1
|
Fenske GJ, Pouzou JG, Pouillot R, Taylor DD, Costard S, Zagmutt FJ. The genomic and epidemiological virulence patterns of Salmonella enterica serovars in the United States. PLoS One 2023; 18:e0294624. [PMID: 38051743 DOI: 10.1371/journal.pone.0294624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 11/06/2023] [Indexed: 12/07/2023] Open
Abstract
The serovars of Salmonella enterica display dramatic differences in pathogenesis and host preferences. We developed a process (patent pending) for grouping Salmonella isolates and serovars by their public health risk. We collated a curated set of 12,337 S. enterica isolate genomes from human, beef, and bovine sources in the US. After annotating a virulence gene catalog for each isolate, we used unsupervised random forest methods to estimate the proximity (similarity) between isolates based upon the genomic presentation of putative virulence traits We then grouped isolates (virulence clusters) using hierarchical clustering (Ward's method), used non-parametric bootstrapping to assess cluster stability, and externally validated the clusters against epidemiological virulence measures from FoodNet, the National Outbreak Reporting System (NORS), and US federal sampling of beef products. We identified five stable virulence clusters of S. enterica serovars. Cluster 1 (higher virulence) serovars yielded an annual incidence rate of domestically acquired sporadic cases roughly one and a half times higher than the other four clusters combined (Clusters 2-5, lower virulence). Compared to other clusters, cluster 1 also had a higher proportion of infections leading to hospitalization and was implicated in more foodborne and beef-associated outbreaks, despite being isolated at a similar frequency from beef products as other clusters. We also identified subpopulations within 11 serovars. Remarkably, we found S. Infantis and S. Typhimurium subpopulations that significantly differed in genome length and clinical case presentation. Further, we found that the presence of the pESI plasmid accounted for the genome length differences between the S. Infantis subpopulations. Our results show that S. enterica strains associated with highest incidence of human infections share a common virulence repertoire. This work could be updated regularly and used in combination with foodborne surveillance information to prioritize serovars of public health concern.
Collapse
Affiliation(s)
- Gavin J Fenske
- EpiX Analytics, Fort Collins, Colorado, United States of America
| | - Jane G Pouzou
- EpiX Analytics, Fort Collins, Colorado, United States of America
| | - Régis Pouillot
- EpiX Analytics, Fort Collins, Colorado, United States of America
| | - Daniel D Taylor
- EpiX Analytics, Fort Collins, Colorado, United States of America
| | - Solenne Costard
- EpiX Analytics, Fort Collins, Colorado, United States of America
| | | |
Collapse
|
2
|
Hanumunthadu B, Kanji N, Owino N, Ferreira Da Silva C, Robinson H, White R, Ferruzzi P, Nakakana U, Canals R, Pollard AJ, Ramasamy M. Salmonella Vaccine Study in Oxford (SALVO) trial: protocol for an observer-participant blind randomised placebo-controlled trial of the iNTS-GMMA vaccine within a European cohort. BMJ Open 2023; 13:e072938. [PMID: 37963701 PMCID: PMC10649500 DOI: 10.1136/bmjopen-2023-072938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 10/03/2023] [Indexed: 11/16/2023] Open
Abstract
INTRODUCTION Invasive non-typhoidal Salmonellosis (iNTS) is mainly caused by Salmonella enterica serovars Typhimurium and Enteritidis and is estimated to result in 77 500 deaths per year, disproportionately affecting children under 5 years of age in sub-Saharan Africa. Invasive non-typhoidal Salmonellae serovars are increasingly acquiring resistance to first-line antibiotics, thus an effective vaccine would be a valuable tool in reducing morbidity and mortality from infection. While NTS livestock vaccines are in wide use, no licensed vaccines exist for use in humans. Here, a first-in-human study of a novel vaccine (iNTS-GMMA) containing S. Typhimurium and S. Enteritidis Generalised Modules for Membrane Antigens (GMMA) outer membrane vesicles is presented. METHOD AND ANALYSIS The Salmonella Vaccine Study in Oxford is a randomised placebo-controlled participant-observer blind phase I study of the iNTS-GMMA vaccine. Healthy adult volunteers will be randomised to receive three intramuscular injections of the iNTS-GMMA vaccine, containing equal quantities of S. Typhimurium and S. Enteritidis GMMA particles adsorbed on Alhydrogel, or an Alhydrogel placebo at 0, 2 and 6 months. Participants will be sequentially enrolled into three groups: group 1, 1:1 randomisation to low dose iNTS-GMMA vaccine or placebo; group 2, 1:1 randomisation to full dose iNTS-GMMA vaccine or placebo; group 3, 2:1 randomisation to full dose or lower dose (dependant on DSMC reviews of groups 1 and 2) iNTS-GMMA vaccine or placebo.The primary objective is safety and tolerability of the vaccine. The secondary objective is immunogenicity as measured by O-antigen based ELISA. Further exploratory objectives will characterise the expanded human immune profile. ETHICS AND DISSEMINATION Ethical approval for this study has been obtained from the South Central-Oxford A Research Ethics Committee (Ethics REF:22/SC/0059). Appropriate documentation and regulatory approvals have been acquired. Results will be disseminated via peer-reviewed articles and conferences. TRIAL REGISTRATION NUMBER EudraCT Number: 2020-000510-14.
