1
|
Messina NL, Wang M, Forbes EK, Freyne B, Hasang WP, Germano S, Bonnici R, Summons F, Gardiner K, Donath S, Gordon R, Rogerson SJ, Curtis N. The influence of neonatal BCG vaccination on in vitro cytokine responses to Plasmodium falciparum. BMC Immunol 2024; 25:24. [PMID: 38689233 PMCID: PMC11059926 DOI: 10.1186/s12865-024-00611-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 03/11/2024] [Indexed: 05/02/2024] Open
Abstract
BACKGROUND Bacillus Calmette-Guérin (BCG) vaccination has off-target protective effects against infections unrelated to tuberculosis. Among these, murine and human studies suggest that BCG vaccination may protect against malaria. We investigated whether BCG vaccination influences neonatal in vitro cytokine responses to Plasmodium falciparum. Blood samples were collected from 108 participants in the Melbourne Infant Study BCG for Allergy and Infection Reduction (MIS BAIR) randomised controlled trial (Clinical trials registration NCT01906853, registered July 2013), seven days after randomisation to neonatal BCG (n = 66) or no BCG vaccination (BCG-naïve, n = 42). In vitro cytokine responses were measured following stimulation with P. falciparum-infected erythrocytes (PfIE) or E. coli. RESULTS No difference in the measured cytokines were observed between BCG-vaccinated and BCG-naïve neonates following stimulation with PfIE or E. coli. However, age at which blood was sampled was independently associated with altered cytokine responses to PfIE. Being male was also independently associated with increased TNF-a responses to both PfIE and E. coli. CONCLUSION These findings do not support a role for BCG vaccination in influencing in vitro neonatal cytokine responses to P. falciparum. Older neonates are more likely to develop P. falciparum-induced IFN-γ and IFN-γ-inducible chemokine responses implicated in early protection against malaria and malaria pathogenesis.
Collapse
Affiliation(s)
- N L Messina
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia
- The University of Melbourne, Parkville, Australia
| | - M Wang
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia
- The University of Melbourne, Parkville, Australia
- Tsinghua University, Beijing, China
- Department of Ophthalmology, Peking Union Medical College Hospital, Beijing, China
| | - E K Forbes
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia.
| | - B Freyne
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia
- The University of Melbourne, Parkville, Australia
- The Royal Children's Hospital Melbourne, Parkville, Australia
- Department of Paediatric Infectious Diseases, School of Medicine, Children's Health Ireland at Crumlin, University College Dublin, Dublin, Ireland
| | - W P Hasang
- Department of Infectious Diseases, The Doherty Institute, The University of Melbourne, Melbourne, Australia
| | - S Germano
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia
| | - R Bonnici
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia
| | - F Summons
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia
- The University of Melbourne, Parkville, Australia
- The Royal Children's Hospital Melbourne, Parkville, Australia
| | - K Gardiner
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia
- The Royal Children's Hospital Melbourne, Parkville, Australia
| | - S Donath
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia
- The University of Melbourne, Parkville, Australia
| | - R Gordon
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia
- The University of Melbourne, Parkville, Australia
| | - S J Rogerson
- Department of Infectious Diseases, The Doherty Institute, The University of Melbourne, Melbourne, Australia
- Department of Medicine, The Doherty Institute, The University of Melbourne, Melbourne, Australia
| | - N Curtis
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia
- The University of Melbourne, Parkville, Australia
- The Royal Children's Hospital Melbourne, Parkville, Australia
| |
Collapse
|
2
|
Anand N. Antiparasitic activity of the iron-containing milk protein lactoferrin and its potential derivatives against human intestinal and blood parasites. FRONTIERS IN PARASITOLOGY 2024; 2:1330398. [PMID: 39816822 PMCID: PMC11731944 DOI: 10.3389/fpara.2023.1330398] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 12/30/2023] [Indexed: 01/18/2025]
Abstract
An iron-containing milk protein named lactoferrin (Lf) has demonstrated antiparasitic and immunomodulatory properties against a variety of human parasites. This protein has shown its capability to bind and transport iron molecules in the vicinity of the host-pathogen environment. The ability of parasites to sequester the iron molecule and to increase their pathogenicity and survival depends on the availability of iron sources. Lf protein has suggested a iron chelating effect on parasites iron and, hence, has shown its antiparasitic effect. Since the parasites have a complex life cycle and have developed drug resistance, vaccines and other treatments are a handful. Therefore, therapeutic research focusing on natural treatment regimens that target the parasite and are non-toxic to host cells is urgently needed. The antiparasitic efficacy of Lf protein has been extensively studied over the past 40 years using both in vitro and in vivo studies. This review article highlighted past important studies on Lf protein that revealed its potential antiparasitic activity against various intracellular and extracellular intestinal or blood-borne human parasites. This review article structures the role of Lf protein in its various forms, such as native, peptide, and nanoformulation, laying the groundwork for its function as an antiparasitic agent and its possible known mechanisms of action.
Collapse
Affiliation(s)
- Namrata Anand
- Department of Pharmacy Practice and Science, College of Pharmacy, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
3
|
Rubio-Casillas A, Rodriguez-Quintero CM, Redwan EM, Gupta MN, Uversky VN, Raszek M. Do vaccines increase or decrease susceptibility to diseases other than those they protect against? Vaccine 2024; 42:426-440. [PMID: 38158298 DOI: 10.1016/j.vaccine.2023.12.060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 11/16/2023] [Accepted: 12/20/2023] [Indexed: 01/03/2024]
Abstract
Contrary to the long-held belief that the effects of vaccines are specific for the disease they were created; compelling evidence has demonstrated that vaccines can exert positive or deleterious non-specific effects (NSEs). In this review, we compiled research reports from the last 40 years, which were found based on the PubMed search for the epidemiological and immunological studies on the non-specific effects (NSEs) of the most common human vaccines. Analysis of information showed that live vaccines induce positive NSEs, whereas non-live vaccines induce several negative NSEs, including increased female mortality associated with enhanced susceptibility to other infectious diseases, especially in developing countries. These negative NSEs are determined by the vaccination sequence, the antigen concentration in vaccines, the type of vaccine used (live vs. non-live), and also by repeated vaccination. We do not recommend stopping using non-live vaccines, as they have demonstrated to protect against their target disease, so the suggestion is that their detrimental NSEs can be minimized simply by changing the current vaccination sequence. High IgG4 antibody levels generated in response to repeated inoculation with mRNA COVID-19 vaccines could be associated with a higher mortality rate from unrelated diseases and infections by suppressing the immune system. Since most COVID-19 vaccinated countries are reporting high percentages of excess mortality not directly attributable to deaths from such disease, the NSEs of mRNA vaccines on overall mortality should be studied in depth.
