1
|
Vassilopoulou E, Agostoni C, Feketea G, Alberti I, Gianni ML, Milani GP. The Role of Breastfeeding in Acute Respiratory Infections in Infancy. Pediatr Infect Dis J 2024; 43:1090-1099. [PMID: 38986006 DOI: 10.1097/inf.0000000000004454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/12/2024]
Abstract
BACKGROUND Acute respiratory infections (ARIs) affect the respiratory tract, are often caused by viruses such as respiratory syncytial virus and rhinovirus, and present symptoms such as coughing, fever, respiratory distress, and breathing difficulty. The global adherence to exclusive breastfeeding (BF) for the first 6 months of life has reached 44%, supported by the World Health Organization and United Nations International Children's Emergency Fund efforts. BF provides vital nutrients and contributes to infant immune system development, protecting against infections. The role of BF in preventing and reducing complications of ARIs in infants is gaining attention, prompting a review of current data and future research needs. This review aims to summarize the evidence on the role of BF in reducing the risk and severity of ARIs in infants, elucidate the adaptations in breast milk composition during infections, and identify relevant research needs. METHODS AND RESULTS Human milk (HM) is rich in immunoglobulins, antimicrobial peptides, and immunomodulatory factors that protect against various pathogens, including respiratory viruses. Several studies have demonstrated that BF is associated with a significant reduction in hospitalization, oxygen requirements, and mortality in infants with ARIs. The effectiveness of BF varies according to the specific respiratory virus, and a longer duration of exclusive BF appears to enhance its protective effect. It is documented that the composition of HM adjusts dynamically in response to infections, fortifying the infant's immune defenses. Specific immunological components of HM, including leukocytes and immunoglobulins, increase in response to infection in the infant, contributing to the enhancement of the immune defense in infants. Immune-boosting microRNAs enhance immune transfer to the infants and promote early gut maturation, and the HM microbiome along with other factors modifies the infant's gut microbiome and immune system. CONCLUSIONS BF defends infants from respiratory infections, and the investigation of the microRNAs in HM offers new insights into its antiviral properties. The promotion of BF, especially in vulnerable communities, is of paramount importance in alleviating the global burden of ARIs in infancy.
Collapse
Affiliation(s)
- Emilia Vassilopoulou
- From the Pediatric Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Nutritional Sciences and Dietetics, International Hellenic University, Thessaloniki, Greece
| | - Carlo Agostoni
- From the Pediatric Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Gavriela Feketea
- Department of Pharmacology, Toxicology and Clinical Pharmacology, University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Pediatric Allergy Outpatient Clinic, Department of Pediatrics, "Karamandaneio" Children's Hospital of Patra, Patras, Greece
| | - Ilaria Alberti
- From the Pediatric Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Maria Lorella Gianni
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
- Fondazione I.R.C.C.S. Ca' Granda Ospedale Maggiore Policlinico, Neonatal Intensive Care Unit, Milan, Italy
| | - Gregorio Paolo Milani
- From the Pediatric Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
2
|
Baumgartner M, Lang M, Holley H, Crepaz D, Hausmann B, Pjevac P, Moser D, Haller F, Hof F, Beer A, Orgler E, Frick A, Khare V, Evstatiev R, Strohmaier S, Primas C, Dolak W, Köcher T, Klavins K, Rath T, Neurath MF, Berry D, Makristathis A, Muttenthaler M, Gasche C. Mucosal Biofilms Are an Endoscopic Feature of Irritable Bowel Syndrome and Ulcerative Colitis. Gastroenterology 2021; 161:1245-1256.e20. [PMID: 34146566 PMCID: PMC8527885 DOI: 10.1053/j.gastro.2021.06.024] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 06/11/2021] [Accepted: 06/13/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS Irritable bowel syndrome (IBS) and inflammatory bowel diseases result in a substantial reduction in quality of life and a considerable socioeconomic impact. In IBS, diagnosis and treatment options are limited, but evidence for involvement of the gut microbiome in disease pathophysiology is emerging. Here we analyzed the prevalence of endoscopically visible mucosal biofilms in gastrointestinal disease and associated changes in microbiome composition and metabolism. METHODS The presence of mucosal biofilms was assessed in 1426 patients at 2 European university-based endoscopy centers. One-hundred and seventeen patients were selected for in-depth molecular and microscopic analysis using 16S ribosomal RNA gene amplicon-sequencing of colonic biopsies and fecal samples, confocal microscopy with deep learning-based image analysis, scanning electron microscopy, metabolomics, and in vitro biofilm formation assays. RESULTS Biofilms were present in 57% of patients with IBS and 34% of patients with ulcerative colitis compared with 6% of controls (P < .001). These yellow-green adherent layers of the ileum and right-sided colon were microscopically confirmed to be dense bacterial biofilms. 16S-sequencing links the presence of biofilms to a dysbiotic gut microbiome, including overgrowth of Escherichia coli and Ruminococcus gnavus. R. gnavus isolates cultivated from patient biofilms also formed biofilms in vitro. Metabolomic analysis found an accumulation of bile acids within biofilms that correlated with fecal bile acid excretion, linking this phenotype with a mechanism of diarrhea. CONCLUSIONS The presence of mucosal biofilms is an endoscopic feature in a subgroup of IBS and ulcerative colitis with disrupted bile acid metabolism and bacterial dysbiosis. They provide novel insight into the pathophysiology of IBS and ulcerative colitis, illustrating that biofilm can be seen as a tipping point in the development of dysbiosis and disease.
