream infections. Microbial metabolites have also been detected in the plasma of severe malaria patients, potentially contributing to metabolic acidosis and other clinical complications. However, establishing causal relationships requires intervention studies targeting the gut microbiota.
Collapse
Affiliation(s)
- Natthida Sriboonvorakul
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Kesinee Chotivanich
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Udomsak Silachamroon
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Weerapong Phumratanaprapin
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - John H. Adams
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, Tampa, Florida, United States of America
| | - Arjen M. Dondorp
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
| | - Stije J. Leopold
- Department of Internal Medicine, Division of Infectious Diseases, Amsterdam University Medical Center, location AMC, the Netherlands
| |
Collapse
|
30
|
Mandal RK, Schmidt NW. Mechanistic insights into the interaction between the host gut microbiome and malaria. PLoS Pathog 2023; 19:e1011665. [PMID: 37824458 PMCID: PMC10569623 DOI: 10.1371/journal.ppat.1011665] [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: 10/14/2023] Open
Abstract
Malaria is a devastating infectious disease and significant global health burden caused by the bite of a Plasmodium-infected female Anopheles mosquito. Gut microbiota was recently discovered as a risk factor of severe malaria. This review entails the recent advances on the impact of gut microbiota composition on malaria severity and consequence of malaria infection on gut microbiota in mammalian hosts. Additionally, this review provides mechanistic insight into interactions that might occur between gut microbiota and host immunity which in turn can modulate malaria severity. Finally, approaches to modulate gut microbiota composition are discussed. We anticipate this review will facilitate novel hypotheses to move the malaria-gut microbiome field forward.
Collapse
Affiliation(s)
- Rabindra K. Mandal
- Ryan White Center for Pediatric Infectious Diseases and Global Health, Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indiana, United States of America
| | - Nathan W. Schmidt
- Ryan White Center for Pediatric Infectious Diseases and Global Health, Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indiana, United States of America
| |
Collapse
|
31
|
Su X, Stadler RV, Xu F, Wu J. Malaria Genomics, Vaccine Development, and Microbiome. Pathogens 2023; 12:1061. [PMID: 37624021 PMCID: PMC10459703 DOI: 10.3390/pathogens12081061] [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: 07/21/2023] [Revised: 08/14/2023] [Accepted: 08/15/2023] [Indexed: 08/26/2023] Open
Abstract
Recent advances in malaria genetics and genomics have transformed many aspects of malaria research in areas of molecular evolution, epidemiology, transmission, host-parasite interaction, drug resistance, pathogenicity, and vaccine development. Here, in addition to introducing some background information on malaria parasite biology, parasite genetics/genomics, and genotyping methods, we discuss some applications of genetic and genomic approaches in vaccine development and in studying interactions with microbiota. Genetic and genomic data can be used to search for novel vaccine targets, design an effective vaccine strategy, identify protective antigens in a whole-organism vaccine, and evaluate the efficacy of a vaccine. Microbiota has been shown to influence disease outcomes and vaccine efficacy; studying the effects of microbiota in pathogenicity and immunity may provide information for disease control. Malaria genetics and genomics will continue to contribute greatly to many fields of malaria research.
Collapse
Affiliation(s)
- Xinzhuan Su
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA; (R.V.S.); (F.X.); (J.W.)
| | | | | | | |
Collapse
|
32
|
Beyhan YE, Yıldız MR. Microbiota and parasite relationship. Diagn Microbiol Infect Dis 2023; 106:115954. [PMID: 37267741 DOI: 10.1016/j.diagmicrobio.2023.115954] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/29/2023] [Accepted: 04/11/2023] [Indexed: 06/04/2023]
Abstract
The diversity of microbiota is different in each person. Many health problems such as autoimmune diseases, diabetes, cardiovascular diseases, and depression can be caused by microbiota imbalance. Since the parasite needs a host to survive, it interacts closely with the microbiota elements. Blastocystis acts on the inflammatory state of the intestine and may cause various gastrointestinal symptoms, on the contrary, it is more important for gut health because it causes bacterial diversity and richness. Blastocystis is associated with changes in gut microbiota composition, the ultimate indicator of which is the Firmicutes/Bacteroidetes ratio. The Bifidobacterium genus was significantly reduced in IBS patients and Blastocystis, and there is a significant decrease in Faecalibacterium prausnitzii, which has anti-inflammatory properties in Blastocystis infection without IBS. Lactobacillus species reduce the presence of Giardia, and the produced bacteriocins prevent parasite adhesion. The presence of helminths has been strongly associated with the transition from Bacteroidetes to Firmicutes and Clostridia. Contrary to Ascaris, alpha diversity in the intestinal microbiota decreases in chronic Trichuris muris infection, and growth and nutrient metabolism efficiency can be suppressed. Helminth infections indirectly affect mood and behavior in children through their effects on microbiota change. The main and focus of this review is to address the relationship of parasites with microbiota elements and to review the data about what changes they cause. Microbiota studies have gained importance recently and it is thought that it will contribute to the treatment of many diseases as well as in the fight against parasitic diseases in the future.
Collapse
Affiliation(s)
- Yunus E Beyhan
- Department of Parasitology, Van Yüzüncü Yil University Faculty of Medicine, Van, Turkey.
| | - Muhammed R Yıldız
- Department of Parasitology, Van Yüzüncü Yil University Faculty of Medicine, Van, Turkey
| |
Collapse
|
33
|
Talukdar D, Bandopadhyay P, Ray Y, Paul SR, Sarif J, D'Rozario R, Lahiri A, Das S, Bhowmick D, Chatterjee S, Das B, Ganguly D. Association of gut microbial dysbiosis with disease severity, response to therapy and disease outcomes in Indian patients with COVID-19. Gut Pathog 2023; 15:22. [PMID: 37161621 PMCID: PMC10170741 DOI: 10.1186/s13099-023-00546-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 04/13/2023] [Indexed: 05/11/2023] Open
Abstract
BACKGROUND Severe coronavirus disease 2019 (COVID-19) is associated with systemic hyper-inflammation. An adaptive interaction between gut microbiota and host immune systems is important for intestinal homeostasis and systemic immune regulation. The association of gut microbial composition and functions with COVID-19 disease severity is sparse, especially in India. We analysed faecal microbial diversity and abundances in a cohort of Indian COVID-19 patients to identify key signatures in the gut microbial ecology in patients with severe COVID-19 disease as well as in response to different therapies. The composition of the gut microbiome was characterized using 16Sr RNA gene sequences of genomic DNA extracted from faecal samples of 52 COVID-19 patients. Metabolic pathways across the groups were predicted using PICRUSt2. All statistical analyses were done using Vegan in the R environment. Plasma cytokine abundance at recruitment was measured in a multiplex assay. RESULTS The gut microbiome composition of mild and severe patients was found to be significantly different. Immunomodulatory commensals, viz. Lachnospiraceae family members and Bifidobacteria producing butyrate and short-chain fatty acids (SCFAs), were under represented in patients with severe COVID-19, with an increased abundance of opportunistic pathogens like Eggerthella. The higher abundance of Lachnoclostridium in severe disease was reduced in response to convalescent plasma therapy. Specific microbial genera showed distinctive trends in enriched metabolic pathways, strong correlations with blood plasma cytokine levels, and associative link to disease outcomes. CONCLUSION Our study indicates that, along with SARS-CoV-2, a dysbiotic gut microbial community may also play an important role in COVID-19 severity through modulation of host immune responses.
Collapse
Affiliation(s)
- Daizee Talukdar
- Functional Genomics Laboratory, Translational Health Science and Technology Institute, Faridabad, India
| | - Purbita Bandopadhyay
- CSIR-Indian Institute of Chemical Biology, Kolkata, India
- Academy of Scientific and Innovative Research, Ghaziabad, India
| | - Yogiraj Ray
- Department of Medicine, Infectious Diseases and Beleghata General Hospital, Kolkata, India
- Department of Infectious Disease, SSKM Hospital, Kolkata, India
| | - Shekhar Ranjan Paul
- Department of Medicine, Infectious Diseases and Beleghata General Hospital, Kolkata, India
| | - Jafar Sarif
- CSIR-Indian Institute of Chemical Biology, Kolkata, India
- Academy of Scientific and Innovative Research, Ghaziabad, India
| | - Ranit D'Rozario
- CSIR-Indian Institute of Chemical Biology, Kolkata, India
- Academy of Scientific and Innovative Research, Ghaziabad, India
| | - Abhishake Lahiri
- CSIR-Indian Institute of Chemical Biology, Kolkata, India
- Academy of Scientific and Innovative Research, Ghaziabad, India
| | - Santanu Das
- Functional Genomics Laboratory, Translational Health Science and Technology Institute, Faridabad, India
| | - Debaleena Bhowmick
- CSIR-Indian Institute of Chemical Biology, Kolkata, India
- Academy of Scientific and Innovative Research, Ghaziabad, India
| | - Shilpak Chatterjee
- CSIR-Indian Institute of Chemical Biology, Kolkata, India
- Academy of Scientific and Innovative Research, Ghaziabad, India
| | - Bhabatosh Das
- Functional Genomics Laboratory, Translational Health Science and Technology Institute, Faridabad, India.
| | - Dipyaman Ganguly
- CSIR-Indian Institute of Chemical Biology, Kolkata, India.
- Academy of Scientific and Innovative Research, Ghaziabad, India.
| |
Collapse
|
34
|
Drewry LL, Pewe LL, Hancox LS, Van de Wall S, Harty JT. CD4 T Cell-Dependent and -Independent Roles for IFN-γ in Blood-Stage Malaria. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:1305-1313. [PMID: 36939394 PMCID: PMC10121907 DOI: 10.4049/jimmunol.2200899] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 02/27/2023] [Indexed: 03/21/2023]
Abstract
Production of IFN-γ by CD4 T cells is widely theorized to control Plasmodium parasite burden during blood-stage malaria. Surprisingly, the specific and crucial mechanisms through which this highly pleiotropic cytokine acts to confer protection against malarial disease remain largely untested in vivo. Here we used a CD4 T cell-restricted Cre-Lox IFN-γ excision mouse model to test whether and how CD4 T cell-derived IFN-γ controls blood-stage malaria. Although complete absence of IFN-γ compromised control of the acute and the chronic, recrudescent blood-stage infections with P. c. chabaudi, we identified a specific, albeit modest, role for CD4 T cell-derived IFN-γ in limiting parasite burden only during the chronic stages of P. c. chabaudi malaria. CD4 T cell IFN-γ promoted IgG Ab class switching to the IgG2c isotype during P. c. chabaudi malaria in C57BL/6 mice. Unexpectedly, our data do not support gross defects in phagocytic activity in IFN-γ-deficient hosts infected with blood-stage malaria. Together, our data confirm CD4 T cell-dependent roles for IFN-γ but suggest CD4 T cell-independent roles for IFN-γ in immune responses to blood-stage malaria.
Collapse
|
35
|
Dutton CL, Maisha FM, Quinn EB, Morales KL, Moore JM, Mulligan CJ. Maternal Psychosocial Stress Is Associated with Reduced Diversity in the Early Infant Gut Microbiome. Microorganisms 2023; 11:microorganisms11040975. [PMID: 37110398 PMCID: PMC10142543 DOI: 10.3390/microorganisms11040975] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 03/28/2023] [Accepted: 04/05/2023] [Indexed: 04/29/2023] Open
Abstract
The developing infant gut microbiome is highly sensitive to environmental exposures, enabling its evolution into an organ that supports the immune system, confers protection from infection, and facilitates optimal gut and central nervous system function. In this study, we focus on the impact of maternal psychosocial stress on the infant gut microbiome. Forty-seven mother-infant dyads were recruited at the HEAL Africa Hospital in Goma, Democratic Republic of Congo. Extensive medical, demographic, and psychosocial stress data were collected at birth, and infant stool samples were collected at six weeks, three months, and six months. A composite maternal psychosocial stress score was created, based on eight questionnaires to capture a diverse range of stress exposures. Full-length 16S rRNA gene sequences were generated. Infants of mothers with high composite stress scores showed lower levels of gut microbiome beta diversity at six weeks and three months, as well as higher levels of alpha diversity at six months compared to infants of low stress mothers. Longitudinal analyses showed that infants of high stress mothers had lower levels of health-promoting Lactobacillus gasseri and Bifidobacterium pseudocatenulatum at six weeks compared to infants of low stress mothers, but the differences largely disappeared by three to six months. Previous research has shown that L. gasseri can be used as a probiotic to reduce inflammation, stress, and fatigue, as well as to improve mental state, while B. pseudocatenulatum is important in modulating the gut-brain axis in early development and in preventing mood disorders. Our finding of reduced levels of these health-promoting bacteria in infants of high stress mothers suggests that the infant gut microbiome may help mediate the effect of maternal stress on infant health and development.
Collapse
Affiliation(s)
- Christopher L Dutton
- Department of Anthropology, University of Florida, 1115 Turlington Hall, P.O. Box 117305, Gainesville, FL 32611-7305, USA
- Genetics Institute, University of Florida, 2033 Mowry Rd, P.O. Box 103610, Gainesville, FL 32610-3610, USA
- Department of Biology, University of Florida, 220 Bartram Hall, P.O. Box 118525, Gainesville, FL 32611-8525, USA
| | - Felicien Masanga Maisha
- Department of Anthropology, University of Florida, 1115 Turlington Hall, P.O. Box 117305, Gainesville, FL 32611-7305, USA
- Genetics Institute, University of Florida, 2033 Mowry Rd, P.O. Box 103610, Gainesville, FL 32610-3610, USA
- HEAL Africa Hospital, Rue Lyn Lusi No. 111, Goma BP 319, Democratic Republic of the Congo
| | - Edward B Quinn
- Department of Anthropology, University of Florida, 1115 Turlington Hall, P.O. Box 117305, Gainesville, FL 32611-7305, USA
- Genetics Institute, University of Florida, 2033 Mowry Rd, P.O. Box 103610, Gainesville, FL 32610-3610, USA
| | - Katherine Liceth Morales
- Department of Anthropology, University of Florida, 1115 Turlington Hall, P.O. Box 117305, Gainesville, FL 32611-7305, USA
- Genetics Institute, University of Florida, 2033 Mowry Rd, P.O. Box 103610, Gainesville, FL 32610-3610, USA
| | - Julie M Moore
- Department of Infectious Diseases & Immunology, College of Veterinary Medicine, University of Florida, Room V3-111B, P.O. Box 110880, Gainesville, FL 32611-4111, USA
| | - Connie J Mulligan
- Department of Anthropology, University of Florida, 1115 Turlington Hall, P.O. Box 117305, Gainesville, FL 32611-7305, USA
- Genetics Institute, University of Florida, 2033 Mowry Rd, P.O. Box 103610, Gainesville, FL 32610-3610, USA
| |
Collapse
|
36
|
Altered gastrointestinal tract structure and microbiome following cerebral malaria infection. Parasitol Res 2023; 122:789-799. [PMID: 36602586 DOI: 10.1007/s00436-022-07775-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 12/20/2022] [Indexed: 01/06/2023]
Abstract
Cerebral malaria (CM) is the most severe form of malaria with the highest mortality rate and can result in life-long neurological deficits and ongoing comorbidities. Factors contributing to severity of infection and development of CM are not fully elucidated. Recent studies have indicated a key role of the gut microbiome in a range of health conditions that affect the brain, but limited microbiome research has been conducted in the context of malaria. To address this knowledge gap, the impact of CM on the gut microbiome was investigated in mice. C57BL/6J mice were infected with Plasmodium berghei ANKA (PbA) parasites and compared to non-infected controls. Microbial DNA from faecal pellets collected daily for 6-days post-infection were extracted, and microbiome comparisons conducted using 16S rRNA profiling. We identified significant differences in the composition of bacterial communities between the infected and the non-infected groups, including a higher abundance of the genera Akkermansia, Alistipes and Alloprevotella in PbA-infected mice. Furthermore, intestinal samples were collected post-cull for morphological analysis. We determined that the caecal weight was significantly lower, and the small intestine was significantly longer in PbA-infected mice than in the non-infected controls. We concluded that changes in microbial community composition were primarily driven by the infection protocol and, to a lesser extent, by the time of infection. Our findings pave the way for a new area of research and novel intervention strategies to modulate the severity of cerebral malaria disease.
Collapse
|
37
|
Fitri LE, Sardjono TW, Winaris N, Pawestri AR, Endharti AT, Norahmawati E, Handayani D, Kurniawan SN, Azizah S, Alifia LI, Asiyah R, Ayuningtyas TR. Bifidobacterium longum Administration Diminishes Parasitemia and Inflammation During Plasmodium berghei Infection in Mice. J Inflamm Res 2023; 16:1393-1404. [PMID: 37006809 PMCID: PMC10065020 DOI: 10.2147/jir.s400782] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 02/17/2023] [Indexed: 03/28/2023] Open
Abstract
Purpose During Plasmodium berghei (P. berghei) infection, infected erythrocytes are sequestered in gut tissues through microvascular circulation, leading to dysbiosis. This study aimed to investigate the effect of Lactobacillus casei (L. casei) and Bifidobacterium longum (B. longum) administration on the parasitemia level, gut microbiota composition, expression of cluster of differentiation 103 (CD103) in intestinal dendritic and T regulatory cells (T reg), plasma interferon gamma (IFN-γ) and tumor necrosis factor (TNF-α) levels in P. berghei infected mice. Methods P. berghei was inoculated intraperitoneally. Infected mice were randomly divided into 5 groups and treated with either L. casei, B. longum, or the combination of both for 5 days before up to 6 days post-infection (p.i). The control group was treated with phosphate-buffered saline (PBS), while uninfected mice were used as negative control. Levels of CD103 and forkhead box P3 (FoxP3) expression were measured by direct immunofluorescense, while plasma IFN-γ and TNF-α level were determined using enzyme-linked immunosorbent assay (ELISA). Results All treated groups showed an increase in parasitemia from day 2 to day 6 p.i, which was significant at day 2 p.i (p = 0.001), with the group receiving B. longum displaying the lowest degree of parasitemia. Significant reduction in plasma IFN-γ and TNF-α levels was observed in the group receiving B. longum (p = 0.022 and p = 0.026, respectively). The CD103 and FoxP3 expression was highest in the group receiving B. longum (p = 0.01 and p = 0.02, respectively). Conclusion B. longum showed the best protective effect against Plasmodium infection by reducing the degree of parasitemia and modulating the gut immunity. This provides a basis for further research involving probiotic supplementation in immunity modulation of infectious diseases.
Collapse
Affiliation(s)
- Loeki Enggar Fitri
- Department of Parasitology, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
- AIDS, Toxoplasma, Opportunistic Disease, and Malaria (ATOM) Research Group, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
| | - Teguh Wahju Sardjono
- Department of Parasitology, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
- AIDS, Toxoplasma, Opportunistic Disease, and Malaria (ATOM) Research Group, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
- Correspondence: Teguh Wahju Sardjono, Department of Parasitology, Faculty of Medicine Universitas Brawijaya, Jl. Veteran, Malang, 65145, Indonesia, Tel +62 341 569117, Fax +62 341 564755, Email
| | - Nuning Winaris
- Department of Parasitology, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
- AIDS, Toxoplasma, Opportunistic Disease, and Malaria (ATOM) Research Group, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
| | - Aulia Rahmi Pawestri
- Department of Parasitology, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
- AIDS, Toxoplasma, Opportunistic Disease, and Malaria (ATOM) Research Group, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
| | - Agustina Tri Endharti
- Department of Parasitology, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
| | - Eviana Norahmawati
- Department of Anatomical Pathology, Dr. Saiful Anwar Hospital/Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
| | - Dian Handayani
- Department of Nutrition, Faculty of Health Sciences, Universitas Brawijaya, Malang, Indonesia
| | | | - Syafiatul Azizah
- Master Program in Biomedical Sciences, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
| | - Lustyafa Inassani Alifia
- Master Program in Biomedical Sciences, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
- Department of Parasitology, Faculty of Medicine, University of Muhammadiyah Malang, Malang, Indonesia
| | - Rokhmatul Asiyah
- Master Program in Biomedical Sciences, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
- State University of Malang, Malang, Indonesia
| | - Tita Rachma Ayuningtyas
- Master Program in Biomedical Sciences, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
| |
Collapse
|
38
|
Hu X, Zhao J, Zhao J, Yang E, Jia M. Genome-wide liver transcriptomic profiling of a malaria mouse model reveals disturbed immune and metabolic responses. Parasit Vectors 2023; 16:40. [PMID: 36717945 PMCID: PMC9885691 DOI: 10.1186/s13071-023-05672-w] [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: 11/10/2022] [Accepted: 01/16/2023] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND The liver is responsible for a range of functions in vertebrates, such as metabolism and immunity. In malaria, the liver plays a crucial role in the interaction between the parasite and host. Although malarial hepatitis is a common clinical complication of severe malaria, other malaria-related liver changes have been overlooked during the blood stage of the parasite life-cycle, in contrast to the many studies that have focused on parasite invasion of and replication in the liver during the hepatic stage of the parasite. METHODS A rodent model of malaria was established using Plasmodium yoelii strain 17XL, a lethal strain of rodent malaria, for liver transcriptomic profiling. RESULTS Differentially expressed messenger RNAs were associated with innate and adaptive immune responses, while differentially expressed long noncoding RNAs were enriched in the regulation of metabolism-related pathways, such as lipid metabolism. The coexpression network showed that host genes were related to cellular transport and tissue remodeling. Hub gene analysis of P. yoelii indicated that ubiquitination genes that were coexpressed with the host were evolutionarily conserved. CONCLUSIONS Our analysis yielded evidence of activated immune responses, aberrant metabolic processes and tissue remodeling changes in the livers of mice with malaria during the blood stage of the parasite, which provided a systematic outline of liver responses during Plasmodium infection.
Collapse
Affiliation(s)
- Xueyan Hu
- grid.11135.370000 0001 2256 9319Department of Medical Bioinformatics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191 China
| | - Jie Zhao
- grid.11135.370000 0001 2256 9319Department of Microbiology & Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191 China
| | - Junhui Zhao
- grid.11135.370000 0001 2256 9319Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191 China
| | - Ence Yang
- grid.11135.370000 0001 2256 9319Department of Medical Bioinformatics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191 China ,grid.11135.370000 0001 2256 9319Department of Microbiology & Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191 China
| | - Mozhi Jia
- grid.11135.370000 0001 2256 9319Department of Microbiology & Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191 China
| |
Collapse
|
39
|
Farinella DN, Kaur S, Tran V, Cabrera-Mora M, Joyner CJ, Lapp SA, Pakala SB, Nural MV, DeBarry JD, Kissinger JC, Jones DP, Moreno A, Galinski MR, Cordy RJ. Malaria disrupts the rhesus macaque gut microbiome. Front Cell Infect Microbiol 2023; 12:1058926. [PMID: 36710962 PMCID: PMC9880479 DOI: 10.3389/fcimb.2022.1058926] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 12/12/2022] [Indexed: 01/14/2023] Open
Abstract
Previous studies have suggested that a relationship exists between severity and transmissibility of malaria and variations in the gut microbiome, yet only limited information exists on the temporal dynamics of the gut microbial community during a malarial infection. Here, using a rhesus macaque model of relapsing malaria, we investigate how malaria affects the gut microbiome. In this study, we performed 16S sequencing on DNA isolated from rectal swabs of rhesus macaques over the course of an experimental malarial infection with Plasmodium cynomolgi and analyzed gut bacterial taxa abundance across primary and relapsing infections. We also performed metabolomics on blood plasma from the animals at the same timepoints and investigated changes in metabolic pathways over time. Members of Proteobacteria (family Helicobacteraceae) increased dramatically in relative abundance in the animal's gut microbiome during peak infection while Firmicutes (family Lactobacillaceae and Ruminococcaceae), Bacteroidetes (family Prevotellaceae) and Spirochaetes amongst others decreased compared to baseline levels. Alpha diversity metrics indicated decreased microbiome diversity at the peak of parasitemia, followed by restoration of diversity post-treatment. Comparison with healthy subjects suggested that the rectal microbiome during acute malaria is enriched with commensal bacteria typically found in the healthy animal's mucosa. Significant changes in the tryptophan-kynurenine immunomodulatory pathway were detected at peak infection with P. cynomolgi, a finding that has been described previously in the context of P. vivax infections in humans. During relapses, which have been shown to be associated with less inflammation and clinical severity, we observed minimal disruption to the gut microbiome, despite parasites being present. Altogether, these data suggest that the metabolic shift occurring during acute infection is associated with a concomitant shift in the gut microbiome, which is reversed post-treatment.
Collapse
Affiliation(s)
| | - Sukhpreet Kaur
- Department of Biology, Wake Forest University, Winston-Salem, NC, United States
| | - ViLinh Tran
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, School of Medicine, Emory University, Atlanta, GA, United States
| | - Monica Cabrera-Mora
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, School of Medicine, Emory University, Atlanta, GA, United States,Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States
| | - Chester J. Joyner
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States,Department of Infectious Diseases, University of Georgia, Athens, GA, United States
| | - Stacey A. Lapp
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, School of Medicine, Emory University, Atlanta, GA, United States,Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States
| | - Suman B. Pakala
- Institute of Bioinformatics, University of Georgia, Athens, GA, United States
| | - Mustafa V. Nural
- Institute of Bioinformatics, University of Georgia, Athens, GA, United States
| | - Jeremy D. DeBarry
- Institute of Bioinformatics, University of Georgia, Athens, GA, United States,Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, United States,Emory Vaccine Center, Emory University, Atlanta, GA, United States
| | - Jessica C. Kissinger
- Institute of Bioinformatics, University of Georgia, Athens, GA, United States,Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, United States,Department of Genetics, University of Georgia, Athens, GA, United States
| | - Dean P. Jones
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, School of Medicine, Emory University, Atlanta, GA, United States
| | - Alberto Moreno
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, School of Medicine, Emory University, Atlanta, GA, United States,Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States,Division of Infectious Diseases, Department of Medicine, Emory University, Atlanta, GA, United States
| | - Mary R. Galinski
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, School of Medicine, Emory University, Atlanta, GA, United States,Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States,Division of Infectious Diseases, Department of Medicine, Emory University, Atlanta, GA, United States
| | - Regina Joice Cordy
- Department of Biology, Wake Forest University, Winston-Salem, NC, United States,Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States,*Correspondence: Regina Joice Cordy,
| |
Collapse
|
40
|
Liang H, Zhang L, Hoden B, Qu B, Derubeis D, Song X, Zhang D. Delineating the Role of Toll-Like Receptors in Inflammatory Bowel Disease. Methods Mol Biol 2023; 2700:221-228. [PMID: 37603184 DOI: 10.1007/978-1-0716-3366-3_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/22/2023]
Abstract
Toll-like receptors (TLRs) recognize altered gut microbiota triggering an immune response. These responses play a critical role in the pathogenesis and treatment of inflammatory bowel disease (IBD). IBD is characterized by inflammation of the intestinal tracts as in Crohn's disease and ulcerative colitis. However, one challenge in determining the role of a specific TLR in IBD and its underlying mechanism is disparity. Variance in age, gender, race, and ethnicity shows a dramatic difference in the disease incidence, severity, and response to treatment. Delineating the role of TLRs in IBD relies on both a knockout mouse and a disease model. Here, we describe a detailed protocol on how to use nearly identical genetic backgrounds of TLR wild-type and knockout littermate mice in a dextran sodium sulfate (DSS)-induced colitis model.
Collapse
Affiliation(s)
- Hongbin Liang
- Center for Infectious and Inflammatory Diseases, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, USA
| | - Lin Zhang
- Center for Infectious and Inflammatory Diseases, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, USA
| | - Bettina Hoden
- Center for Infectious and Inflammatory Diseases, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, USA
| | - Bo Qu
- Department of Gastroenterology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - David Derubeis
- Center for Infectious and Inflammatory Diseases, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, USA
| | - Xiaotong Song
- Center for Infectious and Inflammatory Diseases, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, USA.
| | - Dekai Zhang
- Center for Infectious and Inflammatory Diseases, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, USA.
- Department of Translational Medical Sciences, College of Medicine, Texas A&M University, Houston, TX, USA.
| |
Collapse
|
41
|
Piazzesi A, Pane S, Putignani L. How Modulations of the Gut Microbiota May Help in Preventing or Treating Parasitic Diseases. CURRENT TROPICAL MEDICINE REPORTS 2022. [DOI: 10.1007/s40475-022-00275-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
42
|
Alloo J, Leleu I, Grangette C, Pied S. Parasite infections, neuroinflammation, and potential contributions of gut microbiota. Front Immunol 2022; 13:1024998. [PMID: 36569929 PMCID: PMC9772015 DOI: 10.3389/fimmu.2022.1024998] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 11/21/2022] [Indexed: 12/12/2022] Open
Abstract
Many parasitic diseases (including cerebral malaria, human African trypanosomiasis, cerebral toxoplasmosis, neurocysticercosis and neuroschistosomiasis) feature acute or chronic brain inflammation processes, which are often associated with deregulation of glial cell activity and disruption of the brain blood barrier's intactness. The inflammatory responses of astrocytes and microglia during parasite infection are strongly influenced by a variety of environmental factors. Although it has recently been shown that the gut microbiota influences the physiology and immunomodulation of the central nervous system in neurodegenerative diseases like Alzheimer's disease and Parkinson's, the putative link in parasite-induced neuroinflammatory diseases has not been well characterized. Likewise, the central nervous system can influence the gut microbiota. In parasite infections, the gut microbiota is strongly perturbed and might influence the severity of the central nervous system inflammation response through changes in the production of bacterial metabolites. Here, we review the roles of astrocytes and microglial cells in the neuropathophysiological processes induced by parasite infections and their possible regulation by the gut microbiota.
Collapse
Affiliation(s)
| | | | | | - Sylviane Pied
- Center for Infection and Immunity of Lille-CIIL, Centre National de la Recherche Scientifique-CNRS UMR 9017-Institut National de la Recherche Scientifique et Médicale-Inserm U1019, Institut Pasteur de Lille, Univ. Lille, Lille, France
| |
Collapse
|
43
|
Donkersley P, Rice A, Graham RI, Wilson K. Gut microbial community supplementation and reduction modulates African armyworm susceptibility to a baculovirus. FEMS Microbiol Ecol 2022; 99:6880154. [PMID: 36473704 PMCID: PMC9764207 DOI: 10.1093/femsec/fiac147] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 11/01/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
Gut microbiota stimulates the immune system and inhibits pathogens, and thus, it is critical for disease prevention. Probiotics represent an effective alternative to antibiotics used for the therapy and prevention of bacterial diseases. Probiotic bacteria are commonly used in vertebrates, although their use in invertebrates is still rare. We manipulated the gut microbiome of the African Armyworm (Spodoptera exempta Walker) using antibiotics and field-collected frass, in an attempt to understand the interactions of the gut microbiome with the nucleopolyhedrovirus, SpexNPV. We found that S. exempta individuals with supplemented gut microbiome were significantly more resistant to SpexNPV, relative to those with a typical laboratory gut microbiome. Illumina MiSeq sequencing revealed the bacterial phyla in the S. exempta gut belonged to 28 different classes. Individuals with an increased abundance of Lactobacillales had a higher probability of surviving viral infection. In contrast, there was an increased abundance of Enterobacteriales and Pseudomonadales in individuals dying from viral infection, corresponding with decreased abundance of these two Orders in surviving caterpillars, suggesting a potential role for them in modulating the interaction between the host and its pathogen. These results have important implications for laboratory studies testing biopesticides.
Collapse
Affiliation(s)
- Philip Donkersley
- Corresponding author: Lancaster Environment Centre, Lancaster University, Lancaster LA1 4YQ, United Kingdom. E-mail:
| | - Annabel Rice
- Lancaster Environment Centre, Lancaster University, Lancaster LA1 4YQ, United Kingdom
| | - Robert I Graham
- Department of Rural Land Use, SRUC, Craibstone Campus, Aberdeen AB21 9YA, United Kingdom
| | - Kenneth Wilson
- Lancaster Environment Centre, Lancaster University, Lancaster LA1 4YQ, United Kingdom
| |
Collapse
|
44
|
Zhou S, Lu Y, Chen J, Pan Z, Pang L, Wang Y, Zhang Q, Strand MR, Chen XX, Huang J. Parasite reliance on its host gut microbiota for nutrition and survival. THE ISME JOURNAL 2022; 16:2574-2586. [PMID: 35941172 PMCID: PMC9561699 DOI: 10.1038/s41396-022-01301-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 07/20/2022] [Accepted: 07/22/2022] [Indexed: 11/12/2022]
Abstract
Studying the microbial symbionts of eukaryotic hosts has revealed a range of interactions that benefit host biology. Most eukaryotes are also infected by parasites that adversely affect host biology for their own benefit. However, it is largely unclear whether the ability of parasites to develop in hosts also depends on host-associated symbionts, e.g., the gut microbiota. Here, we studied the parasitic wasp Leptopilina boulardi (Lb) and its host Drosophila melanogaster. Results showed that Lb successfully develops in conventional hosts (CN) with a gut microbiota but fails to develop in axenic hosts (AX) without a gut microbiota. We determined that developing Lb larvae consume fat body cells that store lipids. We also determined that much larger amounts of lipid accumulate in fat body cells of parasitized CN hosts than parasitized AX hosts. CN hosts parasitized by Lb exhibited large increases in the abundance of the bacterium Acetobacter pomorum in the gut, but did not affect the abundance of Lactobacillus fructivorans which is another common member of the host gut microbiota. However, AX hosts inoculated with A. pomorum and/or L. fructivorans did not rescue development of Lb. In contrast, AX larvae inoculated with A. pomorum plus other identified gut community members including a Bacillus sp. substantially rescued Lb development. Rescue was further associated with increased lipid accumulation in host fat body cells. Insulin-like peptides increased in brain neurosecretory cells of parasitized CN larvae. Lipid accumulation in the fat body of CN hosts was further associated with reduced Bmm lipase activity mediated by insulin/insulin-like growth factor signaling (IIS). Altogether, our results identify a previously unknown role for the gut microbiota in defining host permissiveness for a parasite. Our findings also identify a new paradigm for parasite manipulation of host metabolism that depends on insulin signaling and the gut microbiota.
Collapse
Affiliation(s)
- Sicong Zhou
- Institute of Insect Sciences, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, 310058, China
- Ministry of Agriculture Key Lab of Molecular Biology of Crop Pathogens and Insect Pests, Zhejiang University, Hangzhou, 310058, China
- Key Laboratory of Biology of Crop Pathogens and Insects of Zhejiang Province, Zhejiang University, Hangzhou, 310058, China
| | - Yueqi Lu
- Institute of Insect Sciences, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, 310058, China
- Ministry of Agriculture Key Lab of Molecular Biology of Crop Pathogens and Insect Pests, Zhejiang University, Hangzhou, 310058, China
- Key Laboratory of Biology of Crop Pathogens and Insects of Zhejiang Province, Zhejiang University, Hangzhou, 310058, China
| | - Jiani Chen
- Institute of Insect Sciences, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, 310058, China
- Ministry of Agriculture Key Lab of Molecular Biology of Crop Pathogens and Insect Pests, Zhejiang University, Hangzhou, 310058, China
- Key Laboratory of Biology of Crop Pathogens and Insects of Zhejiang Province, Zhejiang University, Hangzhou, 310058, China
| | - Zhongqiu Pan
- Institute of Insect Sciences, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, 310058, China
- Ministry of Agriculture Key Lab of Molecular Biology of Crop Pathogens and Insect Pests, Zhejiang University, Hangzhou, 310058, China
- Key Laboratory of Biology of Crop Pathogens and Insects of Zhejiang Province, Zhejiang University, Hangzhou, 310058, China
| | - Lan Pang
- Institute of Insect Sciences, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, 310058, China
- Ministry of Agriculture Key Lab of Molecular Biology of Crop Pathogens and Insect Pests, Zhejiang University, Hangzhou, 310058, China
- Key Laboratory of Biology of Crop Pathogens and Insects of Zhejiang Province, Zhejiang University, Hangzhou, 310058, China
| | - Ying Wang
- Institute of Insect Sciences, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, 310058, China
- Ministry of Agriculture Key Lab of Molecular Biology of Crop Pathogens and Insect Pests, Zhejiang University, Hangzhou, 310058, China
- Key Laboratory of Biology of Crop Pathogens and Insects of Zhejiang Province, Zhejiang University, Hangzhou, 310058, China
| | - Qichao Zhang
- Institute of Insect Sciences, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, 310058, China
- Ministry of Agriculture Key Lab of Molecular Biology of Crop Pathogens and Insect Pests, Zhejiang University, Hangzhou, 310058, China
- Key Laboratory of Biology of Crop Pathogens and Insects of Zhejiang Province, Zhejiang University, Hangzhou, 310058, China
| | - Michael R Strand
- Department of Entomology, University of Georgia, Athens, GA, 30602, USA.
| | - Xue-Xin Chen
- Institute of Insect Sciences, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, 310058, China.
- Ministry of Agriculture Key Lab of Molecular Biology of Crop Pathogens and Insect Pests, Zhejiang University, Hangzhou, 310058, China.
- Key Laboratory of Biology of Crop Pathogens and Insects of Zhejiang Province, Zhejiang University, Hangzhou, 310058, China.
- State Key Lab of Rice Biology, Zhejiang University, Hangzhou, 310058, China.
| | - Jianhua Huang
- Institute of Insect Sciences, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, 310058, China.
- Ministry of Agriculture Key Lab of Molecular Biology of Crop Pathogens and Insect Pests, Zhejiang University, Hangzhou, 310058, China.
- Key Laboratory of Biology of Crop Pathogens and Insects of Zhejiang Province, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
45
|
An Overview on the Impact of Microbiota on Malaria Transmission and Severity: Plasmodium-Vector-Host Axis. Acta Parasitol 2022; 67:1471-1486. [PMID: 36264525 DOI: 10.1007/s11686-022-00631-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 10/10/2022] [Indexed: 11/01/2022]
Abstract
PURPOSE Malaria, which is a vector-borne disease caused by Plasmodium sp., continue to become a serious threat, causing more than 600,000 deaths annually, especially in developing countries. Due to the lack of a long-term, and effective vaccine, and an increasing resistance to antimalarials, new strategies are needed for prevention and treatment of malaria. Recently, the impact of microbiota on development and transmission of Plasmodium, and the severity of malaria has only begun to emerge, although its contribution to homeostasis and a wide variety of disorders is well-understood. Further evidence has shown that microbiota of both mosquito and human host play important roles in transmission, progression, and clearance of Plasmodium infection. Furthermore, Plasmodium can cause significant alterations in the host and mosquito gut microbiota, affecting the clinical outcome of malaria. METHODOLOGY In this review, we attempt to summarize results from published studies on the influence of the host microbiota on the outcome of Plasmodium infections in both arthropods and mammalian hosts. CONCLUSION Modifications of microbiota may be an important potential strategy in blocking Plasmodium transmission in vectors and in the diagnosis, treatment, and prevention of malaria in humans in the future.
Collapse
|
46
|
The Role of the Microbiome in Pancreatic Cancer. Cancers (Basel) 2022; 14:cancers14184479. [PMID: 36139638 PMCID: PMC9496841 DOI: 10.3390/cancers14184479] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 09/07/2022] [Accepted: 09/09/2022] [Indexed: 11/29/2022] Open
Abstract
Simple Summary Pancreatic cancer is deadly cancer characterized by dense stroma creating an immunosuppressive tumor microenvironment. Accumulating evidences indicate that the microbiome plays an important role in pancreatic cancer development and progression via the local and systemic inflammation and immune responses. The alteration of the microbiome modulates the tumor microenvironment and immune system in pancreatic cancer, which affects the efficacy of chemotherapies including immune-targeted therapies. Understanding the role of microbiome and underlying mechanisms may lead to novel biomarkers and therapeutic strategies for pancreatic cancer. This review summarizes the current evidence on the role of the microbiome in pancreatic cancer. Abstract Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal malignancies, with little improvement in outcomes in recent decades, although the molecular and phenotypic characterization of PDAC has contributed to advances in tailored therapies. PDAC is characterized by dense stroma surrounding tumor cells, which limits the efficacy of treatment due to the creation of a physical barrier and immunosuppressive environment. Emerging evidence regarding the microbiome in PDAC implies its potential role in the initiation and progression of PDAC. However, the underlying mechanisms of how the microbiome affects the local tumor microenvironment (TME) as well as the systemic immune system have not been elucidated in PDAC. In addition, therapeutic strategies based on the microbiome have not been established. In this review, we summarize the current evidence regarding the role of the microbiome in the development of PDAC and discuss a possible role for the microbiome in the early detection of PDAC in relation to premalignant pancreatic diseases, such as chronic pancreatitis and intraductal papillary mucinous neoplasm (IPMN). In addition, we discuss the potential role of the microbiome in the treatment of PDAC, especially in immunotherapy, although the biomarkers used to predict the efficacy of immunotherapy in PDAC are still unknown. A comprehensive understanding of tumor-associated immune responses, including those involving the microbiome, holds promise for new treatments in PDAC.
Collapse
|
47
|
Olatunde AC, Cornwall DH, Roedel M, Lamb TJ. Mouse Models for Unravelling Immunology of Blood Stage Malaria. Vaccines (Basel) 2022; 10:1525. [PMID: 36146602 PMCID: PMC9501382 DOI: 10.3390/vaccines10091525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 09/05/2022] [Accepted: 09/06/2022] [Indexed: 11/16/2022] Open
Abstract
Malaria comprises a spectrum of disease syndromes and the immune system is a major participant in malarial disease. This is particularly true in relation to the immune responses elicited against blood stages of Plasmodium-parasites that are responsible for the pathogenesis of infection. Mouse models of malaria are commonly used to dissect the immune mechanisms underlying disease. While no single mouse model of Plasmodium infection completely recapitulates all the features of malaria in humans, collectively the existing models are invaluable for defining the events that lead to the immunopathogenesis of malaria. Here we review the different mouse models of Plasmodium infection that are available, and highlight some of the main contributions these models have made with regards to identifying immune mechanisms of parasite control and the immunopathogenesis of malaria.
Collapse
Affiliation(s)
| | | | | | - Tracey J. Lamb
- Department of Pathology, University of Utah, Emma Eccles Jones Medical Research Building, 15 N Medical Drive E, Room 1420A, Salt Lake City, UT 84112, USA
| |
Collapse
|
48
|
Xie Y, Guan W, Zhao Y, Yan S, Guo K, Chen S, Hu X, Shi H, Li J. Deficiency of migration inhibitory factor influences the gut microbiota of C57BL/6 mice infected with Plasmodium berghei ANKA. Front Microbiol 2022; 13:978644. [PMID: 36033889 PMCID: PMC9412183 DOI: 10.3389/fmicb.2022.978644] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 07/25/2022] [Indexed: 12/02/2022] Open
Abstract
Cerebral malaria (CM), as one of the most common complications in severe malaria, has threatened millions of individuals’ neurological health and even their lives. Macrophage migration inhibitory factor (MIF), a pleiotropic proinflammatory factor in humans, seems to be a risk factor for death in patients with CM, but its functional mechanism remains unclear. To verify whether affecting the intestinal microbes of the host was one of the mechanisms by which MIF regulates CM, C57BL/6 mice, including WT + PbA, MIF-KO + PbA and their uninfected controls, were sent for 16S rRNA-based sequencing targeting the V4 region of the intestinal microbiota through the Illumina MiSeq platform. The results showed that OTU clustering, alpha and beta diversity in the four groups involved had evident variation. The relative abundance at different taxonomic levels, especially the dominant intestinal flora, was obviously changed. The LEfSe analysis screened out several biomarkers, including significantly reduced Ligilactobacillus (Lactobacillus murinus) in WPbA mice compared to the WT group and Akkermansia (Akkermansia_muciniphila) in KPbA mice compared to the WPbA group. For MIF KO groups, mice infected with PbA or uninfected showed significant enrichment of producers of short-chain fatty acids, including Dubosiella and Faecalibaculum (Faecalibaculum rodentium) in KPbA, and Lachnospiraceae_NK4A136_group and Firmicutes_bacterium_M10-2 in KO. This study not only further proved the gut microbiota changes in C57BL/6 mice caused by PbA infection, but also found that MIF deletion directly affected the changes in the gut microbiota of C57BL/6 mice before and after PbA infection. This finding reveals a potential mechanism by which MIF regulates CM. Combining MIF with potential microbial biomarkers will provide a promising idea to develop combined drugs for improving CM in the future.
Collapse
|
49
|
Wang Y, Li X, Chen X, Kulyar MFEA, Duan K, Li H, Bhutta ZA, Wu Y, Li K. Gut Fungal Microbiome Responses to Natural Cryptosporidium Infection in Horses. Front Microbiol 2022; 13:877280. [PMID: 35875530 PMCID: PMC9298756 DOI: 10.3389/fmicb.2022.877280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 05/18/2022] [Indexed: 11/13/2022] Open
Abstract
It is critical to characterize changes in the structure and composition of the host fungal community in natural Cryptosporidium infection, because it gives the possible overview of gut microbiome in host homeostasis and disease progression. A total of 168 rectal fecal samples were collected and examined using nPCR. The positive samples were double-checked using 18S rDNA high-throughput sequencing. After confirmation, ITS high-throughput sequencing was utilized to investigate the fungal community’s response to natural Cryptosporidium infection. Results showed that a total of three positive samples (1.79%) were identified with an increased abundance of fungi associated with health hazards, such as class Dothideomycetes, families, i.e., Cladosporiaceae, Glomerellaceae, and genera, i.e., Wickerhamomyces, Talaromyces, Cladosporium, Dactylonectria, and Colletotrichum. On the contrary, taxa associated with favorable physiological effects on the host were shown to have the reverse impact, such as families, i.e., Psathyrellaceae, Pseudeurotiaceae and genera (Beauveria, Nigrospora, and Diversispora). For the first time, we evaluated the condition of natural Cryptosporidium infection in horses in Wuhan, China, and discovered distinct variations in the fungal microbiome in response to natural infection. It might prompt a therapy or prevention strategy to apply specific fungal microorganisms that are probably responsible for decreased susceptibility or increased resistance to infection.
Collapse
Affiliation(s)
- Yaping Wang
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Xuwen Li
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Xiushuang Chen
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | | | - Kun Duan
- China Tobacco Henan Industrial Co., Ltd., Zhengzhou, China
| | - Huade Li
- Sichuan Academy of Grassland Science, Chengdu, China
| | - Zeeshan Ahmad Bhutta
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, South Korea
| | - Yi Wu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Kun Li
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
50
|
Mukherjee D, Chora ÂF, Lone JC, Ramiro RS, Blankenhaus B, Serre K, Ramirez M, Gordo I, Veldhoen M, Varga-Weisz P, Mota MM. Host lung microbiota promotes malaria-associated acute respiratory distress syndrome. Nat Commun 2022; 13:3747. [PMID: 35768411 PMCID: PMC9243033 DOI: 10.1038/s41467-022-31301-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 06/13/2022] [Indexed: 11/14/2022] Open
Abstract
Severe malaria can manifest itself with a variety of well-recognized clinical phenotypes that are highly predictive of death - severe anaemia, coma (cerebral malaria), multiple organ failure, and respiratory distress. The reasons why an infected individual develops one pathology rather than another remain poorly understood. Here we use distinct rodent models of infection to show that the host microbiota is a contributing factor for the development of respiratory distress syndrome and host mortality in the context of malaria infections (malaria-associated acute respiratory distress syndrome, MA-ARDS). We show that parasite sequestration in the lung results in sustained immune activation. Subsequent production of the anti-inflammatory cytokine IL-10 by T cells compromises microbial control, leading to severe lung disease. Notably, bacterial clearance with linezolid, an antibiotic commonly used in the clinical setting to control lung-associated bacterial infections, prevents MA-ARDS-associated lethality. Thus, we propose that the host's anti-inflammatory response to limit tissue damage can result in loss of microbial control, which promotes MA-ARDS. This must be considered when intervening against life-threatening respiratory complications.
Collapse
Affiliation(s)
- Debanjan Mukherjee
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028, Lisboa, Portugal
| | - Ângelo Ferreira Chora
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028, Lisboa, Portugal
| | - Jean-Christophe Lone
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028, Lisboa, Portugal
- School of Life Sciences, University of Essex, Colchester, UK
| | | | - Birte Blankenhaus
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028, Lisboa, Portugal
| | - Karine Serre
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028, Lisboa, Portugal
| | - Mário Ramirez
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028, Lisboa, Portugal
| | - Isabel Gordo
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Marc Veldhoen
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028, Lisboa, Portugal
| | - Patrick Varga-Weisz
- School of Life Sciences, University of Essex, Colchester, UK
- São Paulo Excellence Chair, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Maria M Mota
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028, Lisboa, Portugal.
| |
Collapse
|