Collapse
Affiliation(s)
- Brama Hanumunthadu
- Department of Paediatrics, University of Oxford, Oxford Vaccine Group, Oxford, Oxfordshire, UK
| | - Nasir Kanji
- Department of Paediatrics, University of Oxford, Oxford Vaccine Group, Oxford, Oxfordshire, UK
| | - Nelly Owino
- Department of Paediatrics, University of Oxford, Oxford Vaccine Group, Oxford, Oxfordshire, UK
| | - Carla Ferreira Da Silva
- Department of Paediatrics, University of Oxford, Oxford Vaccine Group, Oxford, Oxfordshire, UK
| | - Hannah Robinson
- Department of Paediatrics, University of Oxford, Oxford Vaccine Group, Oxford, Oxfordshire, UK
- NIHR Oxford Biomedical Research Centre, Oxford, Oxfordshire, UK
| | - Rachel White
- Department of Paediatrics, University of Oxford, Oxford Vaccine Group, Oxford, Oxfordshire, UK
| | | | | | - Rocio Canals
- GSK Vaccines Institute for Global Health, Siena, Italy
| | - Andrew J Pollard
- Department of Paediatrics, University of Oxford, Oxford Vaccine Group, Oxford, Oxfordshire, UK
| | - Maheshi Ramasamy
- Department of Paediatrics, University of Oxford, Oxford Vaccine Group, Oxford, Oxfordshire, UK
| |
Collapse
|
3
|
Grander M, Hoffmann A, Seifert M, Demetz E, Grubwieser P, Pfeifhofer-Obermair C, Haschka D, Weiss G. DMT1 Protects Macrophages from Salmonella Infection by Controlling Cellular Iron Turnover and Lipocalin 2 Expression. Int J Mol Sci 2022; 23:ijms23126789. [PMID: 35743233 PMCID: PMC9223531 DOI: 10.3390/ijms23126789] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 06/10/2022] [Accepted: 06/15/2022] [Indexed: 02/04/2023] Open
Abstract
Macrophages are at the center of innate pathogen control and iron recycling. Divalent metal transporter 1 (DMT1) is essential for the uptake of non-transferrin-bound iron (NTBI) into macrophages and for the transfer of transferrin-bound iron from the endosome to the cytoplasm. As the control of cellular iron trafficking is central for the control of infection with siderophilic pathogens such as Salmonella Typhimurium, a Gram-negative bacterium residing within the phagosome of macrophages, we examined the potential role of DMT1 for infection control. Bone marrow derived macrophages lacking DMT1 (DMT1fl/flLysMCre(+)) present with reduced NTBI uptake and reduced levels of the iron storage protein ferritin, the iron exporter ferroportin and, surprisingly, of the iron uptake protein transferrin receptor. Further, DMT1-deficient macrophages have an impaired control of Salmonella Typhimurium infection, paralleled by reduced levels of the peptide lipocalin-2 (LCN2). LCN2 exerts anti-bacterial activity upon binding of microbial siderophores but also facilitates systemic and cellular hypoferremia. Remarkably, nifedipine, a pharmacological DMT1 activator, stimulates LCN2 expression in RAW264.7 macrophages, confirming its DMT1-dependent regulation. In addition, the absence of DMT1 increases the availability of iron for Salmonella upon infection and leads to increased bacterial proliferation and persistence within macrophages. Accordingly, mice harboring a macrophage-selective DMT1 disruption demonstrate reduced survival following Salmonella infection. This study highlights the importance of DMT1 in nutritional immunity and the significance of iron delivery for the control of infection with siderophilic bacteria.
Collapse
Affiliation(s)
- Manuel Grander
- Department of Internal Medicine II, Medical University of Innsbruck, 6020 Innsbruck, Austria; (M.G.); (A.H.); (M.S.); (E.D.); (P.G.); (C.P.-O.)
- Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Alexander Hoffmann
- Department of Internal Medicine II, Medical University of Innsbruck, 6020 Innsbruck, Austria; (M.G.); (A.H.); (M.S.); (E.D.); (P.G.); (C.P.-O.)
- Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Markus Seifert
- Department of Internal Medicine II, Medical University of Innsbruck, 6020 Innsbruck, Austria; (M.G.); (A.H.); (M.S.); (E.D.); (P.G.); (C.P.-O.)
- Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Egon Demetz
- Department of Internal Medicine II, Medical University of Innsbruck, 6020 Innsbruck, Austria; (M.G.); (A.H.); (M.S.); (E.D.); (P.G.); (C.P.-O.)
| | - Philipp Grubwieser
- Department of Internal Medicine II, Medical University of Innsbruck, 6020 Innsbruck, Austria; (M.G.); (A.H.); (M.S.); (E.D.); (P.G.); (C.P.-O.)
| | - Christa Pfeifhofer-Obermair
- Department of Internal Medicine II, Medical University of Innsbruck, 6020 Innsbruck, Austria; (M.G.); (A.H.); (M.S.); (E.D.); (P.G.); (C.P.-O.)
| | - David Haschka
- Department of Internal Medicine II, Medical University of Innsbruck, 6020 Innsbruck, Austria; (M.G.); (A.H.); (M.S.); (E.D.); (P.G.); (C.P.-O.)
- Correspondence: (D.H.); (G.W.)
| | - Günter Weiss
- Department of Internal Medicine II, Medical University of Innsbruck, 6020 Innsbruck, Austria; (M.G.); (A.H.); (M.S.); (E.D.); (P.G.); (C.P.-O.)
- Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, 6020 Innsbruck, Austria
- Correspondence: (D.H.); (G.W.)
| |
Collapse
|
4
|
Post A, Kaboré B, Berendsen M, Diallo S, Traore O, Arts RJW, Netea MG, Joosten LAB, Tinto H, Jacobs J, de Mast Q, van der Ven A. Altered Ex-Vivo Cytokine Responses in Children With Asymptomatic Plasmodium falciparum Infection in Burkina Faso: An Additional Argument to Treat Asymptomatic Malaria? Front Immunol 2021; 12:614817. [PMID: 34177883 PMCID: PMC8220162 DOI: 10.3389/fimmu.2021.614817] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 05/17/2021] [Indexed: 11/30/2022] Open
Abstract
Introduction Patients with clinical malaria have an increased risk for bacterial bloodstream infections. We hypothesized that asymptomatic malaria parasitemia increases susceptibility for bacterial infections through an effect on the innate immune system. We measured circulating cytokine levels and ex-vivo cytokine production capacity in asymptomatic malaria and compared with controls. Methods Data were collected from asymptomatic participants <5 years old with and without positive malaria microscopy, as well as from hospitalized patients <5 years old with clinical malaria, bacteremia, or malaria/bacteremia co-infections in a malaria endemic region of Burkina Faso. Circulating cytokines (TNF-α, IFN-γ, IL-6, IL-10) were measured using multiplex assays. Whole blood from asymptomatic participants with and without positive malaria microscopy were ex-vivo stimulated with S. aureus, E. coli LPS and Salmonella Typhimurium; cytokine concentrations (TNF-α, IFN-γ, IL-1β, IL-6, IL-10) were measured on supernatants using ELISA. Results Included were children with clinical malaria (n=118), bacteremia (n=22), malaria and bacteremia co-infection (n=9), asymptomatic malaria (n=125), and asymptomatic controls (n=237). Children with either clinical or asymptomatic malaria had higher plasma cytokine concentrations than controls. Cytokine concentrations correlated positively with malaria parasite density with the strongest correlation for IL-10 in both asymptomatic (r=0.63) and clinical malaria (r=0.53). Patients with bacteremia had lower circulating IL-10, TNF-α and IFN-γ and higher IL-6 concentrations, compared to clinical malaria. Ex-vivo whole blood cytokine production to LPS and S. aureus was significantly lower in asymptomatic malaria compared to controls. Whole blood IFN-γ and IL-10 production in response to Salmonella was also lower in asymptomatic malaria. Interpretation In children with asymptomatic malaria, cytokine responses upon ex-vivo bacterial stimulation are downregulated. Further studies are needed to explore if the suggested impaired innate immune response to bacterial pathogens also translates into impaired control of pathogens such as Salmonella spp.