Collapse
Affiliation(s)
- Maximilian Baumgartner
- Division of Gastroenterology and Hepatology, Department of Internal Medicine 3, Medical University of Vienna, Vienna, Austria
| | - Michaela Lang
- Division of Gastroenterology and Hepatology, Department of Internal Medicine 3, Medical University of Vienna, Vienna, Austria; Centre for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystem Science, Division of Microbial Ecology, University of Vienna, Vienna, Austria
| | - Hunter Holley
- Division of Gastroenterology and Hepatology, Department of Internal Medicine 3, Medical University of Vienna, Vienna, Austria; Centre for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystem Science, Division of Microbial Ecology, University of Vienna, Vienna, Austria
| | - Daniel Crepaz
- Centre for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystem Science, Division of Microbial Ecology, University of Vienna, Vienna, Austria
| | - Bela Hausmann
- Joint Microbiome Facility of the Medical University of Vienna and the University of Vienna, Vienna, Austria; Division of Microbiology, Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Petra Pjevac
- Centre for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystem Science, Division of Microbial Ecology, University of Vienna, Vienna, Austria; Joint Microbiome Facility of the Medical University of Vienna and the University of Vienna, Vienna, Austria
| | - Doris Moser
- Department of Cranio-Maxillofacial and Oral Surgery, Medical University of Vienna, Vienna, Austria
| | - Felix Haller
- Division of Gastroenterology and Hepatology, Department of Internal Medicine 3, Medical University of Vienna, Vienna, Austria
| | - Fabian Hof
- Division of Gastroenterology and Hepatology, Department of Internal Medicine 3, Medical University of Vienna, Vienna, Austria
| | - Andrea Beer
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Elisabeth Orgler
- Division of Gastroenterology and Hepatology, Department of Internal Medicine 3, Medical University of Vienna, Vienna, Austria
| | - Adrian Frick
- Division of Gastroenterology and Hepatology, Department of Internal Medicine 3, Medical University of Vienna, Vienna, Austria
| | - Vineeta Khare
- Division of Gastroenterology and Hepatology, Department of Internal Medicine 3, Medical University of Vienna, Vienna, Austria
| | - Rayko Evstatiev
- Division of Gastroenterology and Hepatology, Department of Internal Medicine 3, Medical University of Vienna, Vienna, Austria
| | - Susanne Strohmaier
- Center for Public Health, Department of Epidemiology, Medical University of Vienna, Vienna, Austria
| | - Christian Primas
- Division of Gastroenterology and Hepatology, Department of Internal Medicine 3, Medical University of Vienna, Vienna, Austria
| | - Werner Dolak
- Division of Gastroenterology and Hepatology, Department of Internal Medicine 3, Medical University of Vienna, Vienna, Austria
| | | | - Kristaps Klavins
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Timo Rath
- Ludwig Demling Endoscopy Center of Excellence, Division of Gastroenterology, Friedrich-Alexander-University, Erlangen, Germany
| | - Markus F Neurath
- Ludwig Demling Endoscopy Center of Excellence, Division of Gastroenterology, Friedrich-Alexander-University, Erlangen, Germany
| | - David Berry
- Centre for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystem Science, Division of Microbial Ecology, University of Vienna, Vienna, Austria; Joint Microbiome Facility of the Medical University of Vienna and the University of Vienna, Vienna, Austria
| | - Athanasios Makristathis
- Joint Microbiome Facility of the Medical University of Vienna and the University of Vienna, Vienna, Austria; Division of Microbiology, Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Markus Muttenthaler
- Faculty of Chemistry, Institute of Biological Chemistry, University of Vienna, Vienna, Austria; Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Christoph Gasche
- Division of Gastroenterology and Hepatology, Department of Internal Medicine 3, Medical University of Vienna, Vienna, Austria; Loha for Life, Center for Gastroenterlogy and Iron Deficiency, Vienna, Austria.
| |
Collapse
|
3
|
Thuthikkadu Indhuprakash S, Karthikeyan M, Gopal G, Ambi SV, Sekaran S, Palaniappan B, Diraviyam T. Antibody therapy against antibiotic-resistant diarrheagenic Escherichia coli: a systematic review. Immunotherapy 2021; 13:1305-1320. [PMID: 34463122 DOI: 10.2217/imt-2021-0079] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Over four billion episodes of diarrhea occur annually in developing countries with diarrheagenic Escherichia coli (DEC) outbreaks also being reported, until now bacterial diarrhea is conventionally addressed by the antibiotic treatment regimes. In recent decades, the emergence of antimicrobial-resistant strains has become a major obstacle in diarrheal treatment; hence, novel and ideal therapeutics are needed. Notably, 80% of DEC is resistant to first-class antibiotics. Among the existing strategies, passive immunization is considered as an alternative to combat drug-resistant bacteria. Antibodies specific to an antigen can be used for prophylactic and therapeutic purposes. In this review, we have systematically discussed the effect of passive immunotherapy to combat DEC and explored the types and advancements in antibodies used against antibiotic-resistant DEC.
Collapse
Affiliation(s)
- Srichandrasekar Thuthikkadu Indhuprakash
- Department of Bioengineering, Centre for Research in Infectious Diseases (CRID), School of Chemical & Biotechnology, SASTRA Deemed-to-be-University, Thanjavur, 613401, Tamil Nadu, India
| | - Mukunthan Karthikeyan
- Department of Biotechnology, Centre for Research in Infectious Diseases (CRID), School of Chemical & Biotechnology, SASTRA Deemed-to-be-University, Thanjavur, 613401, Tamil Nadu, India
| | - Gayathri Gopal
- Department of Bioengineering, Centre for Research in Infectious Diseases (CRID), School of Chemical & Biotechnology, SASTRA Deemed-to-be-University, Thanjavur, 613401, Tamil Nadu, India
| | - Senthil Visaga Ambi
- Department of Bioengineering, Centre for Research in Infectious Diseases (CRID), School of Chemical & Biotechnology, SASTRA Deemed-to-be-University, Thanjavur, 613401, Tamil Nadu, India
| | - Saravanan Sekaran
- Department of Pharmacology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai-77, Tamil Nadu, India
| | - Balamurugan Palaniappan
- Department of Biotechnology, Centre for Research in Infectious Diseases (CRID), School of Chemical & Biotechnology, SASTRA Deemed-to-be-University, Thanjavur, 613401, Tamil Nadu, India
| | - Thirumalai Diraviyam
- Department of Bioengineering, Centre for Research in Infectious Diseases (CRID), School of Chemical & Biotechnology, SASTRA Deemed-to-be-University, Thanjavur, 613401, Tamil Nadu, India
| |
Collapse
|
4
|
Caballero-Flores G, Sakamoto K, Zeng MY, Wang Y, Hakim J, Matus-Acuña V, Inohara N, Núñez G. Maternal Immunization Confers Protection to the Offspring against an Attaching and Effacing Pathogen through Delivery of IgG in Breast Milk. Cell Host Microbe 2019; 25:313-323.e4. [PMID: 30686564 DOI: 10.1016/j.chom.2018.12.015] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 11/02/2018] [Accepted: 12/19/2018] [Indexed: 12/15/2022]
Abstract
Owing to immature immune systems and impaired colonization resistance mediated by the microbiota, infants are more susceptible to enteric infections. Maternal antibodies can provide immunity, with maternal vaccination offering a protective strategy. We find that oral infection of adult females with the enteric pathogen Citrobacter rodentium protects dams and offspring against oral challenge. Parenteral immunization of dams with heat-inactivated C. rodentium reduces pathogen loads and mortality in offspring but not mothers. IgG, but not IgA or IgM, transferred through breast milk to the intestinal lumen of suckling offspring, coats the pathogen and reduces intestinal colonization. Protective IgG largely recognizes virulence factors encoded within the locus of enterocyte effacement (LEE) pathogenicity island, including the adhesin Intimin and T3SS filament EspA, which are major antigens conferring protection. Thus, pathogen-specific IgG in breast milk induced during maternal infection or immunization protects neonates against infection with an attaching and effacing pathogen.