Collapse
Affiliation(s)
- Alberto Rubio-Casillas
- Autlan Regional Hospital, Health Secretariat, Autlan 48900, Jalisco, Mexico; Biology Laboratory, Autlan Regional Preparatory School, University of Guadalajara, Autlan 48900, Jalisco, Mexico.
| | | | - Elrashdy M Redwan
- Biological Science Department, Faculty of Science, King Abdulaziz University, P.O. Box 80203, Jeddah 21589, Saudi Arabia; Therapeutic and Protective Proteins Laboratory, Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City for Scientific Research and Technology Applications, New Borg EL-Arab, Alexandria 21934, Egypt.
| | - Munishwar Nath Gupta
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, Hauz Khas, New Delhi 110016, India.
| | - Vladimir N Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA.
| | - Mikolaj Raszek
- Merogenomics (Genomic Sequencing Consulting), Edmonton, AB T5J 3R8, Canada.
| |
Collapse
|
4
|
de Araujo ACVSC, Mambelli F, Sanches RO, Marinho FV, Oliveira SC. Current Understanding of Bacillus Calmette-Guérin-Mediated Trained Immunity and Its Perspectives for Controlling Intracellular Infections. Pathogens 2023; 12:1386. [PMID: 38133271 PMCID: PMC10745672 DOI: 10.3390/pathogens12121386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 11/20/2023] [Accepted: 11/23/2023] [Indexed: 12/23/2023] Open
Abstract
The bacillus Calmette-Guérin (BCG) is an attenuated bacterium derived from virulent Mycobacterium bovis. It is the only licensed vaccine used for preventing severe forms of tuberculosis in children. Besides its specific effects against tuberculosis, BCG administration is also associated with beneficial non-specific effects (NSEs) following heterologous stimuli in humans and mice. The NSEs from BCG could be related to both adaptive and innate immune responses. The latter is also known as trained immunity (TI), a recently described biological feature of innate cells that enables functional improvement based on metabolic and epigenetic reprogramming. Currently, the mechanisms related to BCG-mediated TI are the focus of intense research, but many gaps are still in need of elucidation. This review discusses the present understanding of TI induced by BCG, exploring signaling pathways that are crucial to a trained phenotype in hematopoietic stem cells and monocytes/macrophages lineage. It focuses on BCG-mediated TI mechanisms, including the metabolic-epigenetic axis and the inflammasome pathway in these cells against intracellular pathogens. Moreover, this study explores the TI in different immune cell types, its ability to protect against various intracellular infections, and the integration of trained innate memory with adaptive memory to shape next-generation vaccines.
Collapse
Affiliation(s)
- Ana Carolina V. S. C. de Araujo
- Departamento de Genética, Ecologia e Evolução, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil;
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-900, SP, Brazil;
| | - Fábio Mambelli
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-900, SP, Brazil;
| | - Rodrigo O. Sanches
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (R.O.S.); (F.V.M.)
| | - Fábio V. Marinho
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (R.O.S.); (F.V.M.)
| | - Sergio C. Oliveira
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-900, SP, Brazil;
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (R.O.S.); (F.V.M.)
| |
Collapse
|
5
|
Zhu J, Liu J, Yan C, Wang D, Pan W. Trained immunity: a cutting edge approach for designing novel vaccines against parasitic diseases? Front Immunol 2023; 14:1252554. [PMID: 37868995 PMCID: PMC10587610 DOI: 10.3389/fimmu.2023.1252554] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 09/25/2023] [Indexed: 10/24/2023] Open
Abstract
The preventive situation of parasitosis, a global public health burden especially for developing countries, is not looking that good. Similar to other infections, vaccines would be the best choice for preventing and controlling parasitic infection. However, ideal antigenic molecules for vaccine development have not been identified so far, resulting from the complicated life history and enormous genomes of the parasites. Furthermore, the suppression or down-regulation of anti-infectious immunity mediated by the parasites or their derived molecules can compromise the effect of parasitic vaccines. Comparing the early immune profiles of several parasites in the permissive and non-permissive hosts, a robust innate immune response is proposed to be a critical event to eliminate the parasites. Therefore, enhancing innate immunity may be essential for designing novel and effective parasitic vaccines. The newly emerging trained immunity (also termed innate immune memory) has been increasingly recognized to provide a novel perspective for vaccine development targeting innate immunity. This article reviews the current status of parasitic vaccines and anti-infectious immunity, as well as the conception, characteristics, and mechanisms of trained immunity and its research progress in Parasitology, highlighting the possible consideration of trained immunity in designing novel vaccines against parasitic diseases.
Collapse
Affiliation(s)
- Jinhang Zhu
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
- The Second Clinical Medical College, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jiaxi Liu
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Chao Yan
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Dahui Wang
- Liangshan College (Li Shui) China, Lishui University, Lishui, Zhejiang, China
| | - Wei Pan
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
6
|
Ferreras-Colino E, Moreno I, Gortázar C, Sevilla I, Agulló-Ros I, Domínguez L, Juste R, Risalde MA, Domínguez M. Oral immunization with heat-inactivated Mycobacterium bovis reduces local parasite dissemination and hepatic granuloma development in mice infected with Leishmania amazonensis. Res Vet Sci 2023; 162:104963. [PMID: 37517297 DOI: 10.1016/j.rvsc.2023.104963] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 07/01/2023] [Accepted: 07/21/2023] [Indexed: 08/01/2023]
Abstract
Aiming to explore whether oral immunization with heat-inactivated Mycobacterium bovis (HIMB) protects mice against Leishmania infection, 18 female BALB/c mice were randomly assigned to the immunized group, that received oral HIMB, or the control group, and were infected by inoculation of 10,000 Leishmania amazonensis promastigotes in the footpad. Spleen culture was positive in 55.55% of immunized mice and in 100% of control mice (p = 0.082). The number of immunolabeled amastigotes number in the popliteal lymph node was lower in the immunized group (p = 0.009). The immunized group presented fewer mature granulomas in the liver (p = 0.005) and more Lys + macrophages (p = 0.002) and fewer CD3+ T lymphocytes (p < 0.001) per hepatic granuloma. We conclude that immunization with HIMB via the oral route limited local parasite dissemination and hepatic granuloma development in mice challenged with Leishmania amazonensis through stimulation of macrophages, which is compatible with trained immunity.