Collapse
Affiliation(s)
- Annelies Post
- Department of Internal Medicine, Radboud Centre for Infectious Diseases, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Berenger Kaboré
- Department of Internal Medicine, Radboud Centre for Infectious Diseases, Radboud University Medical Centre, Nijmegen, Netherlands.,IRSS/Clinical Research Unit of Nanoro (CRUN), Nanoro, Burkina Faso
| | - Mike Berendsen
- Department of Internal Medicine, Radboud Centre for Infectious Diseases, Radboud University Medical Centre, Nijmegen, Netherlands.,Bandim Health Project, Institute of Clinical Research, University of Southern Denmark and Odense University Hospital, Odense, Denmark
| | - Salou Diallo
- IRSS/Clinical Research Unit of Nanoro (CRUN), Nanoro, Burkina Faso
| | - Ousmane Traore
- IRSS/Clinical Research Unit of Nanoro (CRUN), Nanoro, Burkina Faso
| | - Rob J W Arts
- Department of Internal Medicine, Radboud Centre for Infectious Diseases, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Mihai G Netea
- Department of Internal Medicine, Radboud Centre for Infectious Diseases, Radboud University Medical Centre, Nijmegen, Netherlands.,Department for Genomics & Immunoregulation, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| | - Leo A B Joosten
- Department of Internal Medicine, Radboud Centre for Infectious Diseases, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Halidou Tinto
- IRSS/Clinical Research Unit of Nanoro (CRUN), Nanoro, Burkina Faso.,Institut Supérieur des Sciences de la Santé, Université Nazi Boni de Bobo-Dioulasso, Bobo-Dioulasso, Burkina Faso
| | - Jan Jacobs
- Department of Clinical Sciences, Institute of Tropical Medicine, Antwerp, Belgium.,Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Quirijn de Mast
- Department of Internal Medicine, Radboud Centre for Infectious Diseases, Radboud University Medical Centre, Nijmegen, Netherlands
| | - André van der Ven
- Department of Internal Medicine, Radboud Centre for Infectious Diseases, Radboud University Medical Centre, Nijmegen, Netherlands
| |
Collapse
|
5
|
Events associated with susceptibility to invasive Salmonella enterica serovar Typhi in BALB/c mice previously infected with Plasmodium berghei ANKA. Sci Rep 2021; 11:2730. [PMID: 33526848 PMCID: PMC7851127 DOI: 10.1038/s41598-021-82330-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 01/06/2021] [Indexed: 02/02/2023] Open
Abstract
Numerous mechanisms have been proposed to explain why patients with malaria are more susceptible to bloodstream invasions by Salmonella spp., however there are still several unknown critical factors regarding the pathogenesis of coinfection. From a coinfection model, in which an S. enterica serovar Typhi (S_Typhi) was chosen to challenge mice that had been infected 24 h earlier with Plasmodium berghei ANKA (P.b_ANKA), we evaluated the influence of malaria on cytokine levels, the functional activity of femoral bone marrow-derived macrophages and neutrophils, and intestinal permeability. The cytokine profile over eight days of coinfection showed exacerbation in the cytokines MCP-1, IFNγ and TNFα in relation to the increase seen in animals with malaria. The cytokine profile was associated with a considerably reduced neutrophil and macrophage count and a prominent dysfunction, especially in ex vivo neutrophils in coinfected mice, though without bacterial modulation that could influence the invasion capacity of ex vivo S_Typhi obtained from liver macerate in non-phagocyte cells. Finally, irregularities in the integrity of intestinal tissue evidenced ruptures in the enterocyte layer, a presence of mononuclear leukocytes in the enterocyte layer, an increase of goblet cells in the enterocyte layer and a high volume of leukocyte infiltrate in the sub-mucosa were greatly increased in coinfected animals. Increases of mononuclear leukocytes in the enterocyte layer and volume of leukocyte infiltrate in the sub-mucosa were also seen in monoinfected animals with P. berghei ANKA. Our findings suggest malaria causes a disarrangement of intestinal homeostasis, exacerbation of proinflammatory cytokines and dysfunction in neutrophils that render the host susceptible to bacteremia by Salmonella spp.
Collapse
|
6
|
Dos Santos LI, Torres TA, Diniz SQ, Gonçalves R, Caballero-Flores G, Núñez G, Gazzinelli RT, Maloy KJ, Ribeiro do V Antonelli L. Disrupted Iron Metabolism and Mortality during Co-infection with Malaria and an Intestinal Gram-Negative Extracellular Pathogen. Cell Rep 2021; 34:108613. [PMID: 33440153 PMCID: PMC8655499 DOI: 10.1016/j.celrep.2020.108613] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 10/31/2020] [Accepted: 12/16/2020] [Indexed: 11/29/2022] Open
Abstract
Individuals with malaria exhibit increased morbidity and mortality when infected with Gram-negative (Gr−) bacteria. To explore this experimentally, we performed co-infection of mice with Plasmodium chabaudi and Citrobacter rodentium, an extracellular Gr− bacterial pathogen that infects the large intestine. While single infections are controlled effectively, co-infection results in enhanced virulence that is characterized by prolonged systemic bacterial persistence and high mortality. Mortality in co-infected mice is associated with disrupted iron metabolism, elevated levels of plasma heme, and increased mitochondrial reactive oxygen species (ROS) production by phagocytes. In addition, iron acquisition by the bacterium plays a key role in pathogenesis because co-infection with a mutant C. rodentium strain lacking a critical iron acquisition pathway does not cause mortality. These results indicate that disrupted iron metabolism may drive mortality during co-infection with C. rodentium and P. chabaudi by both altering host immune responses and facilitating bacterial persistence. Co-infection with malaria and a Gram-negative bacterial pathogen leads to high mortality Co-infection leads to elevated plasma heme and systemic bacterial persistence Iron acquisition is critical for bacterial persistence and mortality
Collapse
Affiliation(s)
- Luara Isabela Dos Santos
- Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte 30190-009, Minas Gerais, Brazil; Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK
| | - Thais Abdala Torres
- Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte 30190-009, Minas Gerais, Brazil; Instituto de Ciências Biológicas, Departamento de Bioquimica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | - Suelen Queiroz Diniz
- Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte 30190-009, Minas Gerais, Brazil; Instituto de Ciências Biológicas, Departamento de Bioquimica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | - Ricardo Gonçalves
- Departamento de Patologia Geral, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Minhas Gerais, Brazil
| | - Gustavo Caballero-Flores
- Department of Pathology and Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Gabriel Núñez
- Department of Pathology and Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Ricardo Tostes Gazzinelli
- Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte 30190-009, Minas Gerais, Brazil; Instituto de Ciências Biológicas, Departamento de Bioquimica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil; University of Massachusetts Medical School, Worcester, MA 01605-2324, USA
| | - Kevin Joseph Maloy
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK; Institute of Infection, Immunity and Inflammation, University of Glasgow, Sir Graeme Davies Building, 120 University Place, Glasgow G12 8TA, Scotland.