Collapse
Affiliation(s)
- Gustavo Caballero-Flores
- Department of Pathology and Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Kei Sakamoto
- Department of Pathology and Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Melody Y Zeng
- Department of Pathology and Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Yaqiu Wang
- Department of Pathology and Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Program in Human Biology, School of Integrative and Global Majors, University of Tsukuba, Tsukuba 305-0006, Japan
| | - Jill Hakim
- Department of Pathology and Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Violeta Matus-Acuña
- Programa de Ecología Genómica, Centro de Ciencias Genómicas, Universidad Nacional Autónoma de México, Cuernavaca, Morelos 62210, Mexico; School of Natural Resources and Environment, University of Michigan, Ann Arbor, MI 48109, USA
| | - Naohiro Inohara
- 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.
| |
Collapse
|
5
|
Torres AG. Maternal immunity, a way to confer protection against enteropathogenic Escherichia coli. JORNAL DE PEDIATRIA (VERSÃO EM PORTUGUÊS) 2017. [DOI: 10.1016/j.jpedp.2017.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
|
6
|
|
7
|
Abstract
ABSTRACT
Antibodies can impact pathogens in the presence or in the absence of effector cells or effector molecules such as complement, and experiments can often sort out with precision the mechanisms by which an antibody inhibits a pathogen
in vitro
. In addition,
in vivo
models, particularly those engineered to knock in or knock out effector cells or effector molecules, are excellent tools for understanding antibody functions. However, it is highly likely that multiple antibody functions occur simultaneously or sequentially in the presence of an infecting organism
in vivo
. The most critical incentive for measuring antibody functions is to provide a basis for vaccine development and for the development of therapeutic antibodies. In this respect, some functions, such as virus neutralization, serve to inhibit the acquisition of a pathogen or limit its pathogenesis. However, antibodies can also enhance replication or contribute to pathogenesis. This review emphasizes those antibody functions that are potentially beneficial to the host. In addition, this review will focus on the effects of antibodies on organisms themselves, rather than on the toxins the organisms may produce.
Collapse
|
8
|
Forthal DN. Functions of Antibodies. Microbiol Spectr 2014; 2:1-17. [PMID: 25215264 PMCID: PMC4159104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023] Open
Affiliation(s)
- Donald N. Forthal
- Chief, Infectious Diseases, University of California, Irvine, 3044 Hewitt Hall, Irvine, CA 92617, 949-824-3366
| |
Collapse
|
9
|
Physiopathological effects of Escherichia coli O157:H7 inoculation in weaned calves fed with colostrum containing antibodies to EspB and Intimin. Vaccine 2014; 32:3823-9. [DOI: 10.1016/j.vaccine.2014.04.073] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Revised: 04/18/2014] [Accepted: 04/22/2014] [Indexed: 11/22/2022]
|
10
|
Gavilanes-Parra S, Mendoza-Hernández G, Chávez-Berrocal ME, Girón JA, Orozco-Hoyuela G, Manjarrez-Hernández A. Identification of secretory immunoglobulin A antibody targets from human milk in cultured cells infected with enteropathogenic Escherichia coli (EPEC). Microb Pathog 2013; 64:48-56. [PMID: 24036180 DOI: 10.1016/j.micpath.2013.09.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Revised: 08/30/2013] [Accepted: 09/02/2013] [Indexed: 12/27/2022]
Abstract
Enteropathogenic Escherichia coli (EPEC) uses a type III secretion system (T3SS) to inject effectors into host cells and alter cellular physiology. The aim of the present study was to identify targets of human secretory immunoglobulin A (sIgA) antibodies from the proteins delivered by EPEC into HEp-2 cells after infection. Bacterial proteins delivered into EPEC-infected cells were obtained in sub-cellular fractions (cytoplasmic, membrane, and cytoskeleton) and probed with sIgA antibodies from human milk and analyzed by Western blotting. These sIgA antibodies reacted with Tir and EspB in the cytoplasmic and membrane fractions, and with intimin in the membrane fraction mainly. The sIgA also identified an EPEC surface-associated Heat-shock protein 70 (Hsp70) in HEp-2 cells infected with EPEC. Purified Hsp70 from EPEC was able to bind to HEp-2 cells, suggesting adhesive properties in this protein. EspC secreted to the medium reacted strongly with the sIgA antibodies. An EPEC 115 kDa protein, unrelated to the EspC protein, was detected in the cytoplasm of infected HEp-2 cells, suggesting that this is a new protein translocated by EPEC. The results suggest that there is a strong host antibody response to Tir and intimin, which are essential proteins for attaching and effacing (A/E) pathogen mediated disease.
Collapse
Affiliation(s)
- Sandra Gavilanes-Parra
- Departamento de Salud Publica, Facultad de Medicina, Universidad Nacional Autónoma de México, D. F. 04510, Mexico
| | | | | | | | | | | |
Collapse
|
11
|
Rabinovitz BC, Gerhardt E, Tironi Farinati C, Abdala A, Galarza R, Vilte DA, Ibarra C, Cataldi A, Mercado EC. Vaccination of pregnant cows with EspA, EspB, γ-intimin, and Shiga toxin 2 proteins from Escherichia coli O157:H7 induces high levels of specific colostral antibodies that are transferred to newborn calves. J Dairy Sci 2012; 95:3318-26. [PMID: 22612965 DOI: 10.3168/jds.2011-5093] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2011] [Accepted: 02/05/2012] [Indexed: 01/09/2023]
Abstract
Enterohemorrhagic Escherichia coli (EHEC) O157:H7 is a major cause of intestinal disease and hemolytic uremic syndrome, a serious systemic complication that particularly affects children. Cattle are primary reservoirs for EHEC O157:H7 and the main source of infection for humans. Vaccination of cattle with different combinations of bacterial virulence factors has shown efficacy in decreasing EHEC O157:H7 shedding. It is, therefore, important to demonstrate whether vaccination of pregnant cows with EHEC O157:H7 induces high titers of transferable antibodies to avoid early colonization of calves by the bacteria. In this study we evaluated the ability of EspA, EspB, the C-terminal fragment of 280 amino acids of γ-intimin (γ-intimin C₂₈₀) and inactivated Shiga toxin (Stx) 2 proteins to induce specific antibodies in colostrum and their passive transference to colostrum-fed calves. Friesian pregnant cows immunized by the intramuscular route mounted significantly high serum and colostrum IgG responses against EspB and γ-intimin C₂₈₀ that were efficiently transferred to their calves. Antibodies to EspB and γ-intimin C₂₈₀ were detected in milk samples of vaccinated cows at d 40 postparturition. Significant Stx2-neutralizing titers were also observed in colostrum from Stx2-vaccinated cows and sera from colostrum-fed calves. The results presented showed that bovine colostrum with increased levels of antibodies against EHEC O157:H7 may be obtained by systemic immunization of pregnant cows, and that these specific antibodies are efficiently transferred to newborn calves by feeding colostrum. Hyperimmune colostrum and milk may be an alternative to protect calves from early colonization by EHEC O157:H7 and a possible key source of antibodies to block colonization and toxic activity of this bacterium.