Collapse
Affiliation(s)
- Elisa Ferreras-Colino
- SaBio, Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo 12, 13005 Ciudad Real, Spain
| | - Inmaculada Moreno
- Unidad de Inmunología Microbiana, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Carretera Pozuelo-Majadahonda km 2, 28220 Majadahonda, Madrid, Spain
| | - Christian Gortázar
- SaBio, Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo 12, 13005 Ciudad Real, Spain
| | - Iker Sevilla
- Animal Health Department, NEIKER-Instituto Vasco de Investigación y Desarrollo Agrario, Derio, Bizkaia, Spain
| | - Irene Agulló-Ros
- Departamento de Anatomía y Anatomía Patológica Comparadas y Toxicología, Grupo de Investigación GISAZ, UIC Zoonosis y Enfermedades Emergentes ENZOEM, Universidad de Córdoba, Córdoba, Spain
| | - Lucas Domínguez
- VISAVET Health Surveillance Center, Universidad Complutense de Madrid, Madrid, Spain
| | - Ramón Juste
- Animal Health Department, NEIKER-Instituto Vasco de Investigación y Desarrollo Agrario, Derio, Bizkaia, Spain
| | - Maria A Risalde
- Departamento de Anatomía y Anatomía Patológica Comparadas y Toxicología, Grupo de Investigación GISAZ, UIC Zoonosis y Enfermedades Emergentes ENZOEM, Universidad de Córdoba, Córdoba, Spain; CIBERINFEC, ISCIII - CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain.
| | - Mercedes Domínguez
- Unidad de Inmunología Microbiana, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Carretera Pozuelo-Majadahonda km 2, 28220 Majadahonda, Madrid, Spain
| |
Collapse
|
7
|
Liu X, Xiao C, Guan P, Chen Q, You L, Kong H, Qin W, Dou P, Li Q, Li Y, Jiao Y, Zhong Z, Yang J, Wang X, Wang Q, Zhao J, Xu Z, Zhang H, Li R, Gao P, Xu G. Metabolomics acts as a powerful tool for comprehensively evaluating vaccines approved under emergency: a CoronaVac retrospective study. Front Immunol 2023; 14:1168308. [PMID: 37520533 PMCID: PMC10375237 DOI: 10.3389/fimmu.2023.1168308] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 06/26/2023] [Indexed: 08/01/2023] Open
Abstract
INTRODUCTION To control the COVID-19 pandemic, great efforts have been made to realize herd immunity by vaccination since 2020. Unfortunately, most of the vaccines against COVID-19 were approved in emergency without a full-cycle and comprehensive evaluation process as recommended to the previous vaccines. Metabolome has a close tie with the phenotype and can sensitively reflect the responses to stimuli, rendering metabolomic analysis have the potential to appraise and monitor vaccine effects authentically. METHODS In this study, a retrospective study was carried out for 330 Chinese volunteers receiving recommended two-dose CoronaVac, a vaccine approved in emergency in 2020. Venous blood was sampled before and after vaccination at 5 separate time points for all the recipients. Routine clinical laboratory analysis, metabolomic and lipidomic analysis data were collected. RESULTS AND DISCUSSION It was found that the serum antibody-positive rate of this population was around 81.82%. Most of the laboratory parameters were slightly perturbated within the relevant reference intervals after vaccination. The metabolomic and lipidomic analyses showed that the metabolic shift after inoculation was mainly in the glycolysis, tricarboxylic acid cycle, amino acid metabolism, urea cycle, as well as microbe-related metabolism (bile acid metabolism, tryptophan metabolism and phenylalanine metabolism). Time-course metabolome changes were found in parallel with the progress of immunity establishment and peripheral immune cell counting fluctuation, proving metabolomics analysis was an applicable solution to evaluate immune effects complementary to traditional antibody detection. Taurocholic acid, lysophosphatidylcholine 16:0 sn-1, glutamic acid, and phenylalanine were defined as valuable metabolite markers to indicate the establishment of immunity after vaccination. Integrated with the traditional laboratory analysis, this study provided a feasible metabolomics-based solution to relatively comprehensively evaluate vaccines approved under emergency.