| | | |
Collapse
|
7
|
Iron in immune cell function and host defense. Semin Cell Dev Biol 2020; 115:27-36. [PMID: 33386235 DOI: 10.1016/j.semcdb.2020.12.005] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 12/17/2020] [Accepted: 12/17/2020] [Indexed: 12/13/2022]
Abstract
The control over iron availability is crucial under homeostatic conditions and even more in the case of an infection. This results from diverse properties of iron: first, iron is an important trace element for the host as well as for the pathogen for various cellular and metabolic processes, second, free iron catalyzes Fenton reaction and is therefore producing reactive oxygen species as a part of the host defense machinery, third, iron exhibits important effects on immune cell function and differentiation and fourth almost every immune activation in turn impacts on iron metabolism and spatio-temporal iron distribution. The central importance of iron in the host and microbe interplay and thus for the course of infections led to diverse strategies to restrict iron for invading pathogens. In this review, we focus on how iron restriction to the pathogen is a powerful innate immune defense mechanism of the host called "nutritional immunity". Important proteins in the iron-host-pathogen interplay will be discussed as well as the influence of iron on the efficacy of innate and adaptive immunity. Recently described processes like ferritinophagy and ferroptosis are further covered in respect to their impact on inflammation and infection control and how they impact on our understanding of the interaction of host and pathogen.
Collapse
|
8
|
Alamer E, Carpio VH, Ibitokou SA, Kirtley ML, Phoenix IR, Opata MM, Wilson KD, Cong Y, Dann SM, Chopra AK, Stephens R. Dissemination of non-typhoidal Salmonella during Plasmodium chabaudi infection affects anti-malarial immunity. Parasitol Res 2019; 118:2277-2285. [PMID: 31119381 DOI: 10.1007/s00436-019-06349-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 05/09/2019] [Indexed: 01/18/2023]
Abstract
Malaria-associated bacteremia accounts for up to one-third of deaths from severe malaria, and non-typhoidal Salmonella (NTS) has been reported as a major complication of severe malarial infection. Patients who develop NTS bacteremia during Plasmodium infection show higher mortality rates than individuals with malaria alone. Systemic bacteremia can be caused by a wound or translocation from epithelial or endothelial sites. NTS is an intestinal pathogen, however the contribution of bacterial translocation from the intestinal tract during Plasmodium infection is not well studied. Here, we investigated the integrity of the intestinal barrier function of P. chabaudi-infected mice using large molecules and Salmonella infection. Intestinal histology and the adaptive immune response to malaria were also studied using light microscopy and flow cytometry. P. chabaudi infection compromised intestinal barrier function, which led to increased intestinal cellular infiltration. In addition, we observed increased serum lipopolysaccharide binding protein and leakage of soluble molecules from the intestine into the blood in infected mice. Plasmodium infection also increased intestinal translocation and dissemination of NTS to the liver. The adaptive immune response to P. chabaudi infection was also significantly impacted by NTS translocation. Reduced B and T cell activation were observed in co-infected animals, suggesting interference in the malaria-specific immune responses by bacteremia. These studies demonstrate that P. chabaudi infection induces failure of the barrier function of the intestinal wall and enhanced intestinal bacterial translocation, affecting anti-malarial immunity.
Collapse
Affiliation(s)
- Edrous Alamer
- Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX, 77555, USA
- Department of Medical Laboratories Technology, College of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
| | - Victor H Carpio
- Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX, 77555, USA
| | - Samad A Ibitokou
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, 301 University Blvd., Galveston, TX, 77555, USA
| | - Michelle L Kirtley
- Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX, 77555, USA
| | - Inaia R Phoenix
- Department of Pathology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX, 77555, USA
| | - Michael M Opata
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, 301 University Blvd., Galveston, TX, 77555, USA
| | - Kyle D Wilson
- Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX, 77555, USA
| | - Yingzi Cong
- Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX, 77555, USA
| | - Sara M Dann
- Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX, 77555, USA
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, 301 University Blvd., Galveston, TX, 77555, USA
| | - Ashok K Chopra
- Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX, 77555, USA
| | - Robin Stephens
- Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX, 77555, USA.
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, 301 University Blvd., Galveston, TX, 77555, USA.
| |
Collapse
|
9
|
Lim SH, Methé BA, Knoll BM, Morris A, Obaro SK. Invasive non-typhoidal Salmonella in sickle cell disease in Africa: is increased gut permeability the missing link? J Transl Med 2018; 16:239. [PMID: 30165857 PMCID: PMC6116559 DOI: 10.1186/s12967-018-1622-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 08/25/2018] [Indexed: 02/07/2023] Open
Abstract
Non-typhoidal Salmonella usually induces self-limiting gastroenteritis. However, in many parts of Africa, especially in individuals who are malnourished, infected with malaria, or have sickle cell disease, the organism causes serious and potentially fatal systemic infections. Since the portal of entry of non-typhoidal Salmonella into the systemic circulation is by way of the intestine, we argue that an increased gut permeability plays a vital role in the initiation of invasive non-typhoidal Salmonella in these patients. Here, we will appraise the evidence supporting a breach in the intestinal barrier and propose the mechanisms for the increased risks for invasive non-typhoidal Salmonella infections in these individuals.