Collapse
Affiliation(s)
- B C Rabinovitz
- Instituto de Patobiología, Instituto Nacional de Tecnología Agropecuaria-INTA, Nicolás Repetto y De los Reseros, 1686 Hurlingham, Buenos Aires, Argentina
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Beutin L, Martin A. Outbreak of Shiga toxin-producing Escherichia coli (STEC) O104:H4 infection in Germany causes a paradigm shift with regard to human pathogenicity of STEC strains. J Food Prot 2012; 75:408-18. [PMID: 22289607 DOI: 10.4315/0362-028x.jfp-11-452] [Citation(s) in RCA: 163] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
An outbreak that comprised 3,842 cases of human infections with enteroaggregative hemorrhagic Escherichia coli (EAHEC) O104:H4 occurred in Germany in May 2011. The high proportion of adults affected in this outbreak and the unusually high number of patients that developed hemolytic uremic syndrome makes this outbreak the most dramatic since enterohemorrhagic E. coli (EHEC) strains were first identified as agents of human disease. The characteristics of the outbreak strain, the way it spread among humans, and the clinical signs resulting from EAHEC infections have changed the way Shiga toxin-producing E. coli strains are regarded as human pathogens in general. EAHEC O104:H4 is an emerging E. coli pathotype that is endemic in Central Africa and has spread to Europe and Asia. EAHEC strains have evolved from enteroaggregative E. coli by uptake of a Shiga toxin 2a (Stx2a)-encoding bacteriophage. Except for Stx2a, no other EHEC-specific virulence markers including the locus of enterocyte effacement are present in EAHEC strains. EAHEC O104:H4 colonizes humans through aggregative adherence fimbrial pili encoded by the enteroaggregative E. coli plasmid. The aggregative adherence fimbrial colonization mechanism substitutes for the locus of enterocyte effacement functions for bacterial adherence and delivery of Stx2a into the human intestine, resulting clinically in hemolytic uremic syndrome. Humans are the only known natural reservoir known for EAHEC. In contrast, Shiga toxin-producing E. coli and EHEC are associated with animals as natural hosts. Contaminated sprouted fenugreek seeds were suspected as the primary vehicle of transmission of the EAHEC O104:H4 outbreak strain in Germany. During the outbreak, secondary transmission (human to human and human to food) was important. Epidemiological investigations revealed fenugreek seeds as the source of entry of EAHEC O104:H4 into the food chain; however, microbiological analysis of seeds for this pathogen produced negative results. The survival of EAHEC in seeds and the frequency of human carriers of EAHEC should be investigated for a better understanding of EAHEC transmission routes.
Collapse
Affiliation(s)
- Lothar Beutin
- National Reference Laboratory for Escherichia coli, Unit 41, Microbial Toxins, Germany.
| | | |
Collapse
|
13
|
Vilte DA, Larzábal M, Garbaccio S, Gammella M, Rabinovitz BC, Elizondo AM, Cantet RJ, Delgado F, Meikle V, Cataldi A, Mercado EC. Reduced faecal shedding of Escherichia coli O157:H7 in cattle following systemic vaccination with γ-intimin C280 and EspB proteins. Vaccine 2011; 29:3962-8. [DOI: 10.1016/j.vaccine.2011.03.079] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2011] [Revised: 03/07/2011] [Accepted: 03/22/2011] [Indexed: 10/18/2022]
|
14
|
Distribution of non-locus of enterocyte effacement pathogenic island-related genes in Escherichia coli carrying eae from patients with diarrhea and healthy individuals in Japan. J Clin Microbiol 2010; 48:4107-14. [PMID: 20844211 DOI: 10.1128/jcm.00677-10] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The relationship to diarrhea of genes located on the pathogenicity islands (PAI) other than the locus of enterocyte effacement (LEE) was investigated. Enteropathogenic Escherichia coli (EPEC), the retention of espC on the EspC PAI, the OI-122 genes (efa1/lifA, nleB), the phylogenetic marker gene yjaA, and the bundle-forming pilus gene bfpA on the EPEC adherence factor (EAF) plasmid were studied. E. coli strains carrying the intimin gene (eae) without the Shiga toxin gene, isolated from patients with diarrhea (n = 83) and healthy individuals (n = 38) in Japan, were evaluated using PCR. The genotypes of eae and espC were identified by heteroduplex mobility assay (HMA). The proportions of strains isolated from individuals with and without diarrhea that carried these genes were as follows: bfpA, 13.3 and 7.9%, respectively; espC, 25.3 and 36.8%; efa1/lifA, 32.5 and 13.2%; nleB, 63.9 and 60.5%; yjaA, 42.2 and 55.3%. Statistical significance (P < 0.05) was achieved only for efa1/lifA. The proportion of strains lacking espC and carrying efa1/lifA was higher for patient-derived strains (30.1%) than for strains from healthy individuals (13.2%), but the difference was not significant. Strains carrying both espC and efa1/lifA were rare (2 strains from patients). Statistical analyses revealed significant relationships between espC and yjaA and between efa1/lifA and nleB, as well as significant inverse relationships between espC and efa1/lifA and between efa1/lifA and yjaA. espC was found in eae HMA types a1, a2, and c2, whereas efa1/lifA was found in types b1, b2, and c1. In addition, 6 polymorphisms of espC were found. The espC, yjaA, efa1/lifA, and nleB genes were mutually dependent, and their distributions were related to eae type, findings that should be considered in future epidemiological studies.
Collapse
|
15
|
Orden JA, De la Fuente R, Yuste M, Martínez-Pulgarín S, Ruiz-Santa-Quiteria JA, Horcajo P, Contreras A, Sánchez A, Corrales JC, Domínguez-Bernal G. Kinetics and role of antibodies against intimin beta in colostrum and in serum from goat kids and longitudinal study of attaching and effacing Escherichia coli in goat kids. CANADIAN JOURNAL OF VETERINARY RESEARCH = REVUE CANADIENNE DE RECHERCHE VETERINAIRE 2010; 74:54-58. [PMID: 20357960 PMCID: PMC2801313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/17/2008] [Accepted: 01/05/2009] [Indexed: 05/29/2023]
Abstract
The presence of antibodies to the intimin beta-binding region (Int280-beta) of attaching and effacing Escherichia coli (AEEC) in serum from 20 goat kids from 2 herds, as well as in goat colostrum, was investigated by enzyme-linked immunosorbent assay. In addition, the onset and subsequent pattern of shedding of AEEC from the same goat kids over a 6-mo period was investigated. All the colostrum and serum samples tested contained antibodies against Int280-beta. The association between the antibody titer and the isolation of AEEC suggests that antibodies to intimin beta do not prevent colonization of the intestine by AEEC in goat kids. The AEEC were generally shed only transiently. Most AEEC isolated from the kids belonged to serogroup O26. Three isolates belonged to serogroup O157. These data show that goat kids may be a reservoir of AEEC that are potentially pathogenic for humans.
Collapse
Affiliation(s)
- José A Orden
- Departamento de Sanidad Animal, Facultad de Veterinaria, Universidad Complutense, Madrid, Spain.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Bovine colostrum contains immunoglobulin G antibodies against intimin, EspA, and EspB and inhibits hemolytic activity mediated by the type three secretion system of attaching and effacing Escherichia coli. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2008; 15:1208-13. [PMID: 18562563 DOI: 10.1128/cvi.00027-08] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Enterohemorrhagic Escherichia coli (EHEC) is the main cause of hemolytic-uremic syndrome, an endemic disease in Argentina which had an incidence in 2005 of 13.9 cases per 100,000 children younger than 5 years old. Cattle appear to be a major reservoir of EHEC, and a serological response to EHEC antigens has been demonstrated in natural and experimental infections. In the current study, antibodies against proteins implicated in EHEC's ability to form attaching and effacing lesions, some of which are exported to the host cell via a type three secretion system (TTSS), were identified in bovine colostrum by Western blot analysis. Twenty-seven (77.0%) of the 35 samples examined contained immunoglobulin G (IgG) antibodies against the three proteins assayed in this study: EspA, EspB, and the carboxy-terminal 280 amino acids of gamma-intimin, an intimin subtype associated mainly with O157:H7 and O145:H- serotypes. Every colostrum sample was able to inhibit, in a range between 45.9 and 96.7%, the TTSS-mediated hemolytic activity of attaching and effacing E. coli. The inhibitory effect was partially mediated by IgG and lactoferrin. In conclusion, we found that early colostrum from cows contains antibodies, lactoferrin, and other unidentified substances that impair TTSS function in attaching and effacing E. coli strains. Bovine colostrum might act by reducing EHEC colonization in newborn calves and could be used as a prophylactic measure to protect non-breast-fed children against EHEC infection in an area of endemicity.
Collapse
|
17
|
Hosea Blewett HJ, Cicalo MC, Holland CD, Field CJ. The immunological components of human milk. ADVANCES IN FOOD AND NUTRITION RESEARCH 2008; 54:45-80. [PMID: 18291304 DOI: 10.1016/s1043-4526(07)00002-2] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Breast-feeding is generally accepted as the optimal method of feeding infants. However, we have yet to fully understand the complex mixture of bioactive compounds contained in human milk. Epidemiological studies have indicated that breast-feeding is associated with health benefits in the infant for many immune-related conditions. Breast milk contains various antimicrobial substances, factors that promote immune development, constituents that promote tolerance/priming of the infant immune system, as well as anti-inflammatory components. This chapter identifies and discusses the immunological compounds in human milk and the available evidence for their effect on the immune system of the infant. Current feeding regimens recommended for infants are based primarily on the current understanding of the nutritional requirements of the neonate, but perhaps will be modified to reflect the consequences on immune function both immediate and later in life.
Collapse
Affiliation(s)
- Heather J Hosea Blewett
- Department of Agricultural, Food and Nutritional Sciences, Alberta Institute for Human Nutrition, University of Alberta, Edmonton, Alberta, T6G 2P5, Canada
| | | | | | | |
Collapse
|
18
|
Schuijffel DF, van Empel PCM, Pennings AMMA, van Putten JPM, Nuijten PJM. Successful selection of cross-protective vaccine candidates for Ornithobacterium rhinotracheale infection. Infect Immun 2005; 73:6812-21. [PMID: 16177359 PMCID: PMC1230975 DOI: 10.1128/iai.73.10.6812-6821.2005] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Ornithobacterium rhinotracheale is a bacterial pathogen known for causing respiratory disease in poultry. In this study, we demonstrate for the first time that cross-protective immunity against different O. rhinotracheale serotypes can be induced by live vaccination. Sera from these live-vaccinated and cross-protected birds were used to identify new vaccine targets by screening an O. rhinotracheale expression library. Out of 20,000 screened plaques, a total of 30 cross-reactive clones were selected for further analysis. Western blot analysis and DNA sequencing identified eight different open reading frames. The genes encoding the eight cross-reactive antigens were amplified, cloned in an expression vector, and expressed in Escherichia coli. Purified recombinant proteins with a molecular mass ranging from 35.9 kDa to 62.9 kDa were mixed and tested as a subunit vaccine for (cross-)protection against challenge with homologous and heterologous O. rhinotracheale serotypes in chickens. Subunit vaccination resulted in the production of antibodies reactive to the recombinant proteins on Western blot, and this eight-valent vaccine conferred both homologous and heterologous protection against O. rhinotracheale challenge in chickens.
Collapse
Affiliation(s)
- D F Schuijffel
- Intervet International BV, Bacteriology R&D, Boxmeer, The Netherlands
| | | | | | | | | |
Collapse
|
19
|
Abstract
This review focuses on the function of the Escherichia coli and Salmonella autotransporters for which a considerable amount of literature is available. Members of the serine protease autotransporters of the Enterobacteriaceae (SPATEs) family are proteins from E. coli and Shigella spp., which, like the Neisseria and Haemophilus influenzae IgA1 proteases and Hap, possess a consensus serine protease motif. The largest subfamily of autotransporters is defined by the AidA conserved domain COG3468 and consists of members from a diverse range of animal and plant pathogens including E. coli, S. enterica, Yersinia pestis. This subfamily, which is composed of more than 55 proteins, possesses some of the best-characterized autotransporter proteins including the S. flexneri mediator of motility IcsA, the major phase-variable E. coli outer membrane protein antigen 43 (Ag43) and the diffuse adhering E. coli (DAEC) adhesin AIDA-I, from which this subfamily derives its name. Another member of the AIDA-I family, and one of the most studied autotransporter proteins, is IcsA. The autotransporter pathway is emerging as the most common mechanism of protein translocation across the gram-negative outer membrane.
Collapse
Affiliation(s)
- Ian R Henderson
- Bacterial Pathogenesis and Genomics Unit, Division of Immunity and Infection, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - James P Nataro
- Center for Vaccine Development, Department of Pediatrics, University of Maryland School of Medicine, 685 W. Baltimore St., Baltimore, MD 21201
| |
Collapse
|
20
|
Carbonare CB, Carbonare SB, Carneiro-Sampaio MMS. Secretory immunoglobulin A obtained from pooled human colostrum and milk for oral passive immunization. Pediatr Allergy Immunol 2005; 16:574-81. [PMID: 16238582 DOI: 10.1111/j.1399-3038.2005.00332.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Passive immunization is useful in cases of immunodeficiencies or infectious diseases, but usually seric gammaglobulin or hyperimmune sera are administered parenterally, providing good systemic immunization, though with low protection of the mucosal surfaces. The oral administration of secretory antibodies, especially surface immunoglobulin (SIg)A, which is perfectly adapted to the mucosal environment, would therefore, be preferable. The aim of the present study was to obtain a SIgA preparation from pooled human colostrum and milk, which should maintain the essential properties of the antibodies suitable for clinical oral administration. IgA preparations were obtained from colostrum and milk pools by salt precipitation. The final products were evaluated in terms of yield and purity, as well as antibody activity to bacterial antigens and toxins and inhibitory activity of bacterial adhesion to epithelial cells. The best yield and purity were achieved when the colostrum pool was used as a source of IgA; the final product showed very few bands of protein contamination. The IgA preparations preserved the antibody reactivity against various microbial antigens, well comparable with the reactivity exhibited by the original milk and colostrum pools. SIgA preparations were able to inhibit greatly the adhesion of enteropathogenic Escherichia coli to Hep-2 cells and the invasion of enteroinvasive E. coli. These promising results show the feasibility of obtaining SIgA suitable for oral use, which may in future be administered to immunodeficient patients with gastrointestinal manifestations, from human colostrum and milk.
Collapse
Affiliation(s)
- Cristiane B Carbonare
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.
| | | | | |
Collapse
|
21
|
Uren TK, Wijburg OLC, Simmons C, Johansen FE, Brandtzaeg P, Strugnell RA. Vaccine-induced protection against gastrointestinal bacterial infections in the absence of secretory antibodies. Eur J Immunol 2005; 35:180-8. [PMID: 15593123 DOI: 10.1002/eji.200425492] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Secretory IgA (SIgA) is widely held to be responsible for the defense of the mucosae against pathogenics and other potentially harmful agents. In this study, polymeric Ig receptor (pIgR) knockout mice, which lack secretory antibodies (SAb), were used to investigate the role of vaccine-elicited SAb in protection against gastrointestinal bacterial infections. An essential role for specific SAb in protection against Vibrio cholerae was evident from experiments showing that vaccinated pIgR(-/-) mice, but not vaccinated C57BL/6 mice, were susceptible to cholera toxin challenge. Vaccination of C57BL/6 mice with Salmonella typhimurium elicited strong antigen-specific, mucosal responses, which blocked in vitro invasion of epithelia. However, vaccinated C57BL/6 and pIgR(-/-) mice were equally resistant to challenge infection with virulent S. typhimurium. Finally, we investigated the importance of SIgA in protection against recurrent infections with Citrobacter rodentium. Although higher numbers of bacteria were detected early after challenge infection in feces of vaccinated pIgR(-/-) mice compared with vaccinated C57BL/6 mice, both mouse strains showed complete clearance after 9 days. These results suggested that, in immune animals, SIgA is crucial for the protection of gastrointestinal surfaces against secreted bacterial toxins, may inhibit early colonization by C. rodentium, but is not essential for protection against re-infection with S. typhimurium or C. rodentium.
Collapse
Affiliation(s)
- Tania K Uren
- The CRC for Vaccine Technology, The University of Melbourne, Department of Microbiology and Immunology, Melbourne, Victoria 3010, Australia
| | | | | | | | | | | |
Collapse
|
22
|
Henderson IR, Navarro-Garcia F, Desvaux M, Fernandez RC, Ala'Aldeen D. Type V protein secretion pathway: the autotransporter story. Microbiol Mol Biol Rev 2004; 68:692-744. [PMID: 15590781 PMCID: PMC539010 DOI: 10.1128/mmbr.68.4.692-744.2004] [Citation(s) in RCA: 595] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Gram-negative bacteria possess an outer membrane layer which constrains uptake and secretion of solutes and polypeptides. To overcome this barrier, bacteria have developed several systems for protein secretion. The type V secretion pathway encompasses the autotransporter proteins, the two-partner secretion system, and the recently described type Vc or AT-2 family of proteins. Since its discovery in the late 1980s, this family of secreted proteins has expanded continuously, due largely to the advent of the genomic age, to become the largest group of secreted proteins in gram-negative bacteria. Several of these proteins play essential roles in the pathogenesis of bacterial infections and have been characterized in detail, demonstrating a diverse array of function including the ability to condense host cell actin and to modulate apoptosis. However, most of the autotransporter proteins remain to be characterized. In light of new discoveries and controversies in this research field, this review considers the autotransporter secretion process in the context of the more general field of bacterial protein translocation and exoprotein function.
Collapse
Affiliation(s)
- Ian R Henderson
- Division of Immunity and Infection, University of Birmingham, Birmingham B15 2TT, UK.
| | | | | | | | | |
Collapse
|
23
|
Beutin L, Krause G, Zimmermann S, Kaulfuss S, Gleier K. Characterization of Shiga toxin-producing Escherichia coli strains isolated from human patients in Germany over a 3-year period. J Clin Microbiol 2004; 42:1099-108. [PMID: 15004060 PMCID: PMC356890 DOI: 10.1128/jcm.42.3.1099-1108.2004] [Citation(s) in RCA: 268] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We have investigated 677 Shiga toxin-producing Escherichia coli (STEC) strains from humans to determine their serotypes, virulence genes, and clinical signs in patients. Six different Shiga toxin types (1, 1c, 2, 2c, 2d, and 2e) were distributed in the STEC strains. Intimin (eae) genes were present in 62.6% of the strains and subtyped into intimins alpha1, beta1, gamma1, epsilon, theta, and eta. Shiga toxin types 1c and 2d were present only in eae-negative STEC strains, and type 2 was significantly (P < 0.001) more frequent in eae-positive STEC strains. Enterohemorrhagic E. coli hemolysin was associated with 96.2% of the eae-positive strains and with 65.2% of the eae-negative strains. Clinical signs in the patients were abdominal pain (8.7%), nonbloody diarrhea (59.2%), bloody diarrhea (14.3%), and hemolytic-uremic syndrome (HUS) (3.5%), and 14.3% of the patients had no signs of gastrointestinal disease or HUS. Infections with eae-positive STEC were significantly (P < 0.001) more frequent in children under 6 years of age than in other age groups, whereas eae-negative STEC infections dominated in adults. The STEC strains were grouped into 74 O:H types by serotyping and by PCR typing of the flagellar (fliC) genes in 221 nonmotile STEC strains. Eleven serotypes (O157:[H7], O26:[H11], O103:H2, O91:[H14], O111:[H8], O145:[H28], O128:H2, O113:[H4], O146:H21, O118:H16, and O76:[H19]) accounted for 69% of all STEC strains. We identified 41 STEC strains belonging to 31 serotypes which had not previously been described as human STEC. Twenty-six of these were positive for intimins alpha1 (one serotype), beta1 (eight serotypes), epsilon (two serotypes), and eta (three serotypes). Our study indicates that different types of STEC strains predominate in infant and adult patients and that new types of STEC strains are present among human isolates.
Collapse
Affiliation(s)
- Lothar Beutin
- Division of Microbial Toxins, Department of Biological Safety, Robert Koch Institute, D-13353 Berlin, Germany.
| | | | | | | | | |
Collapse
|
24
|
Simmons CP, Clare S, Ghaem-Maghami M, Uren TK, Rankin J, Huett A, Goldin R, Lewis DJ, MacDonald TT, Strugnell RA, Frankel G, Dougan G. Central role for B lymphocytes and CD4+ T cells in immunity to infection by the attaching and effacing pathogen Citrobacter rodentium. Infect Immun 2003; 71:5077-86. [PMID: 12933850 PMCID: PMC187366 DOI: 10.1128/iai.71.9.5077-5086.2003] [Citation(s) in RCA: 141] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Citrobacter rodentium, an attaching-effacing bacterial pathogen, establishes an acute infection of the murine colonic epithelium and induces a mild colitis in immunocompetent mice. This study describes the role of T-cell subsets and B lymphocytes in immunity to C. rodentium. C57Bl/6 mice orally infected with C. rodentium resolved infection within 3 to 4 weeks. Conversely, systemic and colonic tissues of RAG1(-/-) mice orally infected with C. rodentium contained high and sustained pathogen loads, and in the colon this resulted in a severe colitis. C57Bl/6 mice depleted of CD4(+) T cells, but not CD8(+) T cells, were highly susceptible to infection and also developed severe colitis. Mice depleted of CD4(+) T cells also had diminished immunoglobulin G (IgG) and IgA antibody responses to two C. rodentium virulence-associated determinants, i.e., EspA and intimin, despite having a massively increased pathogen burden. Mice with an intact T-cell compartment, but lacking B cells ( micro MT mice), were highly susceptible to C. rodentium infection. Systemic immunity, but not mucosal immunity, could be restored by adoptive transfer of convalescent immune sera to infected micro MT mice. Adoptive transfer of immune B cells, but not naïve B cells, provided highly variable immunity to recipient micro MT mice. The results suggest that B-cell-mediated immune responses are central to resolution of a C. rodentium infection but that the mechanism through which this occurs requires further investigation. These data are relevant to understanding immunity to enteric attaching and effacing bacterial pathogens of humans.
Collapse
Affiliation(s)
- Cameron P Simmons
- Centre for Molecular Microbiology and Infection, Department of Biological Sciences, Imperial College of Science, Technology and Medicine, South Kensington, London SW6 1SJ, United Kingdom.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Ochoa TJ, Noguera-Obenza M, Ebel F, Guzman CA, Gomez HF, Cleary TG. Lactoferrin impairs type III secretory system function in enteropathogenic Escherichia coli. Infect Immun 2003; 71:5149-55. [PMID: 12933858 PMCID: PMC187368 DOI: 10.1128/iai.71.9.5149-5155.2003] [Citation(s) in RCA: 81] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Enteropathogenic Escherichia coli (EPEC) is an important cause of infant diarrhea in developing countries. EPEC uses a type III secretory system to deliver effector proteins into the host cell. These proteins cause the characteristic attaching and effacing lesion on enterocytes. Lactoferrin, a glycoprotein present in human milk, inhibits EPEC adherence to mammalian cells. To determine the effect of lactoferrin on the initial host cell attachment step that is mediated by the type III secretory system, we focused on EPEC-induced actin polymerization in HEp2 cells, on the hemolytic activity, and on measurement of E. coli secreted proteins A, B, and D (EspABD). Lactoferrin blocked EPEC-mediated actin polymerization in HEp2 cells and blocked EPEC-induced hemolysis. The mechanism of this inhibition was lactoferrin-mediated degradation of secreted proteins necessary for bacterial contact and pore formation, particularly EspB. The proteolytic effect of lactoferrin was prevented by serine protease inhibitors. This disruption of the type III secretory system implies that lactoferrin could provide broad cross protection against the enteropathogens that share this mechanism.
Collapse
Affiliation(s)
- Theresa J Ochoa
- Division of Pediatric Infectious Diseases, University of Texas-Houston Medical School, Houston, Texas 77040, USA
| | | | | | | | | | | |
Collapse
|
26
|
Noguera-Obenza M, Ochoa TJ, Gomez HF, Guerrero ML, Herrera-Insua I, Morrow AL, Ruiz-Palacios G, Pickering LK, Guzman CA, Cleary TG. Human milk secretory antibodies against attaching and effacing Escherichia coli antigens. Emerg Infect Dis 2003; 9:545-51. [PMID: 12737737 PMCID: PMC2972756 DOI: 10.3201/eid0905.020441] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Secretory immunoglobulin A (sIgA) is a primary factor responsible for preventing attachment of enteropathogens to gut epithelium in breastfeeding infants. We compared the frequency of sIgA to major surface antigens of enterohemorrhagic Escherichia coli (EHEC) in milk of 123 women from the United States and Mexico to determine whether regional differences existed in the frequency of antibodies to these surface antigens. In both groups of women, milk commonly has sIgA against various EHEC lipopolysaccharides, EspA, EspB, intimin, and less frequently against Shiga toxin. The study suggests that persons living in the United States are exposed to attaching/effacing enteropathogens more frequently than is generally assumed. The low frequency of antibodies to Stx1 (in 12% of Mexican and in 22% of U.S. samples) suggests that the rare appearance of hemolytic uremic syndrome in adults is not due to neutralization of toxin at the gut level. Only anti-EspA is found in most milk samples from both populations of women. EspA may represent a useful target for an immunization strategy to prevent EHEC disease in humans.
Collapse
Affiliation(s)
| | | | - Henry F. Gomez
- University of Texas-Houston Medical School, Houston, Texas, USA
| | - M. Lourdes Guerrero
- Instituto Nacional de la Nutricion “Salvador Zubiran,” México City, México, D. F
| | | | | | | | | | - Carlos A. Guzman
- GBF-German National Research Centre for Biotechnology, Braunschweig, Germany
| | | |
Collapse
|
27
|
de Souza Campos Fernandes RC, Quintana Flores VM, Sousa de Macedo Z, Medina-Acosta E. Coproantibodies to the enteropathogenic Escherichia coli vaccine candidates BfpA and EspB in breastfed and artificially fed children. Vaccine 2003; 21:1725-31. [PMID: 12639496 DOI: 10.1016/s0264-410x(02)00525-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A diagnostic test was developed to discriminate active from past enteropathogenic Escherichia coli (EPEC) infection, which uses the affinity-purified recombinant proteins BfpA (bundle-forming pilus (BFP) structural repeating subunit A) and EspB (pore-forming secreted protein B) as reliable markers of virulence to detect antigen-specific coproantibodies by immunoblot analysis, and verification of active typical EPEC infection by gene-specific (bfpA and espB) PCR amplification using DNA extracted directly from specimens and/or culture-enriched preparations. To begin addressing the potential protective role of anti-EPEC antibodies at early age, the prevalence of IgA coproantibodies to these antigens was determined in either breastfed or artificially fed children <2 years of age hospitalized for watery diarrhea.
Collapse
|
28
|
Horne C, Vallance BA, Deng W, Finlay BB. Current progress in enteropathogenic and enterohemorrhagic Escherichia coli vaccines. Expert Rev Vaccines 2002; 1:483-93. [PMID: 12901587 DOI: 10.1586/14760584.1.4.483] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Enteropathogenic and enterohemorrhagic Escherichia coli are important causal agents of infectious diarrhea, particularly amongst pediatric populations. While enteropathogenic E. coli is a significant health threat in developing countries, enterohemorrhagic E. coli causes sporadic, sometimes deadly outbreaks of hemorrhagic colitis, with a serious complication, hemolytic uremic syndrome, ocurring in a proportion of cases. This review discusses the pathogenesis of enterohemorrhagic and enteropathogenic E. coli, the host immune response and the current application of this knowledge towards efficacious vaccine strategies. Several lines of investigation indicate the feasibility of such strategies and justify further development of a vaccine targeting these significant intestinal pathogens.
Collapse
Affiliation(s)
- Cathy Horne
- Biochemistry Labouratory, University of British Columbia, Vancouver, BC, Canada.
| | | | | | | |
Collapse
|
29
|
Yokomizo Y, Watanabe F, Imada Y, Inumaru S, Yanaka T, Tsuji T. Mucosal immunoadjuvant activity of the low toxic recombinant Escherichia coli heat-labile enterotoxin produced by Bacillus brevis for the bacterial subunit or component vaccine in pigs and cattle. Vet Immunol Immunopathol 2002; 87:291-300. [PMID: 12072249 DOI: 10.1016/s0165-2427(02)00055-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
A gene encoding the mature Escherichia coli heat-labile enterotoxin (LT) lacking the nick site in the A subunit by deleting tripeptides was introduced in a vector pNH301 and expressed extracellularly as mutant molecule of holotoxin at high levels in Bacilus brevis HPD31-S5 of the host bacterium. The mucosal adjuvant activities of the produced mutant LT (mLT) preparation were studied in pigs and cattle. Intranasal immunization of pigs with the recombinant subunit vaccine of Erysipelothrix rhusiopathiae or the component vaccine of Bordetella bronchiseptica mixed with the mLT resulted in a substantial enhancement of both mucosal and serum-specific antibody levels. The immunized pigs were also protected when challenge-exposed intradermally with a highly virulent E. rhusiopathiae strain or challenge-exposed intranasally with a highly virulent strain of B. bronchiseptica. The mLT intranasally administered with recombinant intimin (an outer membrane adhesin) of E. coli O157:H7 also induced an elevation of IgA-specific antibody in the nasal secretion and saliva of calves as well as an elevation of IgG1-specific antibody level against the intimin in the sera and colostrum of cows. The three kinds tested protein antigens were poorly immunogenic when antigen administered intranasally alone. The mLT intranasally administered at a higher effective dose did not induce local adverse reactions or diarrhea in pigs and cattle. The present study demonstrates that the recombinant mLT produced using the B. brevis expression system might represent promising immunoadjuvants for the potential application of intranasal vaccines directed against infectious diseases in pigs and cattle.
Collapse
Affiliation(s)
- Y Yokomizo
- National Institute of Animal Health, Kannondai 3-1-5, Ibarakiken, Japan.
| | | | | | | | | | | |
Collapse
|
30
|
Köhler H, Donarski S, Stocks B, Parret A, Edwards C, Schroten H. Antibacterial characteristics in the feces of breast-fed and formula-fed infants during the first year of life. J Pediatr Gastroenterol Nutr 2002; 34:188-93. [PMID: 11840038 DOI: 10.1097/00005176-200202000-00015] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
BACKGROUND Human milk is known to protect infants from a number of infectious diseases. Much less is known about the bioactivity of milk-derived factors in the intestine. In this study, potentially protective characteristics in the feces of breast-fed and formula-fed infants were compared. METHODS The feces of 26 breast-fed and 18 formula-fed infants were collected during the first year of life. In each sample, the concentrations of total protein, immunoglobulin A, and sialic acid were measured. In addition, the effect of the fecal samples was measured on the adhesion of enteropathogenic Escherichia coli (EPEC) to Caco-2 cells and on transepithelial electrical resistance (TER) during an infection. RESULTS In the first month, sialic acid and immunoglobulin A were found in the feces of breast-fed infants in substantially higher concentrations than in the feces of formula fed infants (sialic acid, 1197 +/- 370 microg/ mL versus 31 +/- 19 microg/ mL; immunoglobulin A, 0.11 +/- 7 mg/mL versus 0.3 +/- 1 mg/mL) and thereafter decreased to similar levels in half a year. Adhesion of EPEC to Caco-2 cells was inhibited between 65% and 85% by stools from both groups. The decrease of TER during EPEC infection was unaffected by fecal samples of any origin or age. CONCLUSION Potentially protective factors are present in higher concentrations in the stools of breast-fed infants than in stools of formula-fed infants. Interestingly, feces from breast-fed and formula-fed infants inhibited bacterial adhesion to a similar level, but neither was able to preserve epithelial barrier function.
Collapse
Affiliation(s)
- Henrik Köhler
- University Children's Hospital, Düsseldorf, Germany.
| | | | | | | | | | | |
Collapse
|
31
|
Delahay RM, Frankel G, Knutton S. Intimate interactions of enteropathogenic Escherichia coli at the host cell surface. Curr Opin Infect Dis 2001; 14:559-65. [PMID: 11964876 DOI: 10.1097/00001432-200110000-00009] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Unlike many gastrointestinal pathogens, enteropathogenic Escherichia coli orchestrates the modulation of host cellular and immune responses from the exterior of the infected cell, chiefly via the secreted and translocated components of a type III secretion system. Close inspection of these enteropathogenic Escherichia coli proteins and the interactions they mediate provides an increasingly coherent picture of the pathogenic mechanisms that enteropathogenic Escherichia coli uses to exploit its host.
Collapse
Affiliation(s)
- R M Delahay
- Centre for Molecular Microbiology and Infection, Imperial College of Science, Technology and Medicine, London, UK.
| | | | | |
Collapse
|
32
|
Abstract
Mammalian colostrum offers passive protection to the newborn against a variety of microbial pathogens, in the form of specific immunoglobulin A, G and M antibodies. Sharing maternal immunological memory is in many cases vital for the infant, but may have disastrous consequences, such as involuntary transfer of disease and disturbance of the developing immune system. In most published studies, immune milk preparations are reported to be effective in the prevention of various gastroenteric infections, but not in the treatment of an established infection.
Collapse
Affiliation(s)
- E M Lilius
- Department of Biochemistry and Food Chemistry, University of Turku, Turku, Finland.
| | | |
Collapse
|
33
|
Affiliation(s)
- W T Todd
- Lanarkshire Area Infectious Diseases Unit, Monklands Hospital, Airdale, ML6 OJS, Lanarkshire, UK.
| |
Collapse
|