Collapse
Affiliation(s)
- Xinyu Liu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
- Liaoning Province Key Laboratory of Metabolomics, Dalian, China
| | - Congshu Xiao
- Department of Infection, The Second Hospital of Dalian Medical University, Dalian, China
| | - Pengwei Guan
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
- Liaoning Province Key Laboratory of Metabolomics, Dalian, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Qianqian Chen
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
- Liaoning Province Key Laboratory of Metabolomics, Dalian, China
| | - Lei You
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
- Liaoning Province Key Laboratory of Metabolomics, Dalian, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Hongwei Kong
- Hangzhou Health-Bank Medical Laboratory Co., Ltd., Hangzhou, China
| | - Wangshu Qin
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
- Liaoning Province Key Laboratory of Metabolomics, Dalian, China
| | - Peng Dou
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
- Liaoning Province Key Laboratory of Metabolomics, Dalian, China
| | - Qi Li
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
- Liaoning Province Key Laboratory of Metabolomics, Dalian, China
| | - Yanju Li
- Clinical laboratory, Affiliated Dalian Hospital of Shengjing Hospital of Chinese Medical University, Dalian, China
| | - Ying Jiao
- Nursing Department, Anshan Infectious Disease Hospital, Anshan, China
| | - Zhiwei Zhong
- Department of Infection, The Second Hospital of Dalian Medical University, Dalian, China
| | - Jun Yang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
- Liaoning Province Key Laboratory of Metabolomics, Dalian, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xiaolin Wang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
- Liaoning Province Key Laboratory of Metabolomics, Dalian, China
| | - Qingqing Wang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
- Liaoning Province Key Laboratory of Metabolomics, Dalian, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jinhui Zhao
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
- Liaoning Province Key Laboratory of Metabolomics, Dalian, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zhiliang Xu
- Hangzhou Health-Bank Medical Laboratory Co., Ltd., Hangzhou, China
| | - Hong Zhang
- Internal Department, Women and Children’s Hospital of Anshan City, Anshan, China
| | - Rongkuan Li
- Department of Infection, The Second Hospital of Dalian Medical University, Dalian, China
| | - Peng Gao
- Clinical laboratory, The Second Hospital of Dalian Medical University, Dalian, China
- Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Guowang Xu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
- Liaoning Province Key Laboratory of Metabolomics, Dalian, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
8
|
Hu S, Xiang D, Zhang X, Zhang L, Wang S, Jin K, You L, Huang J. The mechanisms and cross-protection of trained innate immunity. Virol J 2022; 19:210. [PMID: 36482472 PMCID: PMC9733056 DOI: 10.1186/s12985-022-01937-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Accepted: 11/28/2022] [Indexed: 12/13/2022] Open
Abstract
In recent years, the traditional cognition of immunological memory being specific to adaptive immunity has been challenged. Innate immunity can mount enhanced responsiveness upon secondary stimulation, and a phenomenon is termed trained innate immunity. Trained innate immunity is orchestrated by distinct metabolic and epigenetic reprogramming in both circulating myeloid cells and myeloid progenitor cells in bone marrow, leading to long-term resistance to related and non-related pathogens infections. The induction of trained innate immunity can also polarize innate immune cells towards a hyperresponsive phenotype in the tumor microenvironment to exert antitumor effects. This review will discuss the current understanding of innate immune memory and the mechanisms during the induction of innate immunity, including signaling pathways, metabolic changes, and epigenetic rewriting. We also provide an overview of cross-protection against infectious diseases and cancers based on trained innate immunity.
Collapse
Affiliation(s)
- Shiwei Hu
- grid.13402.340000 0004 1759 700XDepartment of Hematology, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Yiwu, Zhejiang China ,Zhejiang Provincial Clinical Research Center for Hematological Disorders, Hangzhou, Zhejiang China
| | - Danhong Xiang
- grid.13402.340000 0004 1759 700XDepartment of Hematology, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Yiwu, Zhejiang China ,Zhejiang Provincial Clinical Research Center for Hematological Disorders, Hangzhou, Zhejiang China
| | - Xinlu Zhang
- grid.13402.340000 0004 1759 700XDepartment of Hematology, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Yiwu, Zhejiang China ,Zhejiang Provincial Clinical Research Center for Hematological Disorders, Hangzhou, Zhejiang China
| | - Lan Zhang
- grid.13402.340000 0004 1759 700XDepartment of Hematology, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Yiwu, Zhejiang China ,Zhejiang Provincial Clinical Research Center for Hematological Disorders, Hangzhou, Zhejiang China
| | - Shengjie Wang
- grid.13402.340000 0004 1759 700XDepartment of Hematology, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Yiwu, Zhejiang China ,Zhejiang Provincial Clinical Research Center for Hematological Disorders, Hangzhou, Zhejiang China
| | - Keyi Jin
- grid.13402.340000 0004 1759 700XDepartment of Hematology, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Yiwu, Zhejiang China ,Zhejiang Provincial Clinical Research Center for Hematological Disorders, Hangzhou, Zhejiang China
| | - Liangshun You
- grid.13402.340000 0004 1759 700XDepartment of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang China ,Zhejiang Provincial Clinical Research Center for Hematological Disorders, Hangzhou, Zhejiang China
| | - Jian Huang
- grid.13402.340000 0004 1759 700XDepartment of Hematology, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Yiwu, Zhejiang China ,grid.13402.340000 0004 1759 700XDepartment of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang China ,Zhejiang Provincial Clinical Research Center for Hematological Disorders, Hangzhou, Zhejiang China
| |
Collapse
|
9
|
Reyes-López M, Ramírez-Rico G, Serrano-Luna J, de la Garza M. Activity of Apo-Lactoferrin on Pathogenic Protozoa. Pharmaceutics 2022; 14:pharmaceutics14081702. [PMID: 36015327 PMCID: PMC9414845 DOI: 10.3390/pharmaceutics14081702] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/10/2022] [Accepted: 08/11/2022] [Indexed: 11/16/2022] Open
Abstract
Parasites and other eventually pathogenic organisms require the ability to adapt to different environmental conditions inside the host to assure survival. Some host proteins have evolved as defense constituents, such as lactoferrin (Lf), which is part of the innate immune system. Lf in its iron-free form (apo-Lf) and its peptides obtained by cleavage with pepsin are microbicides. Parasites confront Lf in mucosae and blood. In this work, the activity of Lf against pathogenic and opportunistic parasites such as Cryptosporidium spp., Eimeria spp., Entamoeba histolytica, Giardia duodenalis, Leishmania spp., Trypanosoma spp., Plasmodium spp., Babesia spp., Toxoplasma gondii, Trichomonas spp., and the free-living but opportunistic pathogens Naegleria fowleri and Acanthamoeba castellani were reviewed. The major effects of Lf could be the inhibition produced by sequestering the iron needed for their survival and the production of oxygen-free radicals to more complicated mechanisms, such as the activation of macrophages to phagocytes with the posterior death of those parasites. Due to the great interest in Lf in the fight against pathogens, it is necessary to understand the exact mechanisms used by this protein to affect their virulence factors and to kill them.