Collapse
Affiliation(s)
- Seah H Lim
- Division of Hematology and Oncology, New York Medical College, Hawthorne, NY, USA. .,Westchester Medical Center Cancer Institute, 19 Bradhurst Avenue, Suite 2575S, Hawthorne, NY, 10532, USA.
| | - Barbara A Methé
- Center for Microbiome in Medicine, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Bettina M Knoll
- Division of Infectious Diseases, New York Medical College, Hawthorne, NY, USA
| | - Alison Morris
- Center for Microbiome in Medicine, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Stephen K Obaro
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
10
|
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
|
11
|
Spottiswoode N, Armitage AE, Williams AR, Fyfe AJ, Biswas S, Hodgson SH, Llewellyn D, Choudhary P, Draper SJ, Duffy PE, Drakesmith H. Role of Activins in Hepcidin Regulation during Malaria. Infect Immun 2017; 85:e00191-17. [PMID: 28893916 PMCID: PMC5695100 DOI: 10.1128/iai.00191-17] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 08/23/2017] [Indexed: 12/21/2022] Open
Abstract
Epidemiological observations have linked increased host iron with malaria susceptibility, and perturbed iron handling has been hypothesized to contribute to the potentially life-threatening anemia that may accompany blood-stage malaria infection. To improve our understanding of these relationships, we examined the pathways involved in regulation of the master controller of iron metabolism, the hormone hepcidin, in malaria infection. We show that hepcidin upregulation in Plasmodium berghei murine malaria infection was accompanied by changes in expression of bone morphogenetic protein (BMP)/sons of mothers against decapentaplegic (SMAD) pathway target genes, a key pathway involved in hepcidin regulation. We therefore investigated known agonists of the BMP/SMAD pathway and found that Bmp gene expression was not increased in infection. In contrast, activin B, which can signal through the BMP/SMAD pathway and has been associated with increased hepcidin during inflammation, was upregulated in the livers of Plasmodium berghei-infected mice; hepatic activin B was also upregulated at peak parasitemia during infection with Plasmodium chabaudi Concentrations of the closely related protein activin A increased in parallel with hepcidin in serum from malaria-naive volunteers infected in controlled human malaria infection (CHMI) clinical trials. However, antibody-mediated neutralization of activin activity during murine malaria infection did not affect hepcidin expression, suggesting that these proteins do not stimulate hepcidin upregulation directly. In conclusion, we present evidence that the BMP/SMAD signaling pathway is perturbed in malaria infection but that activins, although raised in malaria infection, may not have a critical role in hepcidin upregulation in this setting.
Collapse
Affiliation(s)
- Natasha Spottiswoode
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
- Laboratory of Malaria Immunology & Vaccinology, NIAID, NIH, Bethesda, Maryland, USA
| | - Andrew E Armitage
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Andrew R Williams
- Department of Veterinary Disease Biology, University of Copenhagen, Frederiksberg, Denmark
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Alex J Fyfe
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Sumi Biswas
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | - David Llewellyn
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | - Simon J Draper
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Patrick E Duffy
- Laboratory of Malaria Immunology & Vaccinology, NIAID, NIH, Bethesda, Maryland, USA
| | - Hal Drakesmith
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| |
Collapse
|
12
|
Jonker FAM, Te Poel E, Bates I, Boele van Hensbroek M. Anaemia, iron deficiency and susceptibility to infection in children in sub-Saharan Africa, guideline dilemmas. Br J Haematol 2017; 177:878-883. [PMID: 28397964 DOI: 10.1111/bjh.14593] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 01/15/2017] [Accepted: 01/15/2017] [Indexed: 12/31/2022]
Abstract
Globally, anaemia, iron deficiency and infections are responsible for a majority of the morbidity and mortality that occurs among children. As iron is essential for erythropoiesis and the human immune system, as well as a crucial element for many pathogens, these three conditions often interact. This article considers the question - have the studies conducted so far unravelled the potential complex interaction between these factors sufficiently enough to be able to develop universally applicable guidelines about iron treatment in children? It is possible, however, that the area is too complex and diverse, with many sub-populations, and that not universal, but tailor-made guidelines are needed based on some agreed principles.
Collapse
Affiliation(s)
- Femkje A M Jonker
- Global Child Health Group, Emma Children's Hospital, Academic Medical Centre, Amsterdam, The Netherlands
| | - Elodie Te Poel
- Global Child Health Group, Emma Children's Hospital, Academic Medical Centre, Amsterdam, The Netherlands
| | - Imelda Bates
- Liverpool School of Tropical Medicine, University of Liverpool, Liverpool, United Kingdom
| | | |
Collapse
|
13
|
Nguyen TKP, Tran TH, Roberts CL, Graham SM, Marais BJ. Child pneumonia - focus on the Western Pacific Region. Paediatr Respir Rev 2017; 21:102-110. [PMID: 27569107 PMCID: PMC7106312 DOI: 10.1016/j.prrv.2016.07.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 07/12/2016] [Indexed: 01/09/2023]
Abstract
Worldwide, pneumonia is the leading cause of death in infants and young children (aged <5 years). We provide an overview of the global pneumonia disease burden, as well as the aetiology and management practices in different parts of the world, with a specific focus on the WHO Western Pacific Region. In 2011, the Western Pacific region had an estimated 0.11 pneumonia episodes per child-year with 61,900 pneumonia-related deaths in children less than 5 years of age. The majority (>75%) of pneumonia deaths occurred in six countries; Cambodia, China, Laos, Papua New Guinea, the Philippines and Viet Nam. Historically Streptococcus pneumoniae and Haemophilus influenzae were the commonest causes of severe pneumonia and pneumonia-related deaths in young children, but this is changing with the introduction of highly effective conjugate vaccines and socio-economic development. The relative contribution of viruses and atypical bacteria appear to be increasing and traditional case management approaches may require revision to accommodate increased uptake of conjugated vaccines in the Western Pacific region. Careful consideration should be given to risk reduction strategies, enhanced vaccination coverage, improved management of hypoxaemia and antibiotic stewardship.
Collapse
MESH Headings
- Anti-Bacterial Agents/therapeutic use
- Asia, Southeastern/epidemiology
- Child
- Child, Preschool
- Asia, Eastern/epidemiology
- Global Health
- Haemophilus Infections/drug therapy
- Haemophilus Infections/epidemiology
- Haemophilus Infections/mortality
- Haemophilus Infections/prevention & control
- Haemophilus Vaccines/therapeutic use
- Haemophilus influenzae
- Humans
- Hypoxia/therapy
- Infant
- Influenza Vaccines/therapeutic use
- Influenza, Human/epidemiology
- Influenza, Human/mortality
- Influenza, Human/prevention & control
- Influenza, Human/therapy
- Pneumococcal Vaccines/therapeutic use
- Pneumonia/drug therapy
- Pneumonia/epidemiology
- Pneumonia/mortality
- Pneumonia/prevention & control
- Pneumonia, Mycoplasma/drug therapy
- Pneumonia, Mycoplasma/epidemiology
- Pneumonia, Mycoplasma/mortality
- Pneumonia, Pneumococcal/drug therapy
- Pneumonia, Pneumococcal/epidemiology
- Pneumonia, Pneumococcal/mortality
- Pneumonia, Pneumococcal/prevention & control
- Respiratory Syncytial Virus Infections/epidemiology
- Respiratory Syncytial Virus Infections/mortality
- Respiratory Syncytial Virus Infections/therapy
- Streptococcus pneumoniae
- Tuberculosis, Pulmonary/drug therapy
- Tuberculosis, Pulmonary/epidemiology
- Tuberculosis, Pulmonary/mortality
- World Health Organization
Collapse
Affiliation(s)
- T K P Nguyen
- Discipline of Paediatrics and Adolescent Medicine, The Children's Hospital at Westmead, The University of Sydney, Australia; Da Nang Hospital for Women and Children, Da Nang, Viet Nam.
| | - T H Tran
- Da Nang Hospital for Women and Children, Da Nang, Viet Nam
| | - C L Roberts
- Clinical and Population Perinatal Health Research, Kolling Institute, Northern Sydney Local Health District, Sydney, Australia; Sydney Medical School Northern, The University of Sydney, Australia
| | - S M Graham
- Centre for International Child Health, University of Melbourne and Murdoch Children's Research Institute, Australia
| | - B J Marais
- Discipline of Paediatrics and Adolescent Medicine, The Children's Hospital at Westmead, The University of Sydney, Australia
| |
Collapse
|
14
|
Krumkamp R, Kreuels B, Sarpong N, Boahen KG, Foli G, Hogan B, Jaeger A, Reigl L, Zeeb H, Marks F, Adu-Sarkodie Y, May J. Association Between Malaria and Invasive NontyphoidalSalmonellaInfection in a Hospital Study: Accounting for Berkson's Bias. Clin Infect Dis 2016; 62 Suppl 1:S83-9. [DOI: 10.1093/cid/civ950] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
15
|
Orf K, Cunnington AJ. Infection-related hemolysis and susceptibility to Gram-negative bacterial co-infection. Front Microbiol 2015; 6:666. [PMID: 26175727 PMCID: PMC4485309 DOI: 10.3389/fmicb.2015.00666] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 06/18/2015] [Indexed: 01/05/2023] Open
Abstract
Increased susceptibility to co-infection with enteric Gram-negative bacteria, particularly non-typhoidal Salmonella, is reported in malaria and Oroya fever (Bartonella bacilliformis infection), and can lead to increased mortality. Accumulating epidemiological evidence indicates a causal association with risk of bacterial co-infection, rather than just co-incidence of common risk factors. Both malaria and Oroya fever are characterized by hemolysis, and observations in humans and animal models suggest that hemolysis causes the susceptibility to bacterial co-infection. Evidence from animal models implicates hemolysis in the impairment of a variety of host defense mechanisms, including macrophage dysfunction, neutrophil dysfunction, and impairment of adaptive immune responses. One mechanism supported by evidence from animal models and human data, is the induction of heme oxygenase-1 in bone marrow, which impairs the ability of developing neutrophils to mount a competent oxidative burst. As a result, dysfunctional neutrophils become a new niche for replication of intracellular bacteria. Here we critically appraise and summarize the key evidence for mechanisms which may contribute to these very specific combinations of co-infections, and propose interventions to ameliorate this risk.
Collapse
Affiliation(s)
- Katharine Orf
- Section of Paediatrics, Imperial College London London, UK
| | | |
Collapse
|
16
|
Yang J, Barrila J, Roland KL, Kilbourne J, Ott CM, Forsyth RJ, Nickerson CA. Characterization of the Invasive, Multidrug Resistant Non-typhoidal Salmonella Strain D23580 in a Murine Model of Infection. PLoS Negl Trop Dis 2015; 9:e0003839. [PMID: 26091096 PMCID: PMC4474555 DOI: 10.1371/journal.pntd.0003839] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 05/18/2015] [Indexed: 01/09/2023] Open
Abstract
A distinct pathovar of Salmonella enterica serovar Typhimurium, ST313, has emerged in sub-Saharan Africa as a major cause of fatal bacteremia in young children and HIV-infected adults. D23580, a multidrug resistant clinical isolate of ST313, was previously shown to have undergone genome reduction in a manner that resembles that of the more human-restricted pathogen, Salmonella enterica serovar Typhi. It has since been shown through tissue distribution studies that D23580 is able to establish an invasive infection in chickens. However, it remains unclear whether ST313 can cause lethal disease in a non-human host following a natural course of infection. Herein we report that D23580 causes lethal and invasive disease in a murine model of infection following peroral challenge. The LD50 of D23580 in female BALB/c mice was 4.7 x 105 CFU. Tissue distribution studies performed 3 and 5 days post-infection confirmed that D23580 was able to more rapidly colonize the spleen, mesenteric lymph nodes and gall bladder in mice when compared to the well-characterized S. Typhimurium strain SL1344. D23580 exhibited enhanced resistance to acid stress relative to SL1344, which may lend towards increased capability to survive passage through the gastrointestinal tract as well as during its intracellular lifecycle. Interestingly, D23580 also displayed higher swimming motility relative to SL1344, S. Typhi strain Ty2, and the ST313 strain A130. Biochemical tests revealed that D23580 shares many similar metabolic features with SL1344, with several notable differences in the Voges-Proskauer and catalase tests, as well alterations in melibiose, and inositol utilization. These results represent the first full duration infection study using an ST313 strain following the entire natural course of disease progression, and serve as a benchmark for ongoing and future studies into the pathogenesis of D23580. A deadly form of non-typhoidal Salmonella has emerged as a major cause of invasive disease in sub-Saharan Africa. Initial genomic profiling of this novel Salmonella sequence type, ST313, indicated that although it is technically classified as S. Typhimurium (a serovar characterized by a broad host range), it may be evolving towards becoming a more human-specific, ‘typhoid-like’ pathogen. However, it was recently demonstrated that ST313 strains were indeed able to establish an invasive and damaging infection in chickens. Despite these important findings, it remains unclear whether ST313 is able to cause lethal disease in a non-human host, since no study has yet followed the entire natural course of disease progression. As such, there are no data available concerning the median lethal dose (LD50) of any ST313 strain. This is an important metric, as the LD50 value will serve as a benchmark for mechanistic studies focused on understanding the relationship between virulence and the phenotypic and molecular genetic attributes associated with ST313 infections. Here we report that D23580 causes lethal disease in BALB/c mice and determined the LD50 following peroral challenge. Phenotypic characterization revealed distinct differences in tissue distribution, acid stress resistance, and biochemical utilization between D23580 and the ‘classic’ Typhimurium strain SL1344.
Collapse
Affiliation(s)
- Jiseon Yang
- Center for Infectious Diseases and Vaccinology, The Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| | - Jennifer Barrila
- Center for Infectious Diseases and Vaccinology, The Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| | - Kenneth L. Roland
- Center for Infectious Diseases and Vaccinology, The Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| | - Jacquelyn Kilbourne
- Center for Infectious Diseases and Vaccinology, The Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| | - C. Mark Ott
- Biomedical Research and Environmental Sciences Division, NASA Johnson Space Center, Houston, Texas, United States of America
| | - Rebecca J. Forsyth
- Center for Infectious Diseases and Vaccinology, The Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| | - Cheryl A. Nickerson
- Center for Infectious Diseases and Vaccinology, The Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
- School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
- * E-mail:
| |
Collapse
|
17
|
Baumgartner J, Barth-Jaeggi T. Iron interventions in children from low-income and middle-income populations: benefits and risks. Curr Opin Clin Nutr Metab Care 2015; 18:289-94. [PMID: 25807351 DOI: 10.1097/mco.0000000000000168] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
PURPOSE OF REVIEW Children from low- and middle-income countries are particularly vulnerable to develop iron deficiency and iron deficiency anaemia (IDA), which can be prevented or controlled with different iron intervention strategies. However, there is a debate on the efficacy and safety of iron interventions, especially in children from areas with a high infectious disease burden. This review provides an overview of recent trials that investigated the benefits and potential risks of iron interventions in children from low and middle-income countries. RECENT FINDINGS Recent studies showed that intermittent iron supplementation is a promising strategy in reducing iron deficiency and IDA. Only a few studies investigated the effect of iron interventions on developmental outcomes, such as growth and cognition, and provided mixed results. An increasing number of studies reported that iron intervention increases morbidity and causes unfavourable shifts in the gut microbial composition along with increases in intestinal inflammation, particularly in children with a high infectious disease burden. SUMMARY More studies in children from low and middle-income populations are needed that provide evidence for the beneficial effects of iron interventions on functional outcomes beyond alleviating iron deficiency and IDA, and that explore potential mechanisms underlying the negative effects of iron reported in recent trials.
Collapse
Affiliation(s)
- Jeannine Baumgartner
- aCentre of Excellence for Nutrition, North-West University, Potchefstroom, South Africa bLaboratory of Human Nutrition, Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| | | |
Collapse
|
18
|
Faure E. Malarial pathocoenosis: beneficial and deleterious interactions between malaria and other human diseases. Front Physiol 2014; 5:441. [PMID: 25484866 PMCID: PMC4240042 DOI: 10.3389/fphys.2014.00441] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 10/28/2014] [Indexed: 11/28/2022] Open
Abstract
In nature, organisms are commonly infected by an assemblage of different parasite species or by genetically distinct parasite strains that interact in complex ways. Linked to co-infections, pathocoenosis, a term proposed by M. Grmek in 1969, refers to a pathological state arising from the interactions of diseases within a population and to the temporal and spatial dynamics of all of the diseases. In the long run, malaria was certainly one of the most important component of past pathocoenoses. Today this disease, which affects hundreds of millions of individuals and results in approximately one million deaths each year, is always highly endemic in over 20% of the world and is thus co-endemic with many other diseases. Therefore, the incidences of co-infections and possible direct and indirect interactions with Plasmodium parasites are very high. Both positive and negative interactions between malaria and other diseases caused by parasites belonging to numerous taxa have been described and in some cases, malaria may modify the process of another disease without being affected itself. Interactions include those observed during voluntary malarial infections intended to cure neuro-syphilis or during the enhanced activations of bacterial gastro-intestinal diseases and HIV infections. Complex relationships with multiple effects should also be considered, such as those observed during helminth infections. Moreover, reports dating back over 2000 years suggested that co- and multiple infections have generally deleterious consequences and analyses of historical texts indicated that malaria might exacerbate both plague and cholera, among other diseases. Possible biases affecting the research of etiological agents caused by the protean manifestations of malaria are discussed. A better understanding of the manner by which pathogens, particularly Plasmodium, modulate immune responses is particularly important for the diagnosis, cure, and control of diseases in human populations.
Collapse
Affiliation(s)
- Eric Faure
- Aix-Marseille Université, Centre National de la Recherche Scientifique, Centrale Marseille, I2M, UMR 7373Marseille, France
| |
Collapse
|
19
|
Anaemia, iron deficiency and susceptibility to infections. J Infect 2014; 69 Suppl 1:S23-7. [DOI: 10.1016/j.jinf.2014.08.007] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/20/2014] [Indexed: 12/11/2022]
|
20
|
Kortman GAM, Raffatellu M, Swinkels DW, Tjalsma H. Nutritional iron turned inside out: intestinal stress from a gut microbial perspective. FEMS Microbiol Rev 2014; 38:1202-34. [PMID: 25205464 DOI: 10.1111/1574-6976.12086] [Citation(s) in RCA: 184] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 08/27/2014] [Accepted: 08/29/2014] [Indexed: 12/16/2022] Open
Abstract
Iron is abundantly present on earth, essential for most microorganisms and crucial for human health. Human iron deficiency that is nevertheless highly prevalent in developing regions of the world can be effectively treated by oral iron administration. Accumulating evidence indicates that excess of unabsorbed iron that enters the colonic lumen causes unwanted side effects at the intestinal host-microbiota interface. The chemical properties of iron, the luminal environment and host iron withdrawal mechanisms, especially during inflammation, can turn the intestine in a rather stressful milieu. Certain pathogenic enteric bacteria can, however, deal with this stress at the expense of other members of the gut microbiota, while their virulence also seems to be stimulated in an iron-rich intestinal environment. This review covers the multifaceted aspects of nutritional iron stress with respect to growth, composition, metabolism and pathogenicity of the gut microbiota in relation to human health. We aim to present an unpreceded view on the dynamic effects and impact of oral iron administration on intestinal host-microbiota interactions to provide leads for future research and other applications.
Collapse
Affiliation(s)
- Guus A M Kortman
- Department of Laboratory Medicine, The Radboud Institute for Molecular Life Sciences (RIMLS) of the Radboud University Medical Center, Nijmegen, The Netherlands
| | | | | | | |
Collapse
|
21
|
Spottiswoode N, Duffy PE, Drakesmith H. Iron, anemia and hepcidin in malaria. Front Pharmacol 2014; 5:125. [PMID: 24910614 PMCID: PMC4039013 DOI: 10.3389/fphar.2014.00125] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Accepted: 05/11/2014] [Indexed: 12/21/2022] Open
Abstract
Malaria and iron have a complex but important relationship. Plasmodium proliferation requires iron, both during the clinically silent liver stage of growth and in the disease-associated phase of erythrocyte infection. Precisely how the protozoan acquires its iron from its mammalian host remains unclear, but iron chelators can inhibit pathogen growth in vitro and in animal models. In humans, iron deficiency appears to protect against severe malaria, while iron supplementation increases risks of infection and disease. Malaria itself causes profound disturbances in physiological iron distribution and utilization, through mechanisms that include hemolysis, release of heme, dyserythropoiesis, anemia, deposition of iron in macrophages, and inhibition of dietary iron absorption. These effects have significant consequences. Malarial anemia is a major global health problem, especially in children, that remains incompletely understood and is not straightforward to treat. Furthermore, the changes in iron metabolism during a malaria infection may modulate susceptibility to co-infections. The release of heme and accumulation of iron in granulocytes may explain increased vulnerability to non-typhoidal Salmonella during malaria. The redistribution of iron away from hepatocytes and into macrophages may confer host resistance to superinfection, whereby blood-stage parasitemia prevents the development of a second liver-stage Plasmodium infection in the same organism. Key to understanding the pathophysiology of iron metabolism in malaria is the activity of the iron regulatory hormone hepcidin. Hepcidin is upregulated during blood-stage parasitemia and likely mediates much of the iron redistribution that accompanies disease. Understanding the regulation and role of hepcidin may offer new opportunities to combat malaria and formulate better approaches to treat anemia in the developing world.
Collapse
Affiliation(s)
- Natasha Spottiswoode
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of HealthBethesda, MD, USA
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of OxfordOxford, UK
| | - Patrick E. Duffy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of HealthBethesda, MD, USA
| | - Hal Drakesmith
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of OxfordOxford, UK
| |
Collapse
|
22
|
Gonçalves RM, Lima NF, Ferreira MU. Parasite virulence, co-infections and cytokine balance in malaria. Pathog Glob Health 2014; 108:173-8. [PMID: 24854175 DOI: 10.1179/2047773214y.0000000139] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Strong early inflammatory responses followed by a timely production of regulatory cytokines are required to control malaria parasite multiplication without inducing major host pathology. Here, we briefly examine the homeostasis of inflammatory responses to malaria parasite species with varying virulence levels and discuss how co-infections with bacteria, viruses, and helminths can modulate inflammation, either aggravating or alleviating malaria-related morbidity.
Collapse
|
23
|
Phoba MF, De Boeck H, Ifeka BB, Dawili J, Lunguya O, Vanhoof R, Muyembe JJ, Van Geet C, Bertrand S, Jacobs J. Epidemic increase in Salmonella bloodstream infection in children, Bwamanda, the Democratic Republic of Congo. Eur J Clin Microbiol Infect Dis 2013; 33:79-87. [PMID: 23975545 DOI: 10.1007/s10096-013-1931-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Accepted: 07/10/2013] [Indexed: 10/26/2022]
Abstract
Salmonella enterica is the leading cause of bloodstream infection in children in sub-Saharan Africa, but few data are available from Central-Africa. We documented during the period November 2011 to May 2012 an epidemic increase in invasive Salmonella bloodstream infections in HGR Bwamanda, a referral hospital in Equateur Province, DR Congo. Salmonella spp. represented 90.4 % (103 out of 114) of clinically significant blood culture isolates and comprised Salmonella Typhimurium (54.4 %, 56 out of 103), Salmonella Enteritidis (28.2 %, 29 out of 103) and Salmonella Typhi (17.5 %, 18 out of 103), with Salmonella Enteritidis accounting for most of the increase. Most (82 out of 103, 79.6 %) isolates were obtained from children < 5 years old. Median ages of patients infected with Salmonella Typhimurium and Salmonella Enteritidis were 14 months (14 days to 64 years) and 19 months (3 months to 8 years) respectively. Clinical presentation was non-specific; the in-hospital case fatality rate was 11.1 %. More than two thirds (69.7 %, 53 out of 76) of children < 5 years for whom laboratory data were available had Plasmodium falciparum infection. Most (83/85, 97.6 %) non-typhoid Salmonella isolates as well as 6/18 (33.3 %) Salmonella Typhi isolates were multidrug resistant (i.e. resistant to the first-line oral antibiotics amoxicillin, trimethoprim-sulfamethoxazole and chloramphenicol), one (1.0 %) Salmonella Typhimurium had decreased ciprofloxacin susceptibility owing to a point mutation in the gyrA gene (Gly81Cys). Multilocus variable-number tandem-repeat (MLVA) analysis of the Salmonella Enteritidis isolates revealed closely related patterns comprising three major and four minor profiles, with differences limited to one out of five loci. These data show an epidemic increase in clonally related multidrug-resistant Salmonella bloodstream infection in children in DR Congo.
Collapse
Affiliation(s)
- M-F Phoba
- Department of Clinical Microbiology, National Institute for Biomedical Research, Kinshasa, The Democratic Republic of Congo
| | | | | | | | | | | | | | | | | | | |
Collapse
|