1
|
Van Nederveen V, Johnson YS, Ortega E, Soc A, Smith MA, Melton-Celsa AR. Role of aggregative adherence fimbriae from enteroaggregative Escherichia coli isolates in biofilm and colonization. Microb Pathog 2025; 203:107444. [PMID: 40032001 DOI: 10.1016/j.micpath.2025.107444] [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: 08/08/2024] [Revised: 01/10/2025] [Accepted: 02/28/2025] [Indexed: 03/05/2025]
Abstract
Enteroaggregative Escherichia coli (EAEC) are a diverse group of bacteria that cause diarrhea worldwide. EAEC significantly affect travelers to endemic regions, including military personnel, and children in developing countries where EAEC infection is associated with childhood failure-to-thrive. EAEC creates thick biofilms on the intestinal mucosa, a process that is thought to contribute to the development of both diarrhea and childhood failure-to-thrive. Typical EAEC strains encode and produce just one aggregative adherence fimbriae (AAF) out of the five different AAF types. The AAF are required for aggregative adherence to epithelial cells in vitro, but the degree of importance of each of the AAF types in both biofilm formation and pathogenesis is unknown. In this study, we investigated the role of the fimbriae in EAEC biofilms by deleting the major fimbrial subunit gene for the AAF from each of the five AAF categories and observing the impact on biofilm staining from recent EAEC clinical isolates. We found that biofilm was significantly reduced in all strains when the AAF gene was deleted, and that the defect could be overcome by complementation. In this work we also describe a modified murine EAEC model appropriate for colonization studies. In an antibiotic-treated mouse colonization model, some AAF mutant strains were attenuated for colonization, including AAF/II, AAF/IV, and AAF/V isolates. We did not observe complementation of the attenuated colonization phenotype in the mouse model. However, since we found a colonization defect for several EAEC mutant strains of different AAF types, a link between the fimbriae and colonization in the mice is supported. Taken together, our results show that the AAF are required for biofilm formation, and that some AAF contribute to colonization in a mouse model.
Collapse
Affiliation(s)
- Viktoria Van Nederveen
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA
| | - Yuliya Seldina Johnson
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA
| | - Ennzo Ortega
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA
| | - Anthony Soc
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA
| | | | - Angela R Melton-Celsa
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD, USA.
| |
Collapse
|
2
|
Sebastiano M, Chastel O, Eens M, Costantini D. Gene expression provides mechanistic insights into a viral disease in seabirds. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 957:177478. [PMID: 39528216 DOI: 10.1016/j.scitotenv.2024.177478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 11/05/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
Wild animals are exposed to a variety of anthropogenic stressors that may result in loss of physiological homeostasis. One main consequence of this stress exposure is the increased vulnerability to pathogens. We addressed the hypothesis that energetic unbalance and alterations of immune effectors are key proximate mechanisms underlying this vulnerability, by quantifying the gene expression of magnificent frigatebird Fregata magnificens chicks affected by a highly lethal viral disease, whose appearance is favoured by food limitation in this species. A comparison between chicks with and without visible clinical signs of the disease using strict threshold of significance (p-value adjusted<0.05 and log2 fold-change above 1 or below -1) revealed 86 upregulated and 9 downregulated genes in sick chicks. The main differentially expressed genes with several fold difference between healthy and sick chicks were linked to biotic and external stimuli, inflammation and antifungal/antibacterial activity, signaling, and hydrolase activity. We further followed the chicks for several weeks, to identify chicks that became sick over the course of the study, to assess how the gene expression profile of chicks may predict the response to the disease. A comparison between chicks that remained always healthy and chicks that showed the appearance of visible clinical signs of the disease revealed 4 upregulated and 8 downregulated genes in chicks that became sick. The main differentially expressed genes with several fold difference between the two phenotypes were linked to cell development and differentiation, metabolism, and immunity. The results of our study suggest that alterations of the energetic machinery and of specific immune effectors (e.g. toll-like receptor, tetraspanins) underlie the impact of a viral disease on a free-living vertebrate. Our study contributes to a more comprehensive understanding of the host-pathogen interaction in wild animals and the physiological pathways involved, and provides insights for effective wildlife disease monitoring and management strategies.
Collapse
Affiliation(s)
- Manrico Sebastiano
- Behavioural Ecology & Ecophysiology Group, Department of Biology, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium; Unité Physiologie Moléculaire et Adaptation, UMR7221-Muséum National d'Histoire Naturelle, CNRS, Paris, France.
| | - Olivier Chastel
- Centre d'Etudes Biologiques de Chizé (CEBC), UMR 7372 CNRS-Univ. La Rochelle, France
| | - Marcel Eens
- Behavioural Ecology & Ecophysiology Group, Department of Biology, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - David Costantini
- Unité Physiologie Moléculaire et Adaptation, UMR7221-Muséum National d'Histoire Naturelle, CNRS, Paris, France; Department of Ecological and Biological Sciences, University of Tuscia, Largo dell'Università snc, 01100 Viterbo, Italy
| |
Collapse
|
3
|
Egan MS, de Macedo R, Zackular JP. Metals in the gut: microbial strategies to overcome nutritional immunity in the intestinal tract. Metallomics 2024; 16:mfae052. [PMID: 39577845 DOI: 10.1093/mtomcs/mfae052] [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: 06/11/2024] [Accepted: 11/20/2024] [Indexed: 11/24/2024]
Abstract
Trace metals are indispensable nutritional factors for all living organisms. During host-pathogen interactions, they serve as crucial resources that dictate infection outcomes. Accordingly, the host uses a defense strategy known as nutritional immunity, which relies on coordinated metal chelation to mitigate bacterial advances. In response, pathogens employ complex strategies to secure these resources at sites of infection. In the gastrointestinal (GI) tract, the microbiota must also acquire metals for survival, making metals a central line of competition in this complex ecosystem. In this minireview, we outline how bacteria secure iron, zinc, and manganese from the host with a focus on the GI tract. We also reflect on how host dietary changes impact disease outcomes and discuss therapeutic opportunities to target bacterial metal uptake systems. Ultimately, we find that recent discoveries on the dynamics of transition metals at the host-pathogen-microbiota interface have reshaped our understanding of enteric infections and provided insights into virulence strategies, microbial cooperation, and antibacterial strategies.
Collapse
Affiliation(s)
- Marisa S Egan
- Division of Protective Immunity, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Biology, Swarthmore College, Swarthmore, PA 19081, USA
| | - Raquel de Macedo
- Division of Protective Immunity, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Microbiology, Faculdade de Ciências Médicas da Santa Casa de São Paulo, São Paulo, SP 01224-001, Brazil
| | - Joseph P Zackular
- Division of Protective Immunity, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- The Center for Microbial Medicine, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| |
Collapse
|
4
|
Ribeiro SA, Braga EL, Queiroga ML, Clementino MA, Fonseca XM, Belém MO, Magalhães LM, de Sousa JK, de Freitas TM, Veras HN, de Aquino CC, Santos AD, de Moura FR, Dos Santos AA, Havt A, Maciel BL, Lima AA. A New Murine Undernutrition Model Based on Complementary Feeding of Undernourished Children Causes Damage to the Morphofunctional Intestinal Epithelium Barrier. J Nutr 2024; 154:1232-1251. [PMID: 38346539 PMCID: PMC11347815 DOI: 10.1016/j.tjnut.2024.02.001] [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: 10/05/2023] [Revised: 01/26/2024] [Accepted: 02/01/2024] [Indexed: 02/28/2024] Open
Abstract
BACKGROUND Complementary feeding is critical in establishing undernutrition. However, experimental undernourished diets do not represent the amount of nutrients in the complementary diets of undernourished children. OBJECTIVES To develop, validate, and evaluate the impact of a new murine model of undernutrition on the intestinal epithelium, based on the complementary diet of undernourished children from 7 countries with low-socioeconomic power belonging to the Malnutrition-Enteric Diseases (MAL-ED) cohort study. METHODS We used the difference in the percentage of energy, macronutrients, fiber and zinc in the complementary diet of children without undernutrition compared with stunting (height-for-age Z-score < -2) for the MAL-ED diet formulation. Subsequently, C57BL/6 mice were fed a control diet (AIN-93M diet) or MAL-ED diet for 28 d. Weight was measured daily; body composition was measured every 7 d; lactulose:mannitol ratio (LM) and morphometry were evaluated on days 7 and 28; the cotransport test and analysis of intestinal transporters and tight junctions were performed on day 7. RESULTS The MAL-ED diet presented -8.03% energy, -37.46% protein, -24.20% lipid, -10.83% zinc, +5.93% carbohydrate, and +45.17% fiber compared with the control diet. This diet rapidly reduced weight gain and compromised body growth and energy reserves during the chronic period (P < 0.05). In the intestinal epithelial barrier, this diet caused an increase in the LM (P < 0.001) and reduced (P < 0.001) the villous area associated with an increase in FAT/CD36 in the acute period and increased (P < 0.001) mannitol excretion in the chronic period. CONCLUSIONS The MAL-ED diet induced undernutrition in mice, resulting in acute damage to the integrity of the intestinal epithelial barrier and a subsequent increase in the intestinal area during the chronic period. This study introduces the first murine model of undernutrition for the complementary feeding phase, based on data from undernourished children in 7 different countries.
Collapse
Affiliation(s)
- Samilly A Ribeiro
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, Brazil.
| | - Enock Lr Braga
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, Brazil
| | - Marcus L Queiroga
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, Brazil
| | - Marco A Clementino
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, Brazil
| | - Xhaulla Mqc Fonseca
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, Brazil
| | - Mônica O Belém
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, Brazil
| | - Lyvia Mvc Magalhães
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, Brazil
| | - José K de Sousa
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, Brazil
| | - Thiago M de Freitas
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, Brazil
| | - Herlice N Veras
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, Brazil
| | - Cristiane C de Aquino
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, Brazil
| | - Alan Dc Santos
- Núcleo de Estudos Químicos de Micromoléculas da Amazônia (NEQUIMA), Manaus, Brazil
| | - Flávio Rm de Moura
- Núcleo de Estudos Químicos de Micromoléculas da Amazônia (NEQUIMA), Manaus, Brazil
| | - Armênio A Dos Santos
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, Brazil
| | - Alexandre Havt
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, Brazil
| | - Bruna Ll Maciel
- Nutrition Postgraduation Program, Department of Nutrition, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Aldo Am Lima
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, Brazil
| |
Collapse
|
5
|
Jiang F, Yang Y, Mao Z, Cai W, Li G. ArcA positively regulates the expression of virulence genes and contributes to virulence of porcine Shiga toxin-producing enterotoxigenic Escherichia coli. Microbiol Spectr 2023; 11:e0152523. [PMID: 37916813 PMCID: PMC10714933 DOI: 10.1128/spectrum.01525-23] [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: 04/12/2023] [Accepted: 10/09/2023] [Indexed: 11/03/2023] Open
Abstract
IMPORTANCE Enterotoxigenic Escherichia coli (ETEC) cause severe diarrhea in humans and animals, leading to death and huge economic loss worldwide. Thus, elucidation of ETEC's pathogenic mechanisms will provide powerful data for the discovery of drugs serving as prevention or therapeutics against ETEC-caused diarrheal diseases. Here, we report that ArcA plays an essential role in the pathogenicity and virulence regulation in ETEC by positively regulating the expression of several key virulence factors including F18 fimbriae, heat-labile and heat-stable toxins, Shiga toxin 2e, and hemolysin, under microaerobic conditions and in vivo. Moreover, we found that positive regulation of several virulence genes by ArcA requires a global repressor H-NS (histone-like nucleoid structuring), implying that ArcA may exert positive effects by antagonizing H-NS. Collectively, our data established a key role for ArcA in the pathogenicity of porcine ETEC and ETEC strains isolated from human infections. Moreover, our work reveals another layer of regulation in relation to oxygen control of virulence factors in ETEC.
Collapse
Affiliation(s)
- Fengwei Jiang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
- Department of Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, Iowa, USA
| | - Yan Yang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Zhao Mao
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Wentong Cai
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Ganwu Li
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
- Department of Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, Iowa, USA
| |
Collapse
|
6
|
Perlman M, Senger S, Verma S, Carey J, Faherty CS. A foundational approach to culture and analyze malnourished organoids. Gut Microbes 2023; 15:2248713. [PMID: 37724815 PMCID: PMC10512930 DOI: 10.1080/19490976.2023.2248713] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 08/07/2023] [Indexed: 09/21/2023] Open
Abstract
The gastrointestinal (GI) epithelium plays a major role in nutrient absorption, barrier formation, and innate immunity. The development of organoid-based methodology has significantly impacted the study of the GI epithelium, particularly in the fields of mucosal biology, immunity, and host-microbe interactions. Various effects on the GI epithelium, such as genetics and nutrition, impact patients and alter disease states. Thus, incorporating these effects into organoid-based models will facilitate a better understanding of disease progression and offer opportunities to evaluate therapeutic candidates. One condition that has a significant effect on the GI epithelium is malnutrition, and studying the mechanistic impacts of malnutrition would enhance our understanding of several pathologies. Therefore, the goal of this study was to begin to develop methodology to generate viable malnourished organoids with accessible techniques and resources that can be used for a wide array of mechanistic studies. By selectively limiting distinct macronutrient components of organoid media, we were able to successfully culture and evaluate malnourished organoids. Genetic and protein-based analyses were used to validate the approach and confirm the presence of known biomarkers of malnutrition. Additionally, as proof-of-concept, we utilized malnourished organoid-derived monolayers to evaluate the effect of malnourishment on barrier formation and the ability of the bacterial pathogen Shigella flexneri to infect the GI epithelium. This work serves as the basis for new and exciting techniques to alter the nutritional state of organoids and investigate the related impacts on the GI epithelium.
Collapse
Affiliation(s)
- Meryl Perlman
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Stefania Senger
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, Boston, MA, USA
| | - Smriti Verma
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, USA
| | - James Carey
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, Boston, MA, USA
| | - Christina S. Faherty
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
7
|
Ramis G, Murciano F, Orengo J, González-Guijarro B, Cuellar-Flores A, Serrano D, Muñoz Luna A, Sánchez-Uribe P, Martínez-Alarcón L. Is Oral Vaccination against Escherichia coli Influenced by Zinc Oxide? Animals (Basel) 2023; 13:1754. [PMID: 37889667 PMCID: PMC10252008 DOI: 10.3390/ani13111754] [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: 03/31/2023] [Revised: 04/27/2023] [Accepted: 05/22/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND Although zinc oxide has been banned at therapeutic doses in the EU, its use is still legal in most countries with industrial pig farming. This compound has been shown to be very effective in preventing E. coli-related diseases. However, another strategy used to control this pathogen is vaccination, administered parenterally or orally. Oral vaccines contain live strains, with F4 and F18 binding factors. Since zinc oxide prevents E. coli adhesion, it is hypothesised that its presence at therapeutic doses (2500 ppm) may alter the immune response and the protection of intestinal integrity derived from the vaccination of animals. METHODS A group of piglets were orally vaccinated at weaning and divided into two subgroups; one group was fed a feed containing 2500 ppm zinc oxide (V + ZnO) for the first 15 days post-vaccination (dpv) and the other was not (V). Faeces were sampled from the animals at 6, 8, 11, 13, and 15 dpv. Unvaccinated animals without ZnO in their feed (Neg) were sampled simultaneously and, on day 15 post-vaccination, were also compared with a group of unvaccinated animals with ZnO in their feed (ZnO). RESULTS Differences were found in E. coli excretion, with less quantification in the V + ZnO group, and a significant increase in secretory IgA in the V group at 8 dpv, which later equalised with that of the V + ZnO group. There was also some difference in IFNα, IFNγ, IL1α, ILβ, and TNFα gene expression when comparing both vaccinated groups (p < 0.05). However, there was no difference in gene expression for the tight junction (TJ) proteins responsible for intestinal integrity. CONCLUSIONS Although some differences in the excretion of the vaccine strain were found when comparing both vaccinated groups, there are no remarkable differences in immune stimulation or soluble IgA production when comparing animals orally vaccinated against E. coli in combination with the presence or absence of ZnO in their feed. We can conclude that the immune response produced is very similar in both groups.
Collapse
Affiliation(s)
- Guillermo Ramis
- Departamento de Producción Animal, Facultad de Veterinaria, Universidad de Murcia, 30100 Murcia, Spain; (G.R.); (F.M.); (B.G.-G.)
- Instituto Murciano de Investigación en Biomédicina (IMIB), 30120 Murcia, Spain
| | - Francisco Murciano
- Departamento de Producción Animal, Facultad de Veterinaria, Universidad de Murcia, 30100 Murcia, Spain; (G.R.); (F.M.); (B.G.-G.)
| | - Juan Orengo
- Departamento de Producción Animal, Facultad de Veterinaria, Universidad de Murcia, 30100 Murcia, Spain; (G.R.); (F.M.); (B.G.-G.)
| | - Belén González-Guijarro
- Departamento de Producción Animal, Facultad de Veterinaria, Universidad de Murcia, 30100 Murcia, Spain; (G.R.); (F.M.); (B.G.-G.)
| | - Amanda Cuellar-Flores
- Departamento de Producción Animal, Facultad de Veterinaria, Universidad de Murcia, 30100 Murcia, Spain; (G.R.); (F.M.); (B.G.-G.)
| | - Daniel Serrano
- Departamento de Sanidad Animal, Facultad de Veterinaria, Universidad de Murcia, 30100 Murcia, Spain
| | - Antonio Muñoz Luna
- Departamento de Producción Animal, Facultad de Veterinaria, Universidad de Murcia, 30100 Murcia, Spain; (G.R.); (F.M.); (B.G.-G.)
- Instituto Murciano de Investigación en Biomédicina (IMIB), 30120 Murcia, Spain
| | | | - Laura Martínez-Alarcón
- Instituto Murciano de Investigación en Biomédicina (IMIB), 30120 Murcia, Spain
- Unidad para Docencia, Investigación y Calidad (UDICA), Hospital Clínico Universitario Virgen de la Arrixaca, 30120 Murcia, Spain
| |
Collapse
|
8
|
Párraga Solórzano PK, Bastille TS, Radin JN, Kehl-Fie TE. A Manganese-independent Aldolase Enables Staphylococcus aureus To Resist Host-imposed Metal Starvation. mBio 2023; 14:e0322322. [PMID: 36598285 PMCID: PMC9973326 DOI: 10.1128/mbio.03223-22] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 12/05/2022] [Indexed: 01/05/2023] Open
Abstract
The preferred carbon source of Staphylococcus aureus and many other pathogens is glucose, and its consumption is critical during infection. However, glucose utilization increases the cellular demand for manganese, a nutrient sequestered by the host as a defense against invading pathogens. Therefore, bacteria must balance glucose metabolism with the increasing demand that metal-dependent processes, such as glycolysis, impose upon the cell. A critical regulator that enables S. aureus to resist nutritional immunity is the ArlRS two-component system. This work revealed that ArlRS regulates the expression of FdaB, a metal-independent fructose 1,6-bisphosphate aldolase. Further investigation revealed that when S. aureus is metal-starved by the host, FdaB functionally replaces the metal-dependent isozyme FbaA, thereby allowing S. aureus to resist host-imposed metal starvation in culture. Although metal-dependent aldolases are canonically zinc-dependent, this work uncovered that FbaA requires manganese for activity and that FdaB protects S. aureus from manganese starvation. Both FbaA and FdaB contribute to the ability of S. aureus to cause invasive disease in wild-type mice. However, the virulence defect of a strain lacking FdaB was reversed in calprotectin-deficient mice, which have defects in manganese sequestration, indicating that this isozyme contributes to the ability of this pathogen to overcome manganese limitation during infection. Cumulatively, these observations suggest that the expression of the metal-independent aldolase FdaB allows S. aureus to alleviate the increased demand for manganese that glucose consumption imposes, and highlights the cofactor flexibility of even established metalloenzyme families. IMPORTANCE Staphylococcus aureus and other pathogens consume glucose during infection. Glucose utilization increases the demand for transition metals, such as manganese, a nutrient that the host limits as a defense mechanism against invading pathogens. Therefore, pathogenic bacteria must balance glucose and manganese requirements during infection. The two-component system ArlRS is an important regulator that allows S. aureus to adapt to both glucose and manganese starvation. Among the genes regulated by ArlRS is the metal-independent fructose 1,6-bisphosphate aldolase fdaB, which functionally substitutes for the metal-dependent isoenzyme FbaA and enables S. aureus to survive host-imposed manganese starvation. Unexpectedly, and differing from most characterized metal-dependent aldolases, FbaA requires manganese for activity. Cumulatively, these findings reveal a new mechanism for overcoming nutritional immunity as well as the cofactor plasticity of even well-characterized metalloenzyme families.
Collapse
Affiliation(s)
| | - Talina S. Bastille
- Department of Microbiology, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
| | - Jana N. Radin
- Department of Microbiology, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
| | - Thomas E. Kehl-Fie
- Department of Microbiology, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
| |
Collapse
|
9
|
Arredondo-Hernandez R, Siebe C, Castillo-Rojas G, Ponce de León S, López-Vidal Y. The synergistic interaction of systemic inflammation, dysbiosis and antimicrobial resistance promotes growth restriction in children with acute severe malnutrition: An emphasis on Escherichia coli. FRONTIERS IN ANTIBIOTICS 2022; 1:1001717. [PMID: 39816412 PMCID: PMC11732057 DOI: 10.3389/frabi.2022.1001717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 10/05/2022] [Indexed: 01/18/2025]
Abstract
A healthy development is denied to millions of children worldwide as harsh life conditions manifest themselves in an altered inflammation-prone microbiome crosstalk environment. Keynote of this tragedy is that insufficient nutritious amino acid blocks lipids-intake to sustain diverse microbiota, and promotes the generalist strategy followed by Escherichia coli -besides other proteobacteria- of shifting gut metabolism, subverting the site specificity of first immune reaction. Furthermore, it could be hypothesized that selective success lies in their ability to induce inflammation, since this phenomenon also fuels horizontal gene transfer (HGT). In this review, we dilucidate how immune mechanisms of environmental enteric dysfunction affect overgrowth restriction, infectious morbidity rate, and acquired lifelong risks among severe acute malnourished children. Also, despite acknowledging complexities of antimicrobial resistant enrichment, we explore and speculate over the links between virulence regulation and HGT as an indissociable part in the quest for new inflammatory niches by open genome bacteria, particularly when both collide in the most vulnerable.
Collapse
Affiliation(s)
- Rene Arredondo-Hernandez
- Laboratorio de Microbioma, División de Investigación y División de Posgrado, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Christina Siebe
- Instituto de Geología, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Gonzalo Castillo-Rojas
- Programa de Inmunología Molecular Microbiana, Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Samuel Ponce de León
- Laboratorio de Microbioma, División de Investigación y División de Posgrado, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Yolanda López-Vidal
- Programa de Inmunología Molecular Microbiana, Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
10
|
Marginal Zinc Deficiency Aggravated Intestinal Barrier Dysfunction and Inflammation through ETEC Virulence Factors in a Mouse Model of Diarrhea. Vet Sci 2022; 9:vetsci9090507. [PMID: 36136723 PMCID: PMC9503546 DOI: 10.3390/vetsci9090507] [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: 08/02/2022] [Revised: 09/13/2022] [Accepted: 09/13/2022] [Indexed: 11/21/2022] Open
Abstract
Simple Summary Enterotoxigenic Escherichia coli (ETEC) is one of the most common bacterial causes of diarrhea in children and farm animals. Zinc has received widespread attention for its roles in the prevention and treatment of diarrhea. However, zinc is also essential for the pathogenesis of ETEC. This study aimed to explore the accurate effect and mechanisms of marginal zinc deficiency on ETEC k88 infection and host intestinal health. Using the newly developed marginal zinc deficiency and ETEC k88 infection mouse model, we found that marginal zinc deficiency aggravated growth impairment, diarrhea, intestinal morphology, intestinal permeability, and inflammation induced by ETEC k88 infection. Consistently, intestinal ETEC k88 shedding was also higher in mice with marginal zinc deficiency. However, marginal zinc deficiency failed to affect host zinc levels and correspondingly the zinc-receptor GPR39 expression in the jejunum. In addition, marginal zinc deficiency upregulated the relative expression of virulence genes involved in heat-labile and heat-stable enterotoxins, motility, cellular adhesion, and biofilm formation in the cecum content of mice with ETEC infection. These findings provide a new explanation for zinc treatment of ETEC infection. Abstract Zinc is both essential and inhibitory for the pathogenesis of enterotoxigenic Escherichia coli (ETEC). However, the accurate effects and underlying mechanism of marginal zinc deficiency on ETEC infection are not fully understood. Here, a marginal zinc-deficient mouse model was established by feeding mice with a marginal zinc-deficient diet, and ETEC k88 was further administrated to mice after antibiotic disruption of the normal microbiota. Marginal zinc deficiency aggravated growth impairment, diarrhea, intestinal morphology, intestinal permeability, and inflammation induced by ETEC k88 infection. In line with the above observations, marginal zinc deficiency also increased the intestinal ETEC shedding, though the concentration of ETEC in the intestinal content was not different or even decreased in the stool. Moreover, marginal zinc deficiency failed to change the host’s zinc levels, as evidenced by the fact that the serum zinc levels and zinc-receptor GPR39 expression in jejunum were not significantly different in mice with ETEC challenge. Finally, marginal zinc deficiency upregulated the relative expression of virulence genes involved in heat-labile and heat-stable enterotoxins, motility, cellular adhesion, and biofilm formation in the cecum content of mice with ETEC infection. These findings demonstrated that marginal zinc deficiency likely regulates ETEC infection through the virulence factors, whereas it is not correlated with host zinc levels.
Collapse
|
11
|
Samuelson DR, Haq S, Knoell DL. Divalent Metal Uptake and the Role of ZIP8 in Host Defense Against Pathogens. Front Cell Dev Biol 2022; 10:924820. [PMID: 35832795 PMCID: PMC9273032 DOI: 10.3389/fcell.2022.924820] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 05/26/2022] [Indexed: 01/13/2023] Open
Abstract
Manganese (Mn) and Zinc (Zn) are essential micronutrients whose concentration and location within cells are tightly regulated at the onset of infection. Two families of Zn transporters (ZIPs and ZnTs) are largely responsible for regulation of cytosolic Zn levels and to a certain extent, Mn levels, although much less is known regarding Mn. The capacity of pathogens to persevere also depends on access to micronutrients, yet a fundamental gap in knowledge remains regarding the importance of metal exchange at the host interface, often referred to as nutritional immunity. ZIP8, one of 14 ZIPs, is a pivotal importer of both Zn and Mn, yet much remains to be known. Dietary Zn deficiency is common and commonly occurring polymorphic variants of ZIP8 that decrease cellular metal uptake (Zn and Mn), are associated with increased susceptibility to infection. Strikingly, ZIP8 is the only Zn transporter that is highly induced following bacterial exposure in key immune cells involved with host defense against leading pathogens. We postulate that mobilization of Zn and Mn into key cells orchestrates the innate immune response through regulation of fundamental defense mechanisms that include phagocytosis, signal transduction, and production of soluble host defense factors including cytokines and chemokines. New evidence also suggests that host metal uptake may have long-term consequences by influencing the adaptive immune response. Given that activation of ZIP8 expression by pathogens has been shown to influence parenchymal, myeloid, and lymphoid cells, the impact applies to all mucosal surfaces and tissue compartments that are vulnerable to infection. We also predict that perturbations in metal homeostasis, either genetic- or dietary-induced, has the potential to impact bacterial communities in the host thereby adversely impacting microbiome composition. This review will focus on Zn and Mn transport via ZIP8, and how this vital metal transporter serves as a "go to" conductor of metal uptake that bolsters host defense against pathogens. We will also leverage past studies to underscore areas for future research to better understand the Zn-, Mn- and ZIP8-dependent host response to infection to foster new micronutrient-based intervention strategies to improve our ability to prevent or treat commonly occurring infectious disease.
Collapse
Affiliation(s)
- Derrick R. Samuelson
- Division of Pulmonary, Critical Care, and Sleep, Department of Internal Medicine, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Sabah Haq
- Department of Pharmacy Practice and Science, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, United States
| | - Daren L. Knoell
- Department of Pharmacy Practice and Science, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, United States,*Correspondence: Daren L. Knoell,
| |
Collapse
|
12
|
Strain R, Stanton C, Ross RP. Effect of diet on pathogen performance in the microbiome. MICROBIOME RESEARCH REPORTS 2022; 1:13. [PMID: 38045644 PMCID: PMC10688830 DOI: 10.20517/mrr.2021.10] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 02/17/2022] [Accepted: 02/23/2022] [Indexed: 12/05/2023]
Abstract
Intricate interactions among commensal bacteria, dietary substrates and immune responses are central to defining microbiome community composition, which plays a key role in preventing enteric pathogen infection, a dynamic phenomenon referred to as colonisation resistance. However, the impact of diet on sculpting microbiota membership, and ultimately colonisation resistance has been overlooked. Furthermore, pathogens have evolved strategies to evade colonisation resistance and outcompete commensal microbiota by using unique nutrient utilisation pathways, by exploiting microbial metabolites as nutrient sources or by environmental cues to induce virulence gene expression. In this review, we will discuss the interplay between diet, microbiota and their associated metabolites, and how these can contribute to or preclude pathogen survival.
Collapse
Affiliation(s)
- Ronan Strain
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12 YT20, Ireland
- Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork P61 C996, Ireland
| | - Catherine Stanton
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12 YT20, Ireland
- Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork P61 C996, Ireland
| | - R. Paul Ross
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12 YT20, Ireland
- School of Microbiology, University College Cork, College Road, Cork T12 K8AF, Ireland
| |
Collapse
|
13
|
Pei L, Liu J, Huang Z, Iqbal M, Shen Y. Effects of Lactic Acid Bacteria Isolated from Equine on Salmonella-Infected Gut Mouse Model. Probiotics Antimicrob Proteins 2021; 15:469-478. [PMID: 34651283 DOI: 10.1007/s12602-021-09841-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/26/2021] [Indexed: 11/28/2022]
Abstract
The aim of this study was to evaluate the antibacterial potential of lactic acid bacteria (Weissella confuse, Pediococcus acidilactici, and Ligilactobacillus equi) isolated from healthy equine in Wuhan against Salmonella Typhimurium CVCC542-induced mice model on intestinal microflora. In previous studies, these isolated strains showed good probiotic potentials in vitro. In this study, fifty healthy mice were randomly divided into five groups, the blank control group, the control group, the Pediococcus acidilactici group (1 × 108 CFU/day), the Ligilactobacillus equi group (1 × 108 CFU/day), and the Weissella confuse group (1 × 108 CFU/day). The body weight in control group and Weissella confuse group showed significant decreased (P < 0.05, P < 0.01), while Pediococcus acidilactici group and Ligilactobacillus equi group showed good recovering after treatments. The lowest diarrhea rate was shown in Ligilactobacillus equi group after treatment. In histopathology, Ligilactobacillus equi group showed the least structural damage in duodenum, and all probiotic treatment groups showed less damage in cecum. The sequence data and optical transform unit showed that Pediococcus acidilactici group and Ligilactobacillus equi group had higher number than control group, while the diversity data showed that the control group and Weissella confuse group had lower diversity in cecum. Microbial community analysis showed increased abundance of Firmicutes, Bacteroidetes, uncultured_bacterium_f_Muribaculaceae, and Lactobacillus in treatment groups, while potential microbes that can induce intestinal diseases such as Verrucomicrobia, Akkermansia, and Lachnospiraceae_NK4A136_group decreased in the treatment groups. In conclusion, lactic acid bacteria isolated from the healthy horses could alleviate the infection of Salmonella and regulate intestinal flora.
Collapse
Affiliation(s)
- Lulu Pei
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Juanjuan Liu
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Zonghao Huang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Mudassar Iqbal
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Yaoqin Shen
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China.
| |
Collapse
|
14
|
Asempa TE, Abdelraouf K, Nicolau DP. Metallo-β-lactamase resistance in Enterobacteriaceae is an artefact of currently utilized antimicrobial susceptibility testing methods. J Antimicrob Chemother 2021; 75:997-1005. [PMID: 31930305 DOI: 10.1093/jac/dkz532] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 11/26/2019] [Accepted: 11/30/2019] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND MBLs are a major contributor to β-lactam resistance when tested using CAMHB. Despite in vitro resistance, positive outcomes have been reported in MBL-infected patients following carbapenem treatment. The impact of physiological zinc concentrations on this in vitro-in vivo MBL discordance warrants investigation. OBJECTIVES To evaluate meropenem in vitro activity against MBL-producing Enterobacteriaceae in zinc-depleted broth (Chelex-CAMHB, EDTA-CAMHB) and assess meropenem efficacy in murine infection models. METHODS Neutropenic mice received a meropenem human-simulated regimen of 2 g q8h or levofloxacin 750 mg q24h (for model validation). Zinc concentrations were determined in conventional CAMHB, zinc-depleted CAMHB and epithelial lining fluid (ELF) of lung-infected mice. RESULTS All MBL-producing isolates (NDM, n = 25; VIM, n = 3; IMP, n = 2) examined were meropenem resistant in CAMHB and susceptible in zinc-depleted CAMHB (5- to 11-fold reduction), with zinc depletion having no impact on levofloxacin MICs. Zinc concentrations (mean ± SD) in CAMHB were 0.959 ± 0.038 mg/L and in both zinc-depleted CAMHB and ELF were <0.002 mg/L. In vivo, levofloxacin displayed predictable efficacy consistent with its phenotypic profile, while meropenem produced >1 log unit bacterial killing despite in vitro resistance in conventional CAMHB. CONCLUSIONS Results indicate that meropenem in vivo efficacy is best represented by the pharmacodynamic profile generated using MICs determined in zinc-depleted media for MBL-producing Enterobacteriaceae. These translational data suggest that the use of conventional CAMHB for MBL susceptibility testing is inappropriate in distinguishing meaningful in vivo resistance given that zinc concentrations are supraphysiological in conventional CAMHB and negligible at infection sites.
Collapse
Affiliation(s)
- Tomefa E Asempa
- Center for Anti-Infective Research and Development, Hartford Hospital, 80 Seymour Street, Hartford, CT, USA
| | - Kamilia Abdelraouf
- Center for Anti-Infective Research and Development, Hartford Hospital, 80 Seymour Street, Hartford, CT, USA
| | - David P Nicolau
- Center for Anti-Infective Research and Development, Hartford Hospital, 80 Seymour Street, Hartford, CT, USA.,Division of Infectious Diseases, Hartford Hospital, 80 Seymour Street, Hartford, CT, USA
| |
Collapse
|
15
|
Guerrant RL, Bolick DT, Swann JR. Modeling Enteropathy or Diarrhea with the Top Bacterial and Protozoal Pathogens: Differential Determinants of Outcomes. ACS Infect Dis 2021; 7:1020-1031. [PMID: 33901398 PMCID: PMC8154416 DOI: 10.1021/acsinfecdis.0c00831] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
![]()
Developing effective
therapeutics or preventive interventions for
important health threats is greatly enhanced whenever accessible models
can enable the assessment of clinically important outcomes. While
no non-human model is ever perfect, inexpensive in vivo small animal models in such as mice are often of great help in assessing
the relevant efficacy of potential interventions. In addition to acute
diarrhea, the long-term growth and developmental effects of enteric
infections, with or without overt diarrhea, are increasingly recognized.
To address these diverse effects, inexpensive animal models are proving
to be very helpful. Herein, we review the major clinical concerns
with enteric parasitic and bacterial infections that are extremely
common worldwide, especially in vulnerable young children living in
impoverished areas, and the recently published murine models of these
infections and their outcomes. We find that common dietary deficiencies
seen in children in developing areas have striking effects on diarrhea
and enteropathy outcomes in mice. However, these effects differ with
different pathogens. Specifically, the effects of protein or zinc
deficiency differ considerably with different major protozoal and
bacterial pathogens, suggesting different pathogenetic pathways and
intervention effects. The pathogens reviewed are the seven top parasitic
and bacterial pathogens seen in children, namely, Cryptosporidium, Giardia, Campylobacter, Shigella, enterotoxigenic Escherichia coli (ETEC), enteroaggregative E. coli (EAEC), and enteropathogenic E. coli (EPEC).
Collapse
Affiliation(s)
- Richard L. Guerrant
- Center for Global Health Equity, Division of Infectious Diseases and International Health, University of Virginia School of Medicine, Charlottesville, Virginia 22908, United States
| | - David T. Bolick
- Center for Global Health Equity, Division of Infectious Diseases and International Health, University of Virginia School of Medicine, Charlottesville, Virginia 22908, United States
| | - Jonathan R. Swann
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, United Kingdom
- Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, Imperial College London, London SW7 2AZ, United Kingdom
| |
Collapse
|
16
|
Yang YS, Shang Q, Zhang YP, Niu WY, Xue JJ. Synthesis and self-assembly of Salen type Schiff based on o-phenylenediamine organogels in response to Zn2+. INORG CHEM COMMUN 2021. [DOI: 10.1016/j.inoche.2020.108402] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
17
|
Cao Y, Liu J, Zhu W, Qin N, Ren X, Zhu B, Xia X. Impact of dietary components on enteric infectious disease. Crit Rev Food Sci Nutr 2021; 62:4010-4035. [PMID: 33455435 DOI: 10.1080/10408398.2021.1871587] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Diets impact host health in multiple ways and an unbalanced diet could contribute to the initiation or progression of a variety of diseases. Although a wealth of information exists on the connections between diet and chronic metabolic diseases such as cardiovascular disease, diabetes mellitus, etc., how diet influences enteric infectious disease still remain underexplored. The review summarizes the current findings on the link between various dietary components and diverse enteric infectious diseases. Dietary ingredients discussed include macronutrients (carbohydrates, lipids, proteins), micronutrients (vitamins, minerals), and other dietary ingredients (phytonutrients and probiotic supplements). We first describe the importance of enteric infectious diseases and the direct and indirect relationship between diet and enteric infectious diseases. Then we discuss the effects of different dietary components on the susceptibility to or progression of enteric infectious disease. Finally, we delineate current knowledge gap and highlighted future research directions. The literature review revealed that different dietary components affect host resistance to enteric infections through a variety of mechanisms. Dietary components may directly inhibit or bind to enteric pathogens, or indirectly influence enteric infections through modulating immune function and gut microbiota. Elucidating the unique repercussions of different diets on enteric infections in this review may help provide dietary guidelines or design dietary interventions to prevent or alleviate enteric infectious diseases.
Collapse
Affiliation(s)
- Yu Cao
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China.,National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning, China
| | - Jiaxiu Liu
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning, China
| | - Wenxiu Zhu
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning, China
| | - Ningbo Qin
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning, China
| | - Xiaomeng Ren
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning, China
| | - Beiwei Zhu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China.,National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning, China
| | - Xiaodong Xia
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning, China
| |
Collapse
|
18
|
Su SY, Tang QQ. Altered intestinal microflora and barrier injury in severe acute pancreatitis can be changed by zinc. Int J Med Sci 2021; 18:3050-3058. [PMID: 34400875 PMCID: PMC8364456 DOI: 10.7150/ijms.45980] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 06/04/2021] [Indexed: 01/01/2023] Open
Abstract
To investigate the effect of zinc (Zn) supplementation on intestinal microflora changes and bacterial translocation in rats with severe acute pancreatitis (SAP), the rats were divided into the sham surgery (SS), SAP, SS + Zn, and SAP + Zn groups. Saline (0.1 mL/100g) and 5% sodium taurocholate were injected into the pancreaticobiliary duct of the rats in the SS and SAP + Zn groups, respectively. Intraperitoneal injection of 5 mg/kg Zn was performed immediately after injecting saline or 5% sodium taurocholate into the rats in both groups. Serum amylase and Zn levels, plasma endogenous endotoxin, intestinal permeability, and the positive rate of intestinal bacterial translocation were detected, haematoxylin and eosin (H&E) staining was performed, and the pancreatic tissue scores were calculated for each group. In addition, immunohistochemical (IHC) staining was performed to evaluate the expression of IL-1β and TNF-α. Real-time fluorescence quantitative PCR was used to quantify the gene copy numbers of Escherichia, Bifidobacterium, and Lactobacillus in the cecum. The levels of amylase and plasma endotoxin in the SAP group were significantly higher than those in the SS and SS + Zn groups. Intestinal mucosal permeability and intestinal bacterial translocation in the liver, pancreas, and mesenteric lymph nodes were increased in the SAP group. However, the levels of amylase and plasma endotoxin were decreased as a result of zinc supplementation in the SAP group. The expression of IL-1β and TNF-α was also reduced to a greater degree in the SAP + Zn group than in the SAP group. Moreover, alleviated intestinal mucosal permeability and intestinal bacterial translocation in the liver, pancreas, and mesenteric lymph nodes were found in the SAP + Zn group. The results of real-time quantitative PCR showed that the gene copy number of Escherichia increased with time, and the gene copy numbers of Lactobacillus and Bifidobacterium decreased over time. Zn supplementation prevented the release of TNF-α and IL-1β, alleviated intestinal permeability and endotoxemia, reduced bacterial translocation, and inhibited changes in pathogenic intestinal flora in rats with SAP.
Collapse
Affiliation(s)
- Shi-Yue Su
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Hefei 230022, Anhui, China
| | - Qin-Qing Tang
- Department of General Surgery, the First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Hefei 230022, Anhui, China
| |
Collapse
|
19
|
Teh AYH, Cavacini L, Hu Y, Kumru OS, Xiong J, Bolick DT, Joshi SB, Grünwald-Gruber C, Altmann F, Klempner M, Guerrant RL, Volkin DB, Wang Y, Ma JKC. Investigation of a monoclonal antibody against enterotoxigenic Escherichia coli, expressed as secretory IgA1 and IgA2 in plants. Gut Microbes 2021; 13:1-14. [PMID: 33439092 PMCID: PMC7833773 DOI: 10.1080/19490976.2020.1859813] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 11/09/2020] [Accepted: 11/23/2020] [Indexed: 02/04/2023] Open
Abstract
Passive immunization with antibodies is a promising approach against enterotoxigenic Escherichia coli diarrhea, a prevalent disease in LMICs. The objective of this study was to investigate expression of a monoclonal anti-ETEC CfaE secretory IgA antibody in N. benthamiana plants, with a view to facilitating access to ETEC passive immunotherapy. SIgA1 and SIgA2 forms of mAb 68-81 were produced by co-expressing the light and engineered heavy chains with J chain and secretory component in N. benthamiana. Antibody expression and assembly were compared with CHO-derived antibodies by SDS-PAGE, western blotting, size-exclusion chromatography and LC-MS peptide mapping. N-linked glycosylation was assessed by rapid fluorescence/mass spectrometry and LC-ESI-MS. Susceptibility to gastric digestion was assessed in an in vitro model. Antibody function was compared for antigen binding, a Caco-2 cell-based ETEC adhesion assay, an ETEC hemagglutination inhibition assay and a murine in vivo challenge study. SIgA1 assembly appeared superior to SIgA2 in plants. Both sub-classes exhibited resistance to degradation by simulated gastric fluid, comparable to CHO-produced 68-61 SIgA1. The plant expressed SIgAs had more homogeneous N-glycosylation than CHO-derived SIgAs, but no alteration of in vitro functional activity was observed, including antibodies expressed in a plant line engineered for mammalian-like N glycosylation. The plant-derived SIgA2 mAb demonstrated protection against diarrhea in a murine infection model. Although antibody yield and purification need to be optimized, anti-ETEC SIgA antibodies produced in a low-cost plant platform are functionally equivalent to CHO antibodies, and provide promise for passive immunotherapy in LMICs.
Collapse
MESH Headings
- Animals
- Antibodies, Bacterial/genetics
- Antibodies, Bacterial/immunology
- Antibodies, Bacterial/metabolism
- Antibodies, Bacterial/therapeutic use
- Antibodies, Monoclonal/genetics
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/metabolism
- Antibodies, Monoclonal/therapeutic use
- Antibody Affinity
- Bacterial Adhesion/drug effects
- Caco-2 Cells
- Enterotoxigenic Escherichia coli/immunology
- Escherichia coli Infections/microbiology
- Escherichia coli Infections/therapy
- Gastric Acid/metabolism
- Glycosylation
- Humans
- Immunoglobulin A, Secretory/genetics
- Immunoglobulin A, Secretory/immunology
- Immunoglobulin A, Secretory/metabolism
- Immunoglobulin A, Secretory/therapeutic use
- Immunotherapy
- Mice
- Plants, Genetically Modified
- Nicotiana/genetics
- Nicotiana/metabolism
Collapse
Affiliation(s)
- Audrey Y-H Teh
- Molecular Immunology Unit, Institute for Infection and Immunity, St. George’s University of London, London, UK
| | - Lisa Cavacini
- MassBiologics of the University of Massachusetts Medical School, Boston, MA, USA
| | - Yue Hu
- Vaccine Analytics and Formulation Center, Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, USA
| | - Ozan S. Kumru
- Vaccine Analytics and Formulation Center, Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, USA
| | - Jian Xiong
- Vaccine Analytics and Formulation Center, Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, USA
| | - David T. Bolick
- Division of Infectious Disease and International Health, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Sangeeta B. Joshi
- Vaccine Analytics and Formulation Center, Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, USA
| | - Clemens Grünwald-Gruber
- Department for Chemistry, Division of Biochemistry, Universität Für Bodenkultur Wien, Vienna, Austria
| | - Friedrich Altmann
- Department for Chemistry, Division of Biochemistry, Universität Für Bodenkultur Wien, Vienna, Austria
| | - Mark Klempner
- MassBiologics of the University of Massachusetts Medical School, Boston, MA, USA
| | - Richard L. Guerrant
- Division of Infectious Disease and International Health, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - David B. Volkin
- Vaccine Analytics and Formulation Center, Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, USA
| | - Yang Wang
- MassBiologics of the University of Massachusetts Medical School, Boston, MA, USA
| | - Julian K-C. Ma
- Molecular Immunology Unit, Institute for Infection and Immunity, St. George’s University of London, London, UK
| |
Collapse
|
20
|
von Pein JB, Stocks CJ, Schembri MA, Kapetanovic R, Sweet MJ. An alloy of zinc and innate immunity: Galvanising host defence against infection. Cell Microbiol 2020; 23:e13268. [PMID: 32975847 DOI: 10.1111/cmi.13268] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/31/2020] [Accepted: 09/03/2020] [Indexed: 12/16/2022]
Abstract
Innate immune cells such as macrophages and neutrophils initiate protective inflammatory responses and engage antimicrobial responses to provide frontline defence against invading pathogens. These cells can both restrict the availability of certain transition metals that are essential for microbial growth and direct toxic concentrations of metals towards pathogens as antimicrobial responses. Zinc is important for the structure and function of many proteins, however excess zinc can be cytotoxic. In recent years, several studies have revealed that innate immune cells can deliver toxic concentrations of zinc to intracellular pathogens. In this review, we discuss the importance of zinc status during infectious disease and the evidence for zinc intoxication as an innate immune antimicrobial response. Evidence for pathogen subversion of this response is also examined. The likely mechanisms, including the involvement of specific zinc transporters that facilitate delivery of zinc by innate immune cells for metal ion poisoning of pathogens are also considered. Precise mechanisms by which excess levels of zinc can be toxic to microorganisms are then discussed, particularly in the context of synergy with other antimicrobial responses. Finally, we highlight key unanswered questions in this emerging field, which may offer new opportunities for exploiting innate immune responses for anti-infective development.
Collapse
Affiliation(s)
- Jessica B von Pein
- Institute for Molecular Bioscience (IMB), The University of Queensland, St. Lucia, Queensland, Australia.,IMB Centre for Inflammation and Disease Research, The University of Queensland, St. Lucia, Queensland, Australia.,Australian Infectious Diseases Research Centre, The University of Queensland, St. Lucia, Queensland, Australia
| | - Claudia J Stocks
- Institute for Molecular Bioscience (IMB), The University of Queensland, St. Lucia, Queensland, Australia.,IMB Centre for Inflammation and Disease Research, The University of Queensland, St. Lucia, Queensland, Australia.,Australian Infectious Diseases Research Centre, The University of Queensland, St. Lucia, Queensland, Australia
| | - Mark A Schembri
- Australian Infectious Diseases Research Centre, The University of Queensland, St. Lucia, Queensland, Australia.,School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, Queensland, Australia
| | - Ronan Kapetanovic
- Institute for Molecular Bioscience (IMB), The University of Queensland, St. Lucia, Queensland, Australia.,IMB Centre for Inflammation and Disease Research, The University of Queensland, St. Lucia, Queensland, Australia.,Australian Infectious Diseases Research Centre, The University of Queensland, St. Lucia, Queensland, Australia
| | - Matthew J Sweet
- Institute for Molecular Bioscience (IMB), The University of Queensland, St. Lucia, Queensland, Australia.,IMB Centre for Inflammation and Disease Research, The University of Queensland, St. Lucia, Queensland, Australia.,Australian Infectious Diseases Research Centre, The University of Queensland, St. Lucia, Queensland, Australia
| |
Collapse
|
21
|
Zhou JY, Lin HL, Wang Z, Zhang SW, Huang DG, Gao CQ, Yan HC, Wang XQ. Zinc L-Aspartate enhances intestinal stem cell activity to protect the integrity of the intestinal mucosa against deoxynivalenol through activation of the Wnt/β-catenin signaling pathway. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2020; 262:114290. [PMID: 32155551 DOI: 10.1016/j.envpol.2020.114290] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Revised: 02/01/2020] [Accepted: 02/27/2020] [Indexed: 06/10/2023]
Abstract
The micronutrient, zinc, plays a vital role in modulating cellular signaling recognition and enhancing intestinal barrier function. However, the precise mechanisms underlying the zinc regulation of intestinal stem cell (ISC) renewal and regeneration ability, which drive intestinal epithelial turnover to maintain the intestinal barrier, under physiological and pathological conditions are unknown. In this study, we used in vivo mouse plus ex vivo enteroid model to investigate thoroughly the protection efficacy of zinc L-aspartate (Zn-Asp) on intestinal mucosal integrity exposed to deoxynivalenol (DON). The results showed that 10 rather than 20 mg/kg body weight (BW) Zn-Asp (calculation in zinc) significantly increased the jejunum mass and ameliorated mucosa injury caused by 2 mg/kg BW DON treatment, including improvement of the intestinal morphology and barrier, as well as enteroid-forming and -budding efficiency, which was expanded from crypt cells isolated from jejunum of mice in each group. The repair process stimulated by Zn-Asp was also accompanied by increased fluorescence signal intensity of KRT20 and Villin; increased numbers of MUC2+, CAG+, LYZ+, BrdU+ and Ki67+ cells in mouse jejunum; and protein expression of Ki67 and PCNA in the jejunum, crypt and enteroid. Simultaneously, Zn-Asp increased ISC activity to promote intestinal epithelial renewal even under physiological conditions. These results were further verified in ex vivo enteroid culture experiments, which were treated with 100 μmol/L Zn-Asp (calculation in zinc) and 100 ng/mL DON for 72 h. Furthermore, we demonstrated that Zn-Asp improved intestinal integrity or accelerated wound healing along with Wnt/β-catenin signaling upregulation or reactivation. Our findings indicate Zn-Asp, especially Zn, enhances ISC activity to maintain the intestinal integrity by activating the Wnt/β-catenin signaling, which sheds some light upon effective preventive strategies for intestinal injury induced by mycotoxin based on ISCs with exogenous zinc preparations in the proper drugs, health foods or qualified feed.
Collapse
Affiliation(s)
- Jia-Yi Zhou
- College of Animal Science, South China Agricultural University, Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangzhou, Guangdong, China
| | - Hua-Lin Lin
- College of Animal Science, South China Agricultural University, Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangzhou, Guangdong, China
| | - Zhe Wang
- College of Letters & Science, University of Wisconsin-Madison, Madison, WI, United States
| | - Sai-Wu Zhang
- College of Animal Science, South China Agricultural University, Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangzhou, Guangdong, China
| | - Deng-Gui Huang
- College of Animal Science, South China Agricultural University, Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangzhou, Guangdong, China
| | - Chun-Qi Gao
- College of Animal Science, South China Agricultural University, Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangzhou, Guangdong, China; Integrative Microbiology Research Centre, South China Agricultural University, Guangzhou, China
| | - Hui-Chao Yan
- College of Animal Science, South China Agricultural University, Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangzhou, Guangdong, China
| | - Xiu-Qi Wang
- College of Animal Science, South China Agricultural University, Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangzhou, Guangdong, China.
| |
Collapse
|
22
|
Weiskirchen S, Weiper K, Tolba RH, Weiskirchen R. All You Can Feed: Some Comments on Production of Mouse Diets Used in Biomedical Research with Special Emphasis on Non-Alcoholic Fatty Liver Disease Research. Nutrients 2020; 12:nu12010163. [PMID: 31936026 PMCID: PMC7019265 DOI: 10.3390/nu12010163] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 12/25/2019] [Accepted: 12/31/2019] [Indexed: 02/07/2023] Open
Abstract
The laboratory mouse is the most common used mammalian research model in biomedical research. Usually these animals are maintained in germ-free, gnotobiotic, or specific-pathogen-free facilities. In these facilities, skilled staff takes care of the animals and scientists usually don’t pay much attention about the formulation and quality of diets the animals receive during normal breeding and keeping. However, mice have specific nutritional requirements that must be met to guarantee their potential to grow, reproduce and to respond to pathogens or diverse environmental stress situations evoked by handling and experimental interventions. Nowadays, mouse diets for research purposes are commercially manufactured in an industrial process, in which the safety of food products is addressed through the analysis and control of all biological and chemical materials used for the different diet formulations. Similar to human food, mouse diets must be prepared under good sanitary conditions and truthfully labeled to provide information of all ingredients. This is mandatory to guarantee reproducibility of animal studies. In this review, we summarize some information on mice research diets and general aspects of mouse nutrition including nutrient requirements of mice, leading manufacturers of diets, origin of nutrient compounds, and processing of feedstuffs for mice including dietary coloring, autoclaving and irradiation. Furthermore, we provide some critical views on the potential pitfalls that might result from faulty comparisons of grain-based diets with purified diets in the research data production resulting from confounding nutritional factors.
Collapse
Affiliation(s)
- Sabine Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, D-52074 Aachen, Germany; (S.W.); (K.W.)
| | - Katharina Weiper
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, D-52074 Aachen, Germany; (S.W.); (K.W.)
- Institute of Laboratory Animal Science and Experimental Surgery, RWTH University Hospital Aachen, D-52074 Aachen, Germany;
| | - René H. Tolba
- Institute of Laboratory Animal Science and Experimental Surgery, RWTH University Hospital Aachen, D-52074 Aachen, Germany;
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, D-52074 Aachen, Germany; (S.W.); (K.W.)
- Correspondence: ; Tel.: +49-(0)241-80-88683
| |
Collapse
|
23
|
Wang Y, Li A, Zhang L, Waqas M, Mehmood K, Iqbal M, Muyou C, Li Z, Lian Y, Sizhu S, Li J. Probiotic potential of Lactobacillus on the intestinal microflora against Escherichia coli induced mice model through high-throughput sequencing. Microb Pathog 2019; 137:103760. [PMID: 31562897 DOI: 10.1016/j.micpath.2019.103760] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 09/09/2019] [Accepted: 09/24/2019] [Indexed: 12/29/2022]
Abstract
The aim of this study was to evaluate the antibacterial potential of Lactobacillus screened from Tibetan yaks on clinical symptoms and intestinal microflora in enteroinvasive Escherichia coli (EIEC) induced mice model. In vitro study, Lactobacillus reuteri (LR1) exhibited stronger resistance to acid and bile and inhibited the growth of EIEC than Lactobacillus mucosae (LM1). The mice were randomly divided into four groups i.e. the LR1 group (LR1 1 × 109 CFU/day), LM1 group (LM1 1 × 109 CFU/day), blank control group and control group. Mice in control, LR1, and LM1 groups were challenged with EIEC on day 23. The body weight in the control and LM1 groups were significantly decreased after the infection with EIEC (P < 0.05), whereas the body weight of mice in the LR1 group did not change significantly (P > 0.05). The lowest diarrhea rate was recorded in the LR1 group after infection with EIEC. The results showed that the number of pathogens in the control group was higher than that in the experimental groups. The sequence analysis and OTU classification showed that the duodenum, ileum, and cecum of mice in the LR1 group had the highest number of OTUs compared with other groups. Whereas, the diversity analysis showed that in duodenum, ileum and cecum of mice in the LR1 group had the highest abundance and diversity. The composition of intestinal microbes indicated the presence of high proportions of Firmicutes, Proteobacteria and Bacteroidetes. Heat map analysis indicated high abundance of Bdello vibrio in the duodenum of mice in the LR1 group, while many pathogens were found in the different part of intestines in the control group, such as Streptococcus, Clostridium and Pseudomonas. In conclusion, pre-supplementation of LR1 alleviate the clinical symptoms caused by E. coli, and promote a healthy gut flora.
Collapse
Affiliation(s)
- Yaping Wang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, PR China
| | - Aoyun Li
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, PR China
| | - Lihong Zhang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, PR China
| | - Muhammad Waqas
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, PR China
| | - Khalid Mehmood
- University College of Veterinary & Animal Sciences, Islamia University of Bahawalpur, 61100, Pakistan
| | - Mudassar Iqbal
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, PR China; University College of Veterinary & Animal Sciences, Islamia University of Bahawalpur, 61100, Pakistan
| | - Can Muyou
- Institute of Grass Science, Tibet Academy of Agricultural and Animal Husbandry Sciences, Tibet, 860000, PR China
| | - Zhixing Li
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, PR China
| | - Yi Lian
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, PR China
| | - Suolang Sizhu
- College of Animals Husbandry and Veterinary Medicine, Tibet Agriculture and Animal Husbandry University, Linzhi, Tibet, 860000, PR China
| | - Jiakui Li
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, PR China; College of Animals Husbandry and Veterinary Medicine, Tibet Agriculture and Animal Husbandry University, Linzhi, Tibet, 860000, PR China.
| |
Collapse
|
24
|
Salameh E, Morel FB, Zeilani M, Déchelotte P, Marion-Letellier R. Animal Models of Undernutrition and Enteropathy as Tools for Assessment of Nutritional Intervention. Nutrients 2019; 11:nu11092233. [PMID: 31527523 PMCID: PMC6770013 DOI: 10.3390/nu11092233] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 08/24/2019] [Accepted: 09/08/2019] [Indexed: 02/06/2023] Open
Abstract
: Undernutrition is a major public health problem leading to 1 in 5 of all deaths in children under 5 years. Undernutrition leads to growth stunting and/or wasting and is often associated with environmental enteric dysfunction (EED). EED mechanisms leading to growth failure include intestinal hyperpermeability, villus blunting, malabsorption and gut inflammation. As non-invasive methods for investigating gut function in undernourished children are limited, pre-clinical models are relevant to elucidating the pathophysiological processes involved in undernutrition and EED, and to identifying novel therapeutic strategies. In many published models, undernutrition was induced using protein or micronutrient deficient diets, but these experimental models were not associated with EED. Enteropathy models mainly used gastrointestinal injury triggers. These models are presented in this review. We found only a few studies investigating the combination of undernutrition and enteropathy. This highlights the need for further developments to establish an experimental model reproducing the impact of undernutrition and enteropathy on growth, intestinal hyperpermeability and inflammation, that could be suitable for preclinical evaluation of innovative therapeutic intervention.
Collapse
Affiliation(s)
- Emmeline Salameh
- UniRouen, Inserm UMR 1073 Nutrition, Inflammation and Gut-Brain Axis, Normandie University, 76183 Rouen, France.
- Nutriset SAS, 76770 Malaunay, France.
| | | | | | - Pierre Déchelotte
- UniRouen, Inserm UMR 1073 Nutrition, Inflammation and Gut-Brain Axis, Normandie University, 76183 Rouen, France.
- Department of Nutrition, Rouen University Hospital, 76183 Rouen, France.
| | - Rachel Marion-Letellier
- UniRouen, Inserm UMR 1073 Nutrition, Inflammation and Gut-Brain Axis, Normandie University, 76183 Rouen, France.
| |
Collapse
|
25
|
Forgie AJ, Fouhse JM, Willing BP. Diet-Microbe-Host Interactions That Affect Gut Mucosal Integrity and Infection Resistance. Front Immunol 2019; 10:1802. [PMID: 31447837 PMCID: PMC6691341 DOI: 10.3389/fimmu.2019.01802] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 07/17/2019] [Indexed: 12/17/2022] Open
Abstract
The gastrointestinal tract microbiome plays a critical role in regulating host innate and adaptive immune responses against pathogenic bacteria. Disease associated dysbiosis and environmental induced insults, such as antibiotic treatments can lead to increased susceptibility to infection, particularly in a hospital setting. Dietary intervention is the greatest tool available to modify the microbiome and support pathogen resistance. Some dietary components can maintain a healthy disease resistant microbiome, whereas others can contribute to an imbalanced microbial population, impairing intestinal barrier function and immunity. Characterizing the effects of dietary components through the host-microbe axis as it relates to gastrointestinal health is vital to provide evidence-based dietary interventions to mitigate infections. This review will cover the effect of dietary components (carbohydrates, fiber, proteins, fats, polyphenolic compounds, vitamins, and minerals) on intestinal integrity and highlight their ability to modulate host-microbe interactions as to improve pathogen resistance.
Collapse
Affiliation(s)
| | | | - Benjamin P. Willing
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
26
|
Kastl AJ, Terry NA, Wu GD, Albenberg LG. The Structure and Function of the Human Small Intestinal Microbiota: Current Understanding and Future Directions. Cell Mol Gastroenterol Hepatol 2019; 9:33-45. [PMID: 31344510 PMCID: PMC6881639 DOI: 10.1016/j.jcmgh.2019.07.006] [Citation(s) in RCA: 200] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 07/17/2019] [Accepted: 07/17/2019] [Indexed: 02/07/2023]
Abstract
Despite growing literature characterizing the fecal microbiome and its association with health and disease, few studies have analyzed the microbiome of the small intestine. Here, we examine what is known about the human small intestinal microbiota in terms of community structure and functional properties. We examine temporal dynamics of select bacterial populations in the small intestine, and the effects of dietary carbohydrates and fats on shaping these populations. We then evaluate dysbiosis in the small intestine in several human disease models, including small intestinal bacterial overgrowth, short-bowel syndrome, pouchitis, environmental enteric dysfunction, and irritable bowel syndrome. What is clear is that the bacterial biology, and mechanisms of bacteria-induced pathophysiology, are enormously broad and elegant in the small intestine. Studying the small intestinal microbiota is challenged by rapidly fluctuating environmental conditions in these intestinal segments, as well as the complexity of sample collection and bioinformatic analysis. Because the functionality of the digestive tract is determined primarily by the small intestine, efforts must be made to better characterize this unique and important microbial ecosystem.
Collapse
Affiliation(s)
- Arthur J. Kastl
- Division of Gastroenterology, Hepatology, and Nutrition, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania,Correspondence Address correspondence to: Arthur J. Kastl Jr, MD, Division of Gastroenterology, Hepatology, and Nutrition, The Children’s Hospital of Philadelphia, 3401 Civic Center Boulevard, 7NW, Philadelphia, Pennsylvania 19104. fax: (215) 590-3606.
| | - Natalie A. Terry
- Division of Gastroenterology, Hepatology, and Nutrition, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Gary D Wu
- Division of Gastroenterology, Hepatology, and Nutrition, The University of Pennsylvania, Philadelphia, Pennsylvania
| | - Lindsey G. Albenberg
- Division of Gastroenterology, Hepatology, and Nutrition, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| |
Collapse
|
27
|
Abstract
PURPOSE OF REVIEW The current review is to update the results on epidemiology, pathobiology, and genes related to virulence, clinical presentation, molecular diagnosis, antimicrobial resistance, and extraintestinal infection of enteroaggregative Escherichia coli (EAEC). RECENT FINDINGS EAEC subclinical infection was significantly associated with reduced length at 2 years of age and EAEC and coinfections were associated with reduced delta weight-for-length and weight-for-age z-scores in the first 6 months of age in the MAL-ED birth cohort study. EAEC was associated with malnutrition in children 6-24 months of age in prospective case-control studies in Bangladesh and Brazil. Virulence gene-based studies have suggested aggregative fimbriae II may be a major contributor to disease, whereas AggR-activated regulator a marker of less severe disease. The high ability of EAEC colonization likely exacerbates effects of other microbial virulence strategies. Molecular diagnosis has been useful for understanding EAEC burden, although different criteria may relate to different pathogenic outcomes. SUMMARY EAEC gained special interest in the past few years, especially due to association with growth decrements in children with subclinical infections and its important role as a copathogen. Understanding of EAEC pathogenesis advanced but further research is needed for elucidating both microbial and host factors influencing infection outcomes.
Collapse
|
28
|
Wang Y, Li A, Liu J, Mehmood K, Wangdui B, Shi H, Luo X, Zhang H, Li J. L. pseudomesenteroides and L. johnsonii isolated from yaks in Tibet modulate gut microbiota in mice to ameliorate enteroinvasive Escherichia coli-induced diarrhea. Microb Pathog 2019; 132:1-9. [PMID: 30999021 DOI: 10.1016/j.micpath.2019.04.020] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 03/17/2019] [Accepted: 04/11/2019] [Indexed: 01/07/2023]
Abstract
Enteroinvasive Escherichia coli (EIEC) are well-known food-borne pathogens that cause animal intestinal diseases. Lactobacillus is believed to inhibit intestinal pathogens and maintain a healthy gut microbiota. This study aimed to investigate the effects of pre-supplementation of Lactobacillus from yaks (4500m) to prevent the clinical symptoms and the improvement of the disordered flora caused by E. coli infection. Forty healthy mice were randomized to four study groups (n = 10); Leuconostoc pseudomesenteroides (LP1), Lactobacillus johnsonii (LJ1), blank control, and control groups. Mice in the LP1, LJ1, and control groups were intraperitoneally challenged with EIEC O124 (1 × 109 CFU) on day 23. After two days, the mice in control group were recorded for high mortality. The diarrhea in LP1 and LJ1 groups was much lower than that in the control group, and no death was recorded. In histopathology, pre-supplementation of LJ1 and LP1 relieved the damage to the liver, spleen and duodenum caused by E. coli. In addition, the normal intestinal microecology was also affected by infection of EIEC, including an increase in relative abundance of Proteobacteria. At the same time, the beneficial bacteria were increased and harmful bacteria were decreased in different intestinal segments of the LJ1 and LP1 groups compared to the control group. In conclusion, pre-supplementation of LP1 and LJ1 can mitigate EIEC-induced intestinal flora dysbiosis and can also reduce EIEC-associated diarrhea.
Collapse
Affiliation(s)
- Yaping Wang
- College of Animals Husbandry and Veterinary Medicine, Tibet Agriculture and Animal Husbandry University, Linzhi, Tibet, 860000, PR China; College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, PR China
| | - Aoyun Li
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, PR China
| | - Juanjuan Liu
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, PR China
| | - Khalid Mehmood
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, PR China; (c)University College of Veterinary & Animal Sciences, Islamia University of Bahawalpur, 63100, Pakistan
| | - Basang Wangdui
- Institute of Animal Husbandry, Academy of Agriculture and Animal Husbandry, Tibet Autonomous Region, PR China
| | - Hongmei Shi
- Animal Husbandry and Veterinary Science Research Institute of Gannan Prefecture, PR China
| | - Xiaolin Luo
- Sichuan Academy of Grassland Science, Chengdu, Sichuan, 624400, PR China
| | - Hui Zhang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, PR China
| | - Jiakui Li
- College of Animals Husbandry and Veterinary Medicine, Tibet Agriculture and Animal Husbandry University, Linzhi, Tibet, 860000, PR China; College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, PR China.
| |
Collapse
|
29
|
DeBoer MD, Guerrant RL. Ongoing Challenges to Understanding and Interrupting Environmental Enteric Dysfunction. J Pediatr 2019; 210:8-9. [PMID: 30926153 DOI: 10.1016/j.jpeds.2019.02.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 02/27/2019] [Indexed: 11/20/2022]
Affiliation(s)
- Mark D DeBoer
- Pediatric Endocrinology, Department of Pediatrics, University of Virginia, Charlottesville, Virginia
| | - Richard L Guerrant
- Infectious Diseases and International Health, Department of Medicine and, Center for Global Health, University of Virginia, Charlottesville, Virginia.
| |
Collapse
|
30
|
Smith AD, Panickar KS, Urban JF, Dawson HD. Impact of Micronutrients on the Immune Response of Animals. Annu Rev Anim Biosci 2019; 6:227-254. [PMID: 29447473 DOI: 10.1146/annurev-animal-022516-022914] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Vitamins and minerals (micronutrients) play an important role in regulating and shaping an immune response. Deficiencies generally result in inadequate or dysregulated cellular activity and cytokine expression, thereby affecting the immune response. Decreased levels of natural killer, granulocyte, and phagocytic cell activity and T and B cell proliferation and trafficking are associated with inadequate levels of micronutrients, as well as increased susceptibility to various adverse health conditions, including inflammatory disorders, infection, and altered vaccine efficacy. In addition, most studies of micronutrient modulation of immune responses have been done in rodents and humans, thus limiting application to the health and well-being of livestock and companion animals. This exploratory review elucidates the role of vitamins and minerals on immune function and inflammatory responses in animals (pigs, dogs, cats, horses, goats, sheep, and cattle), with reference to rodents and humans.
Collapse
Affiliation(s)
- Allen D Smith
- United States Department of Agriculture, Agricultural Research Service, Beltsville Human Nutrition Research Center, Diet, Genomics & Immunology Laboratory, Beltsville, Maryland 20705-2350, USA;
| | - Kiran S Panickar
- Science & Technology Center, Hills Pet Nutrition Center, Topeka, Kansas 66617, USA
| | - Joseph F Urban
- United States Department of Agriculture, Agricultural Research Service, Beltsville Human Nutrition Research Center, Diet, Genomics & Immunology Laboratory, Beltsville, Maryland 20705-2350, USA;
| | - Harry D Dawson
- United States Department of Agriculture, Agricultural Research Service, Beltsville Human Nutrition Research Center, Diet, Genomics & Immunology Laboratory, Beltsville, Maryland 20705-2350, USA;
| |
Collapse
|
31
|
King SJ, McCole DF. Epithelial-microbial diplomacy: escalating border tensions drive inflammation in inflammatory bowel disease. Intest Res 2019; 17:177-191. [PMID: 30836737 PMCID: PMC6505084 DOI: 10.5217/ir.2018.00170] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Accepted: 02/01/2019] [Indexed: 02/07/2023] Open
Abstract
Inflammatory bowel diseases (IBD) are chronic conditions of the gastrointestinal tract-the main site of host-microbial interaction in the body. Development of IBD is not due to a single event but rather is a multifactorial process where a patient’s genetic background, behavioral habits, and environmental exposures contribute to disease pathogenesis. IBD patients exhibit alterations to gut bacterial populations “dysbiosis” due to the inflammatory microenvironment, however whether this alteration of the gut microbiota precedes inflammation has not been confirmed. Emerging evidence has highlighted the important role of gut microbes in developing measured immune responses and modulating other host responses such as metabolism. Much of the work on the gut microbiota has been correlative and there is an increasing need to understand the intimate relationship between host and microbe. In this review, we highlight how commensal and pathogenic bacteria interact with host intestinal epithelial cells and explore how altered microenvironments impact these connections.
Collapse
Affiliation(s)
- Stephanie J King
- Division of Biomedical Sciences, University of California, Riverside, CA, USA
| | - Declan F McCole
- Division of Biomedical Sciences, University of California, Riverside, CA, USA
| |
Collapse
|
32
|
Q.S. Medeiros PH, Ledwaba SE, Bolick DT, Giallourou N, Yum LK, Costa DV, Oriá RB, Barry EM, Swann JR, Lima AÂM, Agaisse H, Guerrant RL. A murine model of diarrhea, growth impairment and metabolic disturbances with Shigella flexneri infection and the role of zinc deficiency. Gut Microbes 2019; 10:615-630. [PMID: 30712505 PMCID: PMC6748602 DOI: 10.1080/19490976.2018.1564430] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Shigella is one of the major enteric pathogens worldwide. We present a murine model of S. flexneri infection and investigate the role of zinc deficiency (ZD). C57BL/6 mice fed either standard chow (HC) or ZD diets were pretreated with an antibiotic cocktail and received S. flexneri strain 2457T orally. Antibiotic pre-treated ZD mice showed higher S. flexneri colonization than non-treated mice. ZD mice showed persistent colonization for at least 50 days post-infection (pi). S. flexneri-infected mice showed significant weight loss, diarrhea and increased levels of fecal MPO and LCN in both HC and ZD fed mice. S. flexneri preferentially colonized the colon, caused epithelial disruption and inflammatory cell infiltrate, and promoted cytokine production which correlated with weight loss and histopathological changes. Infection with S. flexneri ΔmxiG (critical for type 3 secretion system) did not cause weight loss or diarrhea, and had decreased stool shedding duration and tissue burden. Several biochemical changes related to energy, inflammation and gut-microbial metabolism were observed. Zinc supplementation increased weight gains and reduced intestinal inflammation and stool shedding in ZD infected mice. In conclusion, young antibiotic-treated mice provide a new model of oral S. flexneri infection, with ZD promoting prolonged infection outcomes.
Collapse
Affiliation(s)
- Pedro Henrique Q.S. Medeiros
- Center for Global Health, Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, USA,Institute of Biomedicine, Federal University of Ceara, Fortaleza, Brazil,CONTACT Pedro Henrique Q.S. Medeiros Center for Global Health, Division of Infectious Diseases and International Health, University of Virginia, 345 Crispell Drive, MR6 Room 2711, Charlottesville, VA, USA
| | - Solanka E. Ledwaba
- Center for Global Health, Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, USA
| | - David T. Bolick
- Center for Global Health, Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, USA
| | - Natasa Giallourou
- Division of Computational and Systems Medicine, Department of Surgery and Cancer, Imperial College London, London, UK
| | - Lauren K. Yum
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, USA
| | - Deiziane V.S. Costa
- Center for Global Health, Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, USA,Institute of Biomedicine, Federal University of Ceara, Fortaleza, Brazil
| | - Reinaldo B. Oriá
- Center for Global Health, Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, USA,Institute of Biomedicine, Federal University of Ceara, Fortaleza, Brazil
| | - Eileen M. Barry
- Center for Vaccine Development, University of Maryland, Baltimore, USA
| | - Jonathan R. Swann
- Division of Computational and Systems Medicine, Department of Surgery and Cancer, Imperial College London, London, UK
| | | | - Hervé Agaisse
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, USA
| | - Richard L. Guerrant
- Center for Global Health, Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, USA
| |
Collapse
|
33
|
Alanyl-glutamine Protects Against Damage Induced by Enteroaggregative Escherichia coli Strains in Intestinal Cells. J Pediatr Gastroenterol Nutr 2019; 68:190-198. [PMID: 30247422 DOI: 10.1097/mpg.0000000000002152] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
BACKGROUND Enteroaggregative Escherichia coli (EAEC) is an important pathogen causing enteric infections worldwide. This pathotype is linked to malnutrition in children from developing countries. Alanyl-glutamine (Ala-Gln) is an immune modulator nutrient that acts during intestinal damage and/or inflammation. This study investigated the effect of EAEC infection and Ala-Gln on cell viability, cell death, and inflammation of intestinal epithelium cells (IEC-6). METHODS Cells were infected with an EAEC prototype 042 strain, an EAEC wild-type strain isolated from a Brazilian malnourished child, and a commensal E coli HS. Gene transcription and protein levels of caspases-3, -8, and -9 and cytokine-induced neutrophil chemoattractant 1 (CINC-1/CXCL1) were evaluated using RT-qPCR, western blot analysis, and ELISA. RESULTS Infections with both EAEC strains decreased cell viability and induced apoptosis and necrosis after 24 hours. Ala-Gln supplementation increased cell proliferation and reduced cell death in infected cells. Likewise, EAEC strain 042 significantly increased the transcript levels of caspases-3, -8, and -9 when compared to the control group, and Ala-Gln treatment reversed this effect. Furthermore, EAEC induced CXCL1 protein levels, which were also reduced by Ala-Gln supplementation. CONCLUSION These findings suggest that EAEC infection promotes apoptosis, necrosis, and intestinal inflammation with involvement of caspases. Supplementation of Ala-Gln inhibits cell death, increases cell proliferation, attenuates mediators associated with cell death, and inflammatory pathways in infected cells.
Collapse
|
34
|
Bartelt LA, Bolick DT, Guerrant RL. Disentangling Microbial Mediators of Malnutrition: Modeling Environmental Enteric Dysfunction. Cell Mol Gastroenterol Hepatol 2019; 7:692-707. [PMID: 30630118 PMCID: PMC6477186 DOI: 10.1016/j.jcmgh.2018.12.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 12/12/2018] [Accepted: 12/13/2018] [Indexed: 12/12/2022]
Abstract
Environmental enteric dysfunction (EED) (also referred to as environmental enteropathy) is a subclinical chronic intestinal disorder that is an emerging contributor to early childhood malnutrition. EED is common in resource-limited settings, and is postulated to consist of small intestinal injury, dysfunctional nutrient absorption, and chronic inflammation that results in impaired early child growth attainment. Although there is emerging interest in the hypothetical potential for chemical toxins in the environmental exposome to contribute to EED, the propensity of published data, and hence the focus of this review, implicates a critical role of environmental microbes. Early childhood malnutrition and EED are most prevalent in resource-limited settings where food is limited, and inadequate access to clean water and sanitation results in frequent gastrointestinal pathogen exposures. Even as overt diarrhea rates in these settings decline, silent enteric infections and faltering growth persist. Furthermore, beyond restricted physical growth, EED and/or enteric pathogens also associate with impaired oral vaccine responses, impaired cognitive development, and may even accelerate metabolic syndrome and its cardiovascular consequences. As these potentially costly long-term consequences of early childhood enteric infections increasingly are appreciated, novel therapeutic strategies that reverse damage resulting from nutritional deficiencies and microbial insults in the developing small intestine are needed. Given the inherent limitations in investigating how specific intestinal pathogens directly injure the small intestine in children, animal models provide an affordable and controlled opportunity to elucidate causal sequelae of specific enteric infections, to differentiate consequences of defined nutrient deprivation alone from co-incident enteropathogen insults, and to correlate the resulting gut pathologies with their functional impact during vulnerable early life windows.
Collapse
Affiliation(s)
- Luther A Bartelt
- Division of Infectious Diseases, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Center for Gastrointestinal Biology and Disease, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.
| | - David T Bolick
- Center for Global Health, Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, Virginia
| | - Richard L Guerrant
- Center for Global Health, Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
35
|
Wang Y, Li A, Jiang X, Zhang H, Mehmood K, Zhang L, Jiang J, Waqas M, Iqbal M, Li J. Probiotic Potential of Leuconostoc pseudomesenteroides and Lactobacillus Strains Isolated From Yaks. Front Microbiol 2018; 9:2987. [PMID: 30564222 PMCID: PMC6289064 DOI: 10.3389/fmicb.2018.02987] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 11/19/2018] [Indexed: 12/26/2022] Open
Abstract
The purpose of this study was to evaluate the antibacterial activity and safety of bacterias with probiotic potential isolated from free-ranging Tibetan yaks in high altitude regions of Tibet. For this purpose, one Leuconostoc pseudomesenteroides strain (named P1) and two Lactobacillus johnsonii and Lactobacillus mucosae strains (named LY1 and LY2), respectively, were isolated from fecal samples of Tibetan yaks. The antibacterial activity of the isolates was studied using Escherichia coli (E. coli ATCC 25922), Staphylococcus aureus (S. aureus ATCC 26112), and Salmonella enteritidis (S. enteritidis NCTC 13349) as indicator pathogens. The results showed that LY1 had high antibacterial efficacy against E. coli and S. enteritidis, while P1 had the most powerful bacteriostatic ability against S. aureus. PCR amplification showed that all the isolated strains were positive for Ent P2 (enterocin P-like bacteriocin) and exhibited a high tolerance to bile and low pH. Moreover, the safety of P1, LY1, and LY2 was determined through antibiotic resistance experiments, resistance gene testing, and hemolytic analysis while the antibacterial activity was assessed by in vitro and in vivo experiments. The LY2 strain was abandoned as a potential probiotic due to the detection of the vanA gene. The mice were fed from days 1 to 30 in six groups, the P1-1 (gavaged with P1 1 × 108 CFU/day), P1-2 (gavaged with P1 1 × 109 CFU/day), LY1-1 (gavaged with LY1 1 × 108 CFU/day), LY1-2 (gavaged with LY1 1 × 109 CFU/day), control (gavaged with an equal volume of vehicle), and blank control (gavaged with an equal volume of vehicle) groups. After 30 days, mice in the P1-1, P1-2, LY1-1, LY1-2, and control groups were intraperitoneal challenged with 1 × 108 CFU of E. coli (n = 10) in the abdomen. After 2 days of infection, the mice in the control group showed more severe damage in the liver, spleen and intestine than the mice in the P1-2 and LY1-2 groups. The mice in the P1-2 and LY1-2 groups had lower rates of diarrhea and mortality than other groups. In conclusion, bacteria with probiotic potential isolated from yaks may possibly be effective and safe antibacterial substances, providing a new treatment method to reduce the incidence of diarrhea associated with bacterial diseases in yaks.
Collapse
Affiliation(s)
- Yaping Wang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Aoyun Li
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Xiong Jiang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Hubei Three Gorges Polytechnic, Yichang, China
| | - Hui Zhang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Khalid Mehmood
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- University College of Veterinary and Animal Sciences, The Islamia University of Bahawalpur Pakistan, Bahawalpur, Pakistan
| | - Lihong Zhang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Jinhuan Jiang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Muhammad Waqas
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Mujahid Iqbal
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Jiakui Li
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- College of Animals Husbandry and Veterinary Medicine, Tibet Agriculture and Animal Husbandry University, Linzhi, China
| |
Collapse
|
36
|
Pongkorpsakol P, Buasakdi C, Chantivas T, Chatsudthipong V, Muanprasat C. An agonist of a zinc-sensing receptor GPR39 enhances tight junction assembly in intestinal epithelial cells via an AMPK-dependent mechanism. Eur J Pharmacol 2018; 842:306-313. [PMID: 30459126 DOI: 10.1016/j.ejphar.2018.10.038] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 10/17/2018] [Accepted: 10/25/2018] [Indexed: 02/08/2023]
Abstract
Intestinal barrier function depends on integrity of tight junctions, which serve as barriers to transepithelial influx of noxious substances/microorganisms from gut lumen. The G-protein coupled receptor 39 (GPR39) is a zinc-sensing receptor, which is expressed in several cell types including intestinal epithelial cells (IECs). The main objective of this study was to investigate the effect of GPR39 activation on tight junction assembly in IECs. Treatment with TC-G 1008 (1 μM -10 μM), a GPR39 agonist, and zinc (10 μM -100 μM) increased tight junction assembly in T84 cells. This effect was suppressed by pretreatment with compound C, an inhibitor of AMP-activated protein kinase (AMPK). In addition, western blot analysis revealed that treatment with TC-G 1008 induced AMPK activation in time- and concentration-dependent manners. Interestingly, inhibitors of phospholipase C (PLC) and calcium/calmodulin-dependent protein kinase kinase β (CaMKKβ) abrogated the effect of TC-G 1008 on inducing AMPK activation, tight junction assembly and zonula occludens-1 re-organization. Collectively, this study reveals a novel role of GPR39 in enhancing tight junction assembly in IECs via PLC-CaMKKβ-AMPK pathways. GPR39 agonists may be beneficial in the treatment of diseases associated impaired intestinal barrier function.
Collapse
Affiliation(s)
- Pawin Pongkorpsakol
- Translational Medicine Graduate Program, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Rama VI Road, Rajathevi, Bangkok 10400, Thailand
| | - Chavin Buasakdi
- College of Agricultural and Life Science, University of Wisconsin-Madison, 1450 Linden Dr, Madison, WI 53706, USA
| | - Thanyatorn Chantivas
- Faculty of Medicine Ramathibodi Hospital, Mahidol University, Rama VI Road, Rajathevi, Bangkok 10400, Thailand
| | - Varanuj Chatsudthipong
- Department of Physiology, Faculty of Science, Mahidol University, Rama VI Road, Rajathevi, Bangkok 10400, Thailand
| | - Chatchai Muanprasat
- Translational Medicine Graduate Program, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Rama VI Road, Rajathevi, Bangkok 10400, Thailand; Department of Physiology, Faculty of Science, Mahidol University, Rama VI Road, Rajathevi, Bangkok 10400, Thailand; Excellent Center for Drug Discovery, Faculty of Science, Mahidol University, Rama VI Road, Rajathevi, Bangkok 10400, Thailand; Research Center of Transport Protein for Medical Innovation, Faculty of Science, Mahidol University, Rama VI Road, Rajathevi, Bangkok 10400, Thailand.
| |
Collapse
|
37
|
Essential Role of Zinc and Zinc Transporters in Myeloid Cell Function and Host Defense against Infection. J Immunol Res 2018; 2018:4315140. [PMID: 30417019 PMCID: PMC6207864 DOI: 10.1155/2018/4315140] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 09/27/2018] [Indexed: 12/19/2022] Open
Abstract
Zinc is an essential micronutrient known to play a vital role in host defense against pathogens. Diets that are deficient in zinc lead to impaired immunity and delayed recovery from and worse outcomes following infection. Sustained insufficient zinc intake leads to dysregulation of the innate immune response and increases susceptibility to infection whereas zinc supplementation in at-risk populations has been shown to restore host defense and reduce pathogen-related morbidity and mortality. Upon infection, zinc deficiency leads to increased pathology due to imbalance in key signaling networks that result in excessive inflammation and collateral tissue damage. In particular, zinc impacts macrophage function, a critical front-line cell in host defense, in addition to other immune cells. Deficits in zinc adversely impact macrophage function resulting in dysregulation of phagocytosis, intracellular killing, and cytokine production. An additional work in this field has revealed a vital role for several zinc transporter proteins that are required for proper bioredistribution of zinc within mononuclear cells to achieve an optimal immune response against invading microorganisms. In this review, we will discuss the most recent developments regarding zinc's role in innate immunity and protection against pathogen invasion.
Collapse
|
38
|
Rerksuppaphol S, Rerksuppaphol L. A Randomized Controlled Trial of Zinc Supplementation as Adjuvant Therapy for Dengue Viral Infection in Thai Children. Int J Prev Med 2018; 9:88. [PMID: 30450171 PMCID: PMC6202777 DOI: 10.4103/ijpvm.ijpvm_367_17] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 06/26/2018] [Indexed: 11/16/2022] Open
Abstract
Background: Zinc deficiency is common in developing countries and increases the risk for several infectious diseases. Low serum zinc levels have been reported in children with dengue virus infection (DVI). This study aimed to assess the effects of zinc supplementation on DVI outcomes. Methods: A double-blinded, randomized trial was conducted in 50 children with dengue fever (DF)/dengue hemorrhagic fever admitted to the pediatric unit of MSMC Srinakharinwirot University Hospital, Thailand, between January 2016 and April 2017. Bis-glycinate zinc or placebo was orally administered three times a day for 5 days or until defervescence. The primary outcome was to evaluate the DVI defervescence phase; the secondary outcome was to assess hospitalization length and presence of severe DVI and zinc deficiency. Results: The mean time of defervescence was 29.2 ± 24.0 h in the supplementation group and 38.1 ± 31.5 h in the placebo group (P = 0.270). Meantime of hospital staying was 62.5 ± 23.8 h in the supplementation group and 84.7 ± 34.0 h in placebo group with the mean difference of hospital staying between groups of 22.2 h (95% confidence interval [CI]: 5.5–38.5 h; P = 0.010). Overall prevalence of zinc deficiency was 46%. Serum zinc levels increased from baseline to the end of the study. the mean gain was 26.4 μg/dL (95% CI: 13.6–39.1 μg/dL) in the supplementation group and 14.4 μg/dL (95% CI: 7.4–21.3 μg/dL) in placebo group. No signs of severe DVI were observed in both groups. Zinc supplementation was well tolerated. Conclusions: Overcoming zinc deficiency among Thai children may reduce DF duration and limit the hospitalization, in addition to other advantages that normal serum zinc levels have on overall children health.
Collapse
Affiliation(s)
| | - Lakkana Rerksuppaphol
- Department of Preventive Medicine, Faculty of Medicine, Srinakharinwirot University, Bangkok, Thailand
| |
Collapse
|
39
|
Critical Role of Zinc in a New Murine Model of Enterotoxigenic Escherichia coli Diarrhea. Infect Immun 2018; 86:IAI.00183-18. [PMID: 29661930 PMCID: PMC6013668 DOI: 10.1128/iai.00183-18] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 04/11/2018] [Indexed: 12/11/2022] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) is a major cause of traveler's diarrhea as well as of endemic diarrhea and stunting in children in developing areas. However, a small-mammal model has been badly needed to better understand and assess mechanisms, vaccines, and interventions. We report a murine model of ETEC diarrhea, weight loss, and enteropathy and investigate the role of zinc in the outcomes. ETEC strains producing heat-labile toxins (LT) and heat-stable toxins (ST) that were given to weaned C57BL/6 mice after antibiotic disruption of normal microbiota caused growth impairment, watery diarrhea, heavy stool shedding, and mild to moderate intestinal inflammation, the latter being worse with zinc deficiency. Zinc treatment promoted growth in zinc-deficient infected mice, and subinhibitory levels of zinc reduced expression of ETEC virulence genes cfa1, cexE, sta2, and degP but not of eltA in vitro Zinc supplementation increased shedding and the ileal burden of wild-type (WT) ETEC but decreased shedding and the tissue burden of LT knockout (LTKO) ETEC. LTKO ETEC-infected mice had delayed disease onset and also had less inflammation by fecal myeloperoxidase (MPO) assessment. These findings provide a new murine model of ETEC infection that can help elucidate mechanisms of growth, diarrhea, and inflammatory responses as well as potential vaccines and interventions.
Collapse
|
40
|
Abstract
Transition metals are required cofactors for many proteins that are critical for life, and their concentration within cells is carefully maintained to avoid both deficiency and toxicity. To defend against bacterial pathogens, vertebrate immune proteins sequester metals, in particular zinc, iron, and manganese, as a strategy to limit bacterial acquisition of these necessary nutrients in a process termed "nutritional immunity." In response, bacteria have evolved elegant strategies to access metals and counteract this host defense. In mammals, metal abundance can drastically shift due to changes in dietary intake or absorption from the intestinal tract, disrupting the balance between host and pathogen in the fight for metals and altering susceptibility to disease. This review describes the current understanding of how dietary metals modulate host-microbe interactions and the subsequent impact on the outcome of disease.
Collapse
Affiliation(s)
- Christopher A Lopez
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Eric P Skaar
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA; Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
41
|
Giallourou N, Medlock GL, Bolick DT, Medeiros PHQS, Ledwaba SE, Kolling GL, Tung K, Guerry P, Swann JR, Guerrant RL. A novel mouse model of Campylobacter jejuni enteropathy and diarrhea. PLoS Pathog 2018; 14:e1007083. [PMID: 29791507 PMCID: PMC5988333 DOI: 10.1371/journal.ppat.1007083] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 06/05/2018] [Accepted: 05/09/2018] [Indexed: 01/31/2023] Open
Abstract
Campylobacter infections are among the leading bacterial causes of diarrhea and of 'environmental enteropathy' (EE) and growth failure worldwide. However, the lack of an inexpensive small animal model of enteric disease with Campylobacter has been a major limitation for understanding its pathogenesis, interventions or vaccine development. We describe a robust standard mouse model that can exhibit reproducible bloody diarrhea or growth failure, depending on the zinc or protein deficient diet and on antibiotic alteration of normal microbiota prior to infection. Zinc deficiency and the use of antibiotics create a niche for Campylobacter infection to establish by narrowing the metabolic flexibility of these mice for pathogen clearance and by promoting intestinal and systemic inflammation. Several biomarkers and intestinal pathology in this model also mimic those seen in human disease. This model provides a novel tool to test specific hypotheses regarding disease pathogenesis as well as vaccine development that is currently in progress.
Collapse
Affiliation(s)
- Natasa Giallourou
- Division of Computational and Systems Medicine, Department of Surgery and Cancer, Imperial College, London, United Kingdom
| | - Gregory L. Medlock
- Department of Biomedical Engineering, University of Virginia School of Medicine, Charlottesville, VA, United States of America
| | - David T. Bolick
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, United States of America
| | - Pedro HQS Medeiros
- Institute of Biomedicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Solanka E. Ledwaba
- Department of Microbiology, University of Venda, Thohoyandou, Limpopo, South Africa
| | - Glynis L. Kolling
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, United States of America
| | - Kenneth Tung
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, VA, United States of America
| | - Patricia Guerry
- Enteric Diseases Department, Naval Medical Research Center, Silver Spring, MD, United States of America
| | - Jonathan R. Swann
- Division of Computational and Systems Medicine, Department of Surgery and Cancer, Imperial College, London, United Kingdom
| | - Richard L. Guerrant
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, United States of America
| |
Collapse
|
42
|
Nataro JP, Guerrant RL. Chronic consequences on human health induced by microbial pathogens: Growth faltering among children in developing countries. Vaccine 2017; 35:6807-6812. [DOI: 10.1016/j.vaccine.2017.05.035] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 03/28/2017] [Accepted: 05/10/2017] [Indexed: 02/07/2023]
|
43
|
Abstract
Environmental enteropathy is a chronic condition of the small intestine associated with increased intestinal permeability, mucosal inflammation, malabsorption, and systemic inflammation. It is commonly accompanied by enteric infections and is misleadingly considered a subclinical disease. Potential effects of enteric infections and enteropathy on vaccine responses, child growth, cognitive development, and even later life obesity, diabetes, and metabolic syndrome are increasingly being recognized. Herein, we review the evolving challenges to defining environmental enteropathy and enteric infections, current evidence for the magnitude and determinants of its burden, new assessment tools, and relevant interventions.
Collapse
Affiliation(s)
- Elizabeth T Rogawski
- Department of Public Health Sciences, University of Virginia, PO Box 801379, Carter Harrison Research Building MR-6, 345 Crispell Drive, Room 2520, Charlottesville, VA 22908-1379, USA; Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, PO Box 801379, Carter Harrison Research Building MR-6, 345 Crispell Drive, Room 2520, Charlottesville, VA 22908-1379, USA.
| | - Richard L Guerrant
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, PO Box 801379, Carter Harrison Research Building MR-6, 345 Crispell Drive, Room 2520, Charlottesville, VA 22908-1379, USA
| |
Collapse
|
44
|
Bartelt LA, Bolick DT, Mayneris-Perxachs J, Kolling GL, Medlock GL, Zaenker EI, Donowitz J, Thomas-Beckett RV, Rogala A, Carroll IM, Singer SM, Papin J, Swann JR, Guerrant RL. Cross-modulation of pathogen-specific pathways enhances malnutrition during enteric co-infection with Giardia lamblia and enteroaggregative Escherichia coli. PLoS Pathog 2017; 13:e1006471. [PMID: 28750066 PMCID: PMC5549954 DOI: 10.1371/journal.ppat.1006471] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 06/14/2017] [Indexed: 12/17/2022] Open
Abstract
Diverse enteropathogen exposures associate with childhood malnutrition. To
elucidate mechanistic pathways whereby enteric microbes interact during
malnutrition, we used protein deficiency in mice to develop a new model of
co-enteropathogen enteropathy. Focusing on common enteropathogens in
malnourished children, Giardia lamblia and enteroaggregative
Escherichia coli (EAEC), we provide new insights into
intersecting pathogen-specific mechanisms that enhance malnutrition. We show for
the first time that during protein malnutrition, the intestinal microbiota
permits persistent Giardia colonization and simultaneously
contributes to growth impairment. Despite signals of intestinal injury, such as
IL1α, Giardia-infected mice lack pro-inflammatory intestinal
responses, similar to endemic pediatric Giardia infections.
Rather, Giardia perturbs microbial host co-metabolites of
proteolysis during growth impairment, whereas host nicotinamide utilization
adaptations that correspond with growth recovery increase. EAEC promotes
intestinal inflammation and markers of myeloid cell activation. During
co-infection, intestinal inflammatory signaling and cellular recruitment
responses to EAEC are preserved together with a
Giardia-mediated diminishment in myeloid cell activation.
Conversely, EAEC extinguishes markers of host energy expenditure regulatory
responses to Giardia, as host metabolic adaptations appear
exhausted. Integrating immunologic and metabolic profiles during co-pathogen
infection and malnutrition, we develop a working mechanistic model of how
cumulative diet-induced and pathogen-triggered microbial perturbations result in
an increasingly wasted host. Malnourished children are exposed to multiple sequential, and oftentimes,
persistent enteropathogens. Intestinal microbial disruption and inflammation are
known to contribute to the pathogenesis of malnutrition, but how co-pathogens
interact with each other, with the resident microbiota, or with the host to
alter these pathways is unknown. Using a new model of enteric co-infection with
Giardia lamblia and enteroaggregative Escherichia
coli in mice fed a protein deficient diet, we identify host growth
and intestinal immune responses that are differentially mediated by
pathogen-microbe interactions, including parasite-mediated changes in intestinal
microbial host co-metabolism, and altered immune responses during co-infection.
Our data model how early life cumulative enteropathogen exposures progressively
disrupt intestinal immunity and host metabolism during crucial developmental
periods. Furthermore, studies in this co-infection model reveal new insights
into environmental and microbial determinants of pathogenicity for presently
common, but poorly understood enteropathogens like Giardia
lamblia, that may not conform to existing paradigms of microbial
pathogenesis based on single pathogen-designed models.
Collapse
Affiliation(s)
- Luther A. Bartelt
- Division of Infectious Diseases, Department of Medicine, University of
North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of
America
- Center for Gastrointestinal Biology and Disease, Department of Medicine,
University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United
States of America
- * E-mail:
| | - David T. Bolick
- Division of Infectious Diseases and International Health, Department of
Medicine, University of Virginia, Charlottesville, Virginia, United States of
America
| | - Jordi Mayneris-Perxachs
- Division of Computational and Systems Medicine, Department of Surgery and
Cancer, Imperial College London, United Kingdom
| | - Glynis L. Kolling
- Division of Infectious Diseases and International Health, Department of
Medicine, University of Virginia, Charlottesville, Virginia, United States of
America
| | - Gregory L. Medlock
- Department of Biomedical Engineering, University of Virginia,
Charlottesville, Virginia, United States of America
| | - Edna I. Zaenker
- Division of Infectious Diseases and International Health, Department of
Medicine, University of Virginia, Charlottesville, Virginia, United States of
America
| | - Jeffery Donowitz
- Division of Pediatric Infectious Diseases, Children’s Hospital of
Richmond at Virginia Commonwealth University, Richmond, Virginia, United States
of America
| | - Rose Viguna Thomas-Beckett
- Division of Infectious Diseases, Department of Medicine, University of
North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of
America
| | - Allison Rogala
- Center for Gastrointestinal Biology and Disease, Department of Medicine,
University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United
States of America
| | - Ian M. Carroll
- Center for Gastrointestinal Biology and Disease, Department of Medicine,
University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United
States of America
| | - Steven M. Singer
- Department of Biology, Georgetown University, Washington, DC, United
States of America
| | - Jason Papin
- Department of Biomedical Engineering, University of Virginia,
Charlottesville, Virginia, United States of America
| | - Jonathan R. Swann
- Division of Computational and Systems Medicine, Department of Surgery and
Cancer, Imperial College London, United Kingdom
| | - Richard L. Guerrant
- Division of Infectious Diseases and International Health, Department of
Medicine, University of Virginia, Charlottesville, Virginia, United States of
America
| |
Collapse
|
45
|
Rogawski ET, Guerrant RL, Havt A, Lima IFN, Medeiros PHQS, Seidman JC, McCormick BJJ, Babji S, Hariraju D, Bodhidatta L, Shrestha J, Anania J, Maro A, Samie A, Yori PP, Qureshi S, Mahfuz M, Bessong PO, Kosek MN, Ahmed T, Bhutta ZA, Lang DR, Gottlieb M, Houpt ER, Lima AAM. Epidemiology of enteroaggregative Escherichia coli infections and associated outcomes in the MAL-ED birth cohort. PLoS Negl Trop Dis 2017; 11:e0005798. [PMID: 28742106 PMCID: PMC5542697 DOI: 10.1371/journal.pntd.0005798] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 08/03/2017] [Accepted: 07/11/2017] [Indexed: 11/23/2022] Open
Abstract
Background Enteroaggregative E. coli (EAEC) have been associated with mildly inflammatory diarrhea in outbreaks and in travelers and have been increasingly recognized as enteric pathogens in young children with and without overt diarrhea. We examined the risk factors for EAEC infections and their associations with environmental enteropathy biomarkers and growth outcomes over the first two years of life in eight low-resource settings of the MAL-ED study. Methods EAEC infections were detected by PCR gene probes for aatA and aaiC virulence traits in 27,094 non-diarrheal surveillance stools and 7,692 diarrheal stools from 2,092 children in the MAL-ED birth cohort. We identified risk factors for EAEC and estimated the associations of EAEC with diarrhea, enteropathy biomarker concentrations, and both short-term (one to three months) and long-term (to two years of age) growth. Results Overall, 9,581 samples (27.5%) were positive for EAEC, and almost all children had at least one detection (94.8%) by two years of age. Exclusive breastfeeding, higher enrollment weight, and macrolide use within the preceding 15 days were protective. Although not associated with diarrhea, EAEC infections were weakly associated with biomarkers of intestinal inflammation and more strongly with reduced length at two years of age (LAZ difference associated with high frequency of EAEC detections: -0.30, 95% CI: -0.44, -0.16). Conclusions Asymptomatic EAEC infections were common early in life and were associated with linear growth shortfalls. Associations with intestinal inflammation were small in magnitude, but suggest a pathway for the growth impact. Increasing the duration of exclusive breastfeeding may help prevent these potentially inflammatory infections and reduce the long-term impact of early exposure to EAEC. Enteroaggregative E. coli (EAEC) are pathogens that infect the intestine and can cause diarrhea. They are also commonly identified among young children in low-resource settings, who can carry the pathogen without symptomatic diarrhea. We examined the risk factors for EAEC infections and their associations with child health outcomes over the first two years of life in eight low-resource settings of the MAL-ED study. EAEC infections were detected using molecular methods in more than 30,000 stools collected from 2,092 children in the MAL-ED study. We identified risk factors for EAEC and estimated the associations of EAEC with diarrhea, markers of intestinal health, and child growth. Almost all children were infected with EAEC at least once by two years of age. Exclusive breastfeeding, higher enrollment weight, and recent macrolide antibiotic use were protective against these infections. Although not associated with diarrhea in these children, EAEC infections were associated with intestinal inflammation and reduced length at two years of age. EAEC may impact child development, even in the absence of diarrhea, by causing intestinal inflammation and impairing child growth.
Collapse
Affiliation(s)
- Elizabeth T. Rogawski
- Department of Public Health Sciences, University of Virginia, Charlottesville, Virginia, United States of America
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia, United States of America
- * E-mail:
| | - Richard L. Guerrant
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia, United States of America
| | - Alexandre Havt
- Clinical Research Unit and Institute of Biomedicine, Federal University of Ceara, Fortaleza, Brazil
| | - Ila F. N. Lima
- Clinical Research Unit and Institute of Biomedicine, Federal University of Ceara, Fortaleza, Brazil
| | - Pedro H. Q. S. Medeiros
- Clinical Research Unit and Institute of Biomedicine, Federal University of Ceara, Fortaleza, Brazil
| | - Jessica C. Seidman
- Fogarty International Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Benjamin J. J. McCormick
- Fogarty International Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Sudhir Babji
- Division of Gastrointestinal Sciences, Christian Medical College, Vellore, India
| | - Dinesh Hariraju
- Division of Gastrointestinal Sciences, Christian Medical College, Vellore, India
| | - Ladaporn Bodhidatta
- Department of Enteric Diseases, Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand
| | | | - Japhat Anania
- Haydom Global Health Research Center, Haydom Lutheran Hospital, Haydom, Tanzania
| | - Athanasia Maro
- Haydom Global Health Research Center, Haydom Lutheran Hospital, Haydom, Tanzania
| | - Amidou Samie
- Department of Microbiology, University of Venda, Thohoyandou, South Africa
| | | | - Shahida Qureshi
- Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Mustafa Mahfuz
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - Pascal O. Bessong
- Department of Microbiology, University of Venda, Thohoyandou, South Africa
| | - Margaret N. Kosek
- Asociación Benéfica PRISMA, Iquitos, Peru
- Department of International Health, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Tahmeed Ahmed
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - Zulfiqar A. Bhutta
- Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Dennis R. Lang
- Foundation for the National Institutes of Health, Bethesda, Maryland, United States of America
| | - Michael Gottlieb
- Foundation for the National Institutes of Health, Bethesda, Maryland, United States of America
| | - Eric R. Houpt
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia, United States of America
| | - Aldo A. M. Lima
- Clinical Research Unit and Institute of Biomedicine, Federal University of Ceara, Fortaleza, Brazil
| | | |
Collapse
|
46
|
Nyanga PL, Onyuka J, Webale MK, Were T, Budambula V. Escherichia coli pathotypes and Shigella sero-groups in diarrheic children in Nairobi city, Kenya. GASTROENTEROLOGY AND HEPATOLOGY FROM BED TO BENCH 2017; 10:220-228. [PMID: 29118939 PMCID: PMC5660273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
AIM In the present study, we investigated the prevalence of E. coli pathotypes and Shigella sero-groups and their antimicrobial profiles among diarrheic children in Nairobi city, Kenya. BACKGROUND Although diarrheagenic E. coli pathotypes and Shigella sero-groups are leading causes of diarrhea in children under five years in developing countries, their distribution and antimicrobial resistance vary from place to place and over time in a given region. METHODS In a cross-sectional study, we enrolled diarrheic children (n=354) under five years seeking treatment at Mbagathi Hospital, Nairobi city, Kenya,. Stool samples were collected from all children for bacterial culture. Bacterial isolation and identification was performed by conventional microbiological methods. Polymerase chain amplification was used to detect aspU, aggR, andpcvd432 for EAEC, est and elt for ETEC, eae for EPEC, stx for EHEC, and ipaH for EIEC and Shigella species. Antimicrobial profile was determined by disk diffusion method. RESULTS The prevalence of EAEC, ETEC, EPEC (eae), EIEC (ipaH) was 21.2%, 10.5%, 4.5%, and 0.6%, respectively, while that of mixed infection was 0.6%for ETEC/EAEC and 0.3%for EAEC/EPEC/ETEC. No EHEC strain was isolated. Pathogenetic analysis for EAEC showed that5.9% carried aspU,8.2% possessed both aspU and aggR and 7.1% had a combination of aspU, aggR andpcvd432 while that of ETEC was 2.3% for elt, 6.5% for both elt and est and 1.7% for est. The combination of aspU with aggR, elt and est, and pcvd432 with aggR, aspU and est was 0.3% for each case of ETEC/EAEC mixed infection. The aspU gene co-existed with aggR, pcvd432, eae and elt in the EAEC/EPEC/ETEC mixed infection. The prevalence of S. boydii, S. dysenteriae, S. flexneriand,S. sonnei was 0.8%, 0.6%, 1.7%, and 0.8%, respectively. No E. coli pathotype and shigella co-infection was detected. In addition, both E. coli pathotypes and Shigella species were resistant to ampicillin, trimethoprim/sulfamethoxazole, streptomycin, chloramphenicol and tetracycline while gentamycin and kanamycin resistance occurred in diarrheagenic E. coli. CONCLUSION E. coli pathotypes and Shigella sero-groups harboring virulent genes are important causes of diarrhea in children in Kenya. The increasing spectrum of antibiotic resistance in diarrheagenic E. coli and Shigella species necessitates the development of antimicrobial stewardship education-programs to influence prescribing behavior as well as optimizing the use of effective antimicrobials in Kenya.
Collapse
Affiliation(s)
- Peter Lokamar Nyanga
- Disease Surveillance and Response Unit, Ministry of Health, Nairobi, Kenya.,Department of Medical Laboratory Sciences, Mount Kenya University, Thika, Kenya.
| | - Jackson Onyuka
- School of Health Sciences, Kirinyaga University, Kirinyaga, Kenya.
| | - Mark Kilongosi Webale
- Department of Medical Laboratory Sciences, Masinde Muliro University of Science and Technology, Kakamega, Kenya.
| | - Tom Were
- Department of Environmental Health Sciences, Technical University of Mombasa, Mombasa, Kenya.
| | - Valentine Budambula
- Department of Medical Laboratory Sciences, Mount Kenya University, Thika, Kenya
| |
Collapse
|
47
|
Abstract
Transition metals are required trace elements for all forms of life. Due to their unique inorganic and redox properties, transition metals serve as cofactors for enzymes and other proteins. In bacterial pathogenesis, the vertebrate host represents a rich source of nutrient metals, and bacteria have evolved diverse metal acquisition strategies. Host metal homeostasis changes dramatically in response to bacterial infections, including production of metal sequestering proteins and the bombardment of bacteria with toxic levels of metals. In response, bacteria have evolved systems to subvert metal sequestration and toxicity. The coevolution of hosts and their bacterial pathogens in the battle for metals has uncovered emerging paradigms in social microbiology, rapid evolution, host specificity, and metal homeostasis across domains. This review focuses on recent advances and open questions in our understanding of the complex role of transition metals at the host-pathogen interface.
Collapse
Affiliation(s)
- Lauren D Palmer
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee 37212;
| | - Eric P Skaar
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee 37212;
- Tennessee Valley Healthcare System, US Department of Veterans Affairs, Nashville, Tennessee 37212
| |
Collapse
|
48
|
Mayneris-Perxachs J, Bolick DT, Leng J, Medlock GL, Kolling GL, Papin JA, Swann JR, Guerrant RL. Protein- and zinc-deficient diets modulate the murine microbiome and metabolic phenotype. Am J Clin Nutr 2016; 104:1253-1262. [PMID: 27733402 PMCID: PMC5081716 DOI: 10.3945/ajcn.116.131797] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 08/29/2016] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Environmental enteropathy, which is linked to undernutrition and chronic infections, affects the physical and mental growth of children in developing areas worldwide. Key to understanding how these factors combine to shape developmental outcomes is to first understand the effects of nutritional deficiencies on the mammalian system including the effect on the gut microbiota. OBJECTIVE We dissected the nutritional components of environmental enteropathy by analyzing the specific metabolic and gut-microbiota changes that occur in weaned-mouse models of zinc or protein deficiency compared with well-nourished controls. DESIGN With the use of a 1H nuclear magnetic resonance spectroscopy-based metabolic profiling approach with matching 16S microbiota analyses, the metabolic consequences and specific effects on the fecal microbiota of protein and zinc deficiency were probed independently in a murine model. RESULTS We showed considerable shifts within the intestinal microbiota 14-24 d postweaning in mice that were maintained on a normal diet (including increases in Proteobacteria and striking decreases in Bacterioidetes). Although the zinc-deficient microbiota were comparable to the age-matched, well-nourished profile, the protein-restricted microbiota remained closer in composition to the weaned enterotype with retention of Bacteroidetes. Striking increases in Verrucomicrobia (predominantly Akkermansia muciniphila) were observed in both well-nourished and protein-deficient mice 14 d postweaning. We showed that protein malnutrition impaired growth and had major metabolic consequences (much more than with zinc deficiency) that included altered energy, polyamine, and purine and pyrimidine metabolism. Consistent with major changes in the gut microbiota, reductions in microbial proteolysis and increases in microbial dietary choline processing were observed. CONCLUSIONS These findings are consistent with metabolic alterations that we previously observed in malnourished children. The results show that we can model the metabolic consequences of malnutrition in the mouse to help dissect relevant pathways involved in the effects of undernutrition and their contribution to environmental enteric dysfunction.
Collapse
Affiliation(s)
- Jordi Mayneris-Perxachs
- Division of Computational and Systems Medicine, Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | | | - Joy Leng
- School of Veterinary Medicine, University of Surrey, Guildford, United Kingdom
| | - Greg L Medlock
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA; and
| | | | - Jason A Papin
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA; and
| | - Jonathan R Swann
- Division of Computational and Systems Medicine, Department of Surgery and Cancer, Imperial College London, London, United Kingdom;
| | | |
Collapse
|
49
|
Abstract
PURPOSE OF REVIEW To highlight recent advances in the understanding of nutritional immunology and in the development of novel therapeutics for inflammatory bowel disease (IBD). RECENT FINDINGS We highlight the variety of factors that contribute to the interaction of the immune system and nutrition including the microbiome and the nervous system stimulation of the gut. We describe the potential for therapeutic development in IBD. Further, we review the cellular metabolic effects on immune activation and promising therapeutic targets. Finally, we show how the progression of understanding the role of lanthionine synthetase C-like 2 has encompassed both nutritional and therapeutic advances and led to the development of novel oral small molecule therapeutics for IBD. SUMMARY Nutritional immunology and drug development research centered around immunoregulatory pathways can provide safer and more effective drugs while accelerating the path to cures.
Collapse
|
50
|
Bartelt LA, Bolick DT, Kolling GL, Roche JK, Zaenker EI, Lara AM, Noronha FJ, Cowardin CA, Moore JH, Turner JR, Warren CA, Buck GA, Guerrant RL. Cryptosporidium Priming Is More Effective than Vaccine for Protection against Cryptosporidiosis in a Murine Protein Malnutrition Model. PLoS Negl Trop Dis 2016; 10:e0004820. [PMID: 27467505 PMCID: PMC4965189 DOI: 10.1371/journal.pntd.0004820] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 06/11/2016] [Indexed: 01/21/2023] Open
Abstract
Cryptosporidium is a major cause of severe diarrhea, especially in malnourished children. Using a murine model of C. parvum oocyst challenge that recapitulates clinical features of severe cryptosporidiosis during malnutrition, we interrogated the effect of protein malnutrition (PM) on primary and secondary responses to C. parvum challenge, and tested the differential ability of mucosal priming strategies to overcome the PM-induced susceptibility. We determined that while PM fundamentally alters systemic and mucosal primary immune responses to Cryptosporidium, priming with C. parvum (106 oocysts) provides robust protective immunity against re-challenge despite ongoing PM. C. parvum priming restores mucosal Th1-type effectors (CD3+CD8+CD103+ T-cells) and cytokines (IFNγ, and IL12p40) that otherwise decrease with ongoing PM. Vaccination strategies with Cryptosporidium antigens expressed in the S. Typhi vector 908htr, however, do not enhance Th1-type responses to C. parvum challenge during PM, even though vaccination strongly boosts immunity in challenged fully nourished hosts. Remote non-specific exposures to the attenuated S. Typhi vector alone or the TLR9 agonist CpG ODN-1668 can partially attenuate C. parvum severity during PM, but neither as effectively as viable C. parvum priming. We conclude that although PM interferes with basal and vaccine-boosted immune responses to C. parvum, sustained reductions in disease severity are possible through mucosal activators of host defenses, and specifically C. parvum priming can elicit impressively robust Th1-type protective immunity despite ongoing protein malnutrition. These findings add insight into potential correlates of Cryptosporidium immunity and future vaccine strategies in malnourished children. Cryptosporidium attributable morbidities in malnourished children are increasingly recognized. Exactly how malnutrition interferes with host mucosal immunity to diarrheal pathogens and mucosal vaccine responses remains unclear. Dissecting these interactions in an experimental model of cryptosporidiosis can uncover new insights into novel therapeutic approaches against a pathogen for which effective therapies and vaccines are currently unavailable. We demonstrate that although malnutrition diminishes baseline (primary) Th1-type mucosal immunity these deficits can be partially overcome via non-specific mucosal strategies (S. Typhi and CpG) and completely restored after a sub-clinical (low-dose) exposure to viable C. parvum. These results add insight into preventive strategies to help alleviate Cryptosporidium-specific diarrhea in children in low-resource settings and abrogate prolonged post-infection sequelae.
Collapse
Affiliation(s)
- Luther A. Bartelt
- Division of Infectious Diseases, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- * E-mail:
| | - David T. Bolick
- Division of Infectious Diseases and Center for Global Health, University of Virginia, Charlottesville, Virginia, United States of America
| | - Glynis L. Kolling
- Division of Infectious Diseases and Center for Global Health, University of Virginia, Charlottesville, Virginia, United States of America
| | - James K. Roche
- Division of Infectious Diseases and Center for Global Health, University of Virginia, Charlottesville, Virginia, United States of America
| | - Edna I. Zaenker
- Division of Infectious Diseases and Center for Global Health, University of Virginia, Charlottesville, Virginia, United States of America
| | - Ana M. Lara
- Molecular Biology and Genetics, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Francisco Jose Noronha
- Division of Infectious Diseases and Center for Global Health, University of Virginia, Charlottesville, Virginia, United States of America
| | - Carrie A. Cowardin
- Division of Infectious Diseases and Center for Global Health, University of Virginia, Charlottesville, Virginia, United States of America
| | - John H. Moore
- Division of Infectious Diseases and Center for Global Health, University of Virginia, Charlottesville, Virginia, United States of America
| | - Jerrold R. Turner
- Department of Pathology, The University of Chicago, Chicago, Illinois, United States of America
- Departments of Pathology and Medicine—Gastroenterology, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
| | - Cirle A. Warren
- Division of Infectious Diseases and Center for Global Health, University of Virginia, Charlottesville, Virginia, United States of America
| | - Gregory A. Buck
- Molecular Biology and Genetics, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Richard L. Guerrant
- Division of Infectious Diseases and Center for Global Health, University of Virginia, Charlottesville, Virginia, United States of America
| |
Collapse
|