Collapse
|
10
|
Juste RA, Ferreras-Colino E, de la Fuente J, Domínguez M, Risalde MA, Domínguez L, Cabezas-Cruz A, Gortázar C. Heat inactivated mycobacteria, alpha-gal and zebra fish: insights gained from experiences with two promising trained immunity inductors and a validated animal model. Immunol Suppl 2022; 167:139-153. [PMID: 35752944 DOI: 10.1111/imm.13529] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 06/22/2022] [Indexed: 11/30/2022]
Abstract
Trained immunity (TRAIM) may be defined as a form of memory where innate immune cells such as monocytes, macrophages, dendritic and natural killer (NK) cells undergo an epigenetic reprogramming that enhances their primary defensive capabilities. Cross-pathogen protective TRAIM can be triggered in different hosts by exposure to live microbes or microbe-derived products such as heat-inactivated Mycobacterium bovis or with the glycan α-Gal to elicit protective responses against several pathogens. We review the TRAIM paradigm using two models representing distinct scales of immune sensitization: the whole bacterial cell and one of its building blocks, the polysaccharides or glycans. Observations point out to macrophage lytic capabilities and cytokine regulation as two key components in nonspecific innate immune responses against infections. The study of the TRAIM response deserves attention to better characterize the evolution of host-pathogen cooperation both for identifying the etiology of some diseases and for finding new therapeutic strategies. In this field, the zebrafish provides a convenient and complete biological system that could help to deepen in the knowledge of TRAIM-mediated mechanisms in pathogen-host interactions. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Ramón A Juste
- Animal Health Department, NEIKER-Instituto Vasco de Investigación y Desarrollo Agrario, Derio, Bizkaia, Spain.,NySA. Servicio Regional de Investigación y Desarrollo Agroalimentario (SERIDA), Villaviciosa, Asturias, Spain
| | - Elisa Ferreras-Colino
- SaBio, Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo 12, Ciudad Real, Spain
| | - José de la Fuente
- SaBio, Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo 12, Ciudad Real, Spain.,Department of Veterinary Pathobiology, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK, USA
| | - Mercedes Domínguez
- Unidad de Inmunología Microbiana, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Carretera Pozuelo-Majadahonda km 2, 28220 Majadahonda, Madrid, Spain
| | - María A Risalde
- Departamento de Anatomía y Anatomía Patológica Comparadas y Toxicología, Facultad de Veterinaria, Universidad de Córdoba (UCO), Córdoba, Spain.,CIBERINFEC, ISCIII - CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Lucas Domínguez
- VISAVET Health Surveillance Centre, Complutense University of Madrid, 28040, Madrid, Spain.,Department of Animal Health, Faculty of Veterinary Medicine, Complutense University of Madrid, Madrid, Spain
| | - Alejandro Cabezas-Cruz
- UMR BIPAR, INRAE, ANSES, Ecole Nationale Vétérinaire d'Alfort, Université Paris-Est, Maisons-Alfort, France
| | - Christian Gortázar
- SaBio, Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo 12, Ciudad Real, Spain
| |
Collapse
|
11
|
Aslan G, Alkaya D. One Hundred of Tuberculosis Vaccine: History of Bacille Calmette-Guérin - Could BCG Vaccination Induce Trained Immunity? TURKISH JOURNAL OF IMMUNOLOGY 2022. [DOI: 10.4274/tji.galenos.2022.98598] [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]
|
12
|
Vaz-Rodrigues R, Ferreras-Colino E, Ugarte-Ruíz M, Pesciaroli M, Thomas J, García-Seco T, Sevilla IA, Pérez-Sancho M, Mateo R, Domínguez L, Gortazar C, Risalde MA. Nonspecific protection of heat-inactivated Mycobacterium bovis against Salmonella Choleraesuis infection in pigs. Vet Res 2022; 53:31. [PMID: 35436975 PMCID: PMC9014587 DOI: 10.1186/s13567-022-01047-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 03/17/2022] [Indexed: 11/10/2022] Open
Abstract
AbstractTrained immunity is the capacity of innate immune cells to produce an improved response against a secondary infection after a previous unrelated infection. Salmonellosis represents a public health issue and affects the pig farming industry. In general, vaccination against salmonellosis is still facing problems regarding the control of distinct serovars. Therefore, we hypothesized that an immunostimulant based on heat inactivated Mycobacterium bovis (HIMB) could have an immune training effect in pigs challenged with Salmonella enterica serovar Choleraesuis (S. Choleraesuis) and decided to explore the amplitude of this non-specific immune response. For this purpose, twenty-four 10 days-old female piglets were randomly separated in three groups: immunized group (n = 10) received orally two doses of HIMB prior to the intratracheal S. Choleraesuis-challenge, positive control group (n = 9) that was only challenged with S. Choleraesuis, and negative control group (n = 5) that was neither immunized nor infected. All individuals were necropsied 21 days post-challenge. HIMB improved weight gain and reduced respiratory symptoms and pulmonary lesions caused by S. Choleraesuis in pigs. Pigs immunized with HIMB showed higher cytokine production, especially of serum TNFα and lung CCL28, an important mediator of mucosal trained immunity. Moreover, immunized pigs showed lower levels of the biomarker of lipid oxidation malondialdehyde and higher activity of the antioxidant enzyme superoxide dismutase than untreated challenged pigs. However, the excretion and tissue colonization of S. Choleraesuis remained unaffected. This proof-of-concept study suggests beneficial clinical, pathological, and heterologous immunological effects against bacterial pathogens within the concept of trained immunity, opening avenues for further research.
Collapse
|
13
|
Baindara P, Agrawal S, Franco OL. Host-directed therapies for malaria and tuberculosis: common infection strategies and repurposed drugs. Expert Rev Anti Infect Ther 2022; 20:849-869. [DOI: 10.1080/14787210.2022.2044794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Piyush Baindara
- Department of Molecular Microbiology & Immunology, School of Medicine, University of Missouri, Missouri, Columbia, MO, USA
| | - Sonali Agrawal
- Immunology Division, ICMR-National JALMA Institute for Leprosy and Other Mycobacterial Diseases, Agra, Uttar Pradesh, India
| | - O. L. Franco
- Proteomics Analysis and Biochemical Center, Catholic University of Brasilia, Brasilia, Brazil; S-Inova Biotech, Catholic University Dom Bosco, Campo Grande, MS, Brazil
| |
Collapse
|
14
|
Tangie E, Walters A, Hsu NJ, Fisher M, Magez S, Jacobs M, Keeton R. BCG-mediated protection against M. tuberculosis is sustained post-malaria infection independent of parasite virulence. Immunology 2021; 165:219-233. [PMID: 34775598 DOI: 10.1111/imm.13431] [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] [Received: 07/24/2021] [Revised: 09/15/2021] [Accepted: 09/28/2021] [Indexed: 11/28/2022] Open
Abstract
Tuberculosis (TB) and malaria remain serious threats to global health. Bacillus Calmette-Guerin (BCG), the only licensed vaccine against TB protects against severe disseminated forms of TB in infants but shows poor efficacy against pulmonary TB in adults. Co-infections have been reported as one of the factors implicated in vaccine inefficacy. Given the geographical overlap of malaria and TB in areas where BCG vaccination is routinely administered, we hypothesized that virulence-dependent co-infection with Plasmodium species could alter the BCG-specific immune responses thus resulting in failure to protect against Mycobacterium tuberculosis. We compared virulent Plasmodium berghei and non-virulent Plasmodium chabaudi, their effects on B cells, effector and memory T cells, and the outcome on BCG-induced efficacy against M. tuberculosis infection. We demonstrate that malaria co-infection modulates both B- and T-cell immune responses but does not significantly alter the ability of the BCG vaccine to inhibit the growth of M. tuberculosis irrespective of parasite virulence. This malaria-driven immune regulation may have serious consequences in the early clinical trials of novel vaccines, which rely on vaccine-specific T-cell responses to screen novel vaccines for progression to the more costly vaccine efficacy trials.
Collapse
Affiliation(s)
- Emily Tangie
- Division of Immunology, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, Health Sciences Faculty, University of Cape Town, Observatory, South Africa
| | - Avril Walters
- Division of Immunology, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, Health Sciences Faculty, University of Cape Town, Observatory, South Africa
| | - Nai-Jen Hsu
- Division of Immunology, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, Health Sciences Faculty, University of Cape Town, Observatory, South Africa
| | - Michelle Fisher
- South African Tuberculosis Vaccine Initiative, University of Cape Town, Observatory, South Africa
| | - Stefan Magez
- Laboratory for Cellular and Molecular Immunology (CMIM), Department of Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium.,Department of Biochemistry and Microbiology, Ghent University, Gent, Belgium.,Laboratory for Biomedical Research, Department of Molecular Biotechnology, Environment Technology and Food Technology, Ghent University Global Campus, Incheon, Korea
| | - Muazzam Jacobs
- Division of Immunology, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, Health Sciences Faculty, University of Cape Town, Observatory, South Africa.,National Health Laboratory Service, Cape Town, South Africa.,Infectious Disease Research Unit, University of Cape Town, Observatory, South Africa
| | - Roanne Keeton
- Division of Immunology, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, Health Sciences Faculty, University of Cape Town, Observatory, South Africa
| |
Collapse
|
15
|
Lobo N, Brooks NA, Zlotta AR, Cirillo JD, Boorjian S, Black PC, Meeks JJ, Bivalacqua TJ, Gontero P, Steinberg GD, McConkey D, Babjuk M, Alfred Witjes J, Kamat AM. 100 years of Bacillus Calmette-Guérin immunotherapy: from cattle to COVID-19. Nat Rev Urol 2021; 18:611-622. [PMID: 34131332 PMCID: PMC8204595 DOI: 10.1038/s41585-021-00481-1] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/18/2021] [Indexed: 02/07/2023]
Abstract
Bacillus Calmette-Guérin (BCG) is the most widely used vaccine worldwide and has been used to prevent tuberculosis for a century. BCG also stimulates an anti-tumour immune response, which urologists have harnessed for the treatment of non-muscle-invasive bladder cancer. A growing body of evidence indicates that BCG offers protection against various non-mycobacterial and viral infections. The non-specific effects of BCG occur via the induction of trained immunity and form the basis for the hypothesis that BCG vaccination could be used to protect against the severity of coronavirus disease 2019 (COVID-19). This Perspective article highlights key milestones in the 100-year history of BCG and projects its potential role in the COVID-19 pandemic.
Collapse
Affiliation(s)
- Niyati Lobo
- Department of Urology, MD Anderson Cancer Center, Houston, TX, USA
| | - Nathan A Brooks
- Department of Urology, MD Anderson Cancer Center, Houston, TX, USA
| | - Alexandre R Zlotta
- Division of Urology, Department of Surgery, Sinai Health System, University of Toronto, Toronto, ON, Canada
- Department of Surgical Oncology, Division of Urology, Princess Margaret Cancer Centre, University of Toronto and University Health Network, Toronto, ON, Canada
| | - Jeffrey D Cirillo
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, TX, USA
| | | | | | - Joshua J Meeks
- Northwestern University School of Medicine, Chicago, IL, USA
| | - Trinity J Bivalacqua
- The James Buchanan Brady Urological Institute and Department of Urology, The Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Paolo Gontero
- Division of Urology, Molinette Hospital, University of Torino School of Medicine, Torino, Italy
| | | | - David McConkey
- Johns Hopkins Greenberg Bladder Cancer Institute, Brady Urological Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Marko Babjuk
- Department of Urology, Hospital Motol, Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - J Alfred Witjes
- Department of Urology, Radboud University Nijmegen Medical Centre, Nijmegen, Netherlands
| | - Ashish M Kamat
- Department of Urology, MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
16
|
BCG Provides Short-Term Protection from Experimental Cerebral Malaria in Mice. Vaccines (Basel) 2020; 8:vaccines8040745. [PMID: 33316929 PMCID: PMC7768457 DOI: 10.3390/vaccines8040745] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/05/2020] [Accepted: 12/07/2020] [Indexed: 11/17/2022] Open
Abstract
Clinical and experimental evidence suggests that the tuberculosis vaccine BCG offers protection against unrelated pathogens including the malaria parasite. Cerebral malaria (CM) is the most severe complication associated with Plasmodium falciparum infection in humans and is responsible for most of the fatalities attributed to malaria. We investigated whether BCG protected C57BL/6 mice from P. berghei ANKA (PbA)-induced experimental CM (ECM). The majority of PbA-infected mice that were immunized with BCG showed prolonged survival without developing clinical symptoms of ECM. However, this protective effect waned over time and was associated with the recovery of viable BCG from liver and spleen. Intriguingly, BCG-mediated protection from ECM was not associated with a reduction in parasite burden, indicating that BCG immunization did not improve anti-parasite effector mechanisms. Instead, we found a significant reduction in pro-inflammatory mediators and CD8+ T cells in brains of BCG-vaccinated mice. Together these data suggest that brain recruitment of immune cells involved in the pathogenesis of ECM decreased after BCG vaccination. Understanding the mechanisms underlying the protective effects of BCG on PbA-induced ECM can provide a rationale for developing effective adjunctive therapies to reduce the risk of death and brain damage in CM.
Collapse
|
17
|
Cryptosporidium parvum Subverts Antimicrobial Activity of CRAMP by Reducing Its Expression in Neonatal Mice. Microorganisms 2020; 8:microorganisms8111635. [PMID: 33113928 PMCID: PMC7690728 DOI: 10.3390/microorganisms8111635] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 10/18/2020] [Accepted: 10/18/2020] [Indexed: 12/14/2022] Open
Abstract
Cryptosporidium parvum causes diarrhea in infants under 5 years, in immunosuppressed individuals or in young ruminants. This parasite infects the apical side of ileal epithelial cells where it develops itself and induces inflammation. Antimicrobial peptides (AMPs) are part of the innate immune response, playing a major role in the control of the acute phase of C. parvum infection in neonates. Intestinal AMP production in neonates is characterized by high expressions of Cathelicidin Related Antimicrobial Peptide (CRAMP), the unique cathelicidin in mice known to fight bacterial infections. In this study, we investigated the role of CRAMP during cryptosporidiosis in neonates. We demonstrated that sporozoites are sensitive to CRAMP antimicrobial activity. However, during C. parvum infection the intestinal expression of CRAMP was significantly and selectively reduced, while other AMPs were upregulated. Moreover, despite high CRAMP expression in the intestine of neonates at homeostasis, the depletion of CRAMP did not worsen C. parvum infection. This result might be explained by the rapid downregulation of CRAMP induced by infection. However, the exogenous administration of CRAMP dampened the parasite burden in neonates. Taken together these results suggest that C. parvum impairs the production of CRAMP to subvert the host response, and highlight exogenous cathelicidin supplements as a potential treatment strategy.
Collapse
|
18
|
Lee SH, Chu KB, Kang HJ, Basak S, Kim MJ, Park H, Jin H, Moon EK, Quan FS. Virus-like particles expressing Plasmodium berghei MSP-8 induce protection against P. berghei infection. Parasite Immunol 2020; 42:e12781. [PMID: 32738150 DOI: 10.1111/pim.12781] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 07/22/2020] [Accepted: 07/22/2020] [Indexed: 11/29/2022]
Abstract
AIMS Merozoite surface protein 8 (MSP-8) of Plasmodium parasites plays an important role in erythrocyte invasion and is a potential malaria vaccine candidate. METHODS AND RESULTS In this study, virus-like particles (VLPs) expressing MSP-8 of Plasmodium berghei on the surface of influenza virus matrix protein 1 (M1) core protein were generated for vaccine efficacy assessment. Mice were intramuscularly (IM) immunized with MSP-8 VLPs twice and challenge-infected with P. berghei. We found that VLP vaccination elicited higher levels of P. berghei-specific IgG antibody response in the sera, along with blood CD4+ and CD8+ T-cell response enhancement compared to the naïve control mice. CD4+ and CD8+ effector memory T-cell and memory B-cell responses in the spleen were found to be higher in VLP-immunized mice compared to control mice. VLP vaccination significantly reduced inflammatory cytokine (IFN-γ) response in the spleen and parasitemia levels in blood compared to naïve control mice. CONCLUSIONS These results indicate that MSP-8 containing virus-like particles could be a vaccine candidate for blood-stage vaccine design.
Collapse
Affiliation(s)
- Su-Hwa Lee
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Korea
| | - Ki-Back Chu
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Korea
| | - Hae-Ji Kang
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Korea
| | - Swarnendu Basak
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Korea
| | - Min-Ju Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Korea
| | | | - Hui Jin
- Health Park Co., Ltd., Seoul, Korea
| | - Eun-Kyung Moon
- Department of Medical Zoology, Kyung Hee University School of Medicine, Seoul, Korea
| | - Fu-Shi Quan
- Department of Medical Zoology, Kyung Hee University School of Medicine, Seoul, Korea.,Department of Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, School of Medicine, Graduate school, Kyung Hee University, Seoul, Korea
| |
Collapse
|
19
|
Johnson BS, Laloraya M. A cytokine super cyclone in COVID-19 patients with risk factors: the therapeutic potential of BCG immunization. Cytokine Growth Factor Rev 2020; 54:32-42. [PMID: 32747157 PMCID: PMC7328575 DOI: 10.1016/j.cytogfr.2020.06.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/24/2020] [Accepted: 06/26/2020] [Indexed: 12/17/2022]
Abstract
The seventh human coronavirus SARS-CoV2 belongs to the cluster of extremely pathogenic coronaviruses including SARS-CoV and MERS-CoV, which can cause fatal lower respiratory tract infection. Likewise, SARS-CoV2 infection can be fatal as the disease advances to pneumonia, followed by acute respiratory distress syndrome (ARDS). The development of lethal clinical symptons is associated with an exaggerated production of inflammatory cytokines, referred to as the cytokine storm, is a consequence of a hyperactivated immune response aginst the infection. In this article, we discuss the pathogenic consequences of the cytokine storm and its relationship with COVID-19 associated risk factors. The increased pro-inflammatory immune status in patients with risk factors (diabetes, hypertension, cardiovascular disease, COPD) exacerbates the Cytokine-storm of COVID-19 into a 'Cytokine Super Cyclone'. We also evaluate the antiviral immune responses provided by BCG vaccination and the potential role of 'trained immunity' in early protection against SARS-CoV2.
Collapse
Affiliation(s)
- Betcy Susan Johnson
- Female Reproduction and Metabolic Syndromes Laboratory, Division of Molecular Reproduction, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, 695 014, Kerala, India; Research Centre Kerala University, Senate House Campus, Palayam, Thiruvananthapuram, India
| | - Malini Laloraya
- Female Reproduction and Metabolic Syndromes Laboratory, Division of Molecular Reproduction, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, 695 014, Kerala, India.
| |
Collapse
|
20
|
Jensen KJ, Biering-Sørensen S, Ursing J, Kofoed PEL, Aaby P, Benn CS. Seasonal variation in the non-specific effects of BCG vaccination on neonatal mortality: three randomised controlled trials in Guinea-Bissau. BMJ Glob Health 2020; 5:e001873. [PMID: 32201619 PMCID: PMC7059430 DOI: 10.1136/bmjgh-2019-001873] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Revised: 09/11/2019] [Accepted: 09/15/2019] [Indexed: 11/23/2022] Open
Abstract
The BCG vaccine protects non-specifically against other diseases than tuberculosis. Three randomised controlled trials of early BCG in Guinea-Bissau found a 38% reduction in all-cause neonatal mortality. Little is known about the underlying mechanisms. In Guinea-Bissau, prevalent infectious diseases display distinct seasonality. Revisiting the three trials (>6500 infants) comparing early BCG versus no early BCG in low weight infants on all-cause neonatal mortality over 12 consecutive years, we explored the seasonal variation in BCG’s effect on mortality. In a subgroup of participants, adaptive and innate cytokine responses were measured 4 weeks after randomisation. Consistently over the course of the three trials and 12 years, the effect of BCG on all-cause neonatal mortality was particularly beneficial when administered in November to January, coincident with peaking malaria infections. During these months, BCG was also associated with stronger proinflammatory responses to heterologous challenge. Recent studies have suggested a protective effect of BCG against malaria. BCG may also ameliorate immune-compromising fatal effects of placental malaria in the newborn.
Collapse
Affiliation(s)
- Kristoffer Jarlov Jensen
- Bandim Health Project, University of Southern Denmark, Copenhagen, Denmark.,Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | | | - Johan Ursing
- Department of Infectious Diseases, Danderyd University Hospital, Stockholm, Sweden.,Department of Clinical Sciences, Karolinska Institute, Stockholm, Sweden
| | - Poul-Erik Lund Kofoed
- Department of Pediatrics, Kolding Hospital, Kolding, Denmark.,Bandim Health Project, INDEPTH Network, Bissau, Guinea-Bissau
| | - Peter Aaby
- Bandim Health Project, University of Southern Denmark, Copenhagen, Denmark.,Bandim Health Project, INDEPTH Network, Bissau, Guinea-Bissau
| | - Christine Stabell Benn
- Bandim Health Project, University of Southern Denmark, Copenhagen, Denmark.,OPEN, Institute of Clinical Research, University of Southern Denmark, Odense, Syddanmark, Denmark
| |
Collapse
|
21
|
The science of vaccine safety: Summary of meeting at Wellcome Trust. Vaccine 2020; 38:1869-1880. [PMID: 31987690 DOI: 10.1016/j.vaccine.2020.01.024] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 12/18/2019] [Accepted: 01/07/2020] [Indexed: 12/11/2022]
Abstract
Vaccines are everywhere hugely successful but are also under attack. The reason for the latter is the perception by some people that vaccines are unsafe. However that may be, vaccine safety, life any other scientific subject, must be constantly studied. It was from this point of view that a meeting was organized at the Wellcome Trust in London in May 2019 to assess some aspects of vaccine safety as subjects for scientific study. The objective of the meeting was to assess what is known beyond reasonable doubt and conversely what areas need additional studies. Although the meeting could not cover all aspects of vaccine safety science, many of the most important issues were addressed by a group of about 30 experts to determine what is already known and what additional studies are merited to assess the safety of the vaccines currently in use. The meeting began with reviews of the current situation in different parts of the world, followed by reviews of specific controversial areas, including the incidence of certain conditions after vaccination and the safety of certain vaccine components. Lastly, information about the human papillomavirus vaccine was considered because its safety has been particularly challenged by vaccine opponents. The following is a summary of the meeting findings. In addition to this summary, the meeting organizers will explore opportunities to perform studies that would enlarge knowledge of vaccine safety.
Collapse
|
22
|
Usher NT, Chang S, Howard RS, Martinez A, Harrison LH, Santosham M, Aronson NE. Association of BCG Vaccination in Childhood With Subsequent Cancer Diagnoses: A 60-Year Follow-up of a Clinical Trial. JAMA Netw Open 2019; 2:e1912014. [PMID: 31553471 PMCID: PMC6763973 DOI: 10.1001/jamanetworkopen.2019.12014] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
IMPORTANCE The BCG vaccine is currently the only approved tuberculosis vaccine and is widely administered worldwide, usually during infancy. Previous studies found increased rates of lymphoma and leukemia in BCG-vaccinated populations. OBJECTIVE To determine whether BCG vaccination was associated with cancer rates in a secondary analysis of a BCG vaccine trial. DESIGN, SETTING, AND PARTICIPANTS Retrospective review (60-year follow-up) of a clinical trial in which participants were assigned to the vaccine group by systematic stratification by school district, age, and sex, then randomized by alternation. The original study was conducted at 9 sites in 5 US states between December 1935 and December 1998. Participants were 2963 American Indian and Alaska Native schoolchildren younger than 20 years with no evidence of previous tuberculosis infection. Statistical analysis was conducted between August 2018 and July 2019. INTERVENTIONS Single intradermal injection of either BCG vaccine or saline placebo. MAIN OUTCOMES AND MEASURES The primary outcome was diagnosis of cancer after BCG vaccination. Data on participant interval health and risk factors, including smoking, tuberculosis infection, isoniazid use, and other basic demographic information, were also collected. RESULTS A total of 2963 participants, including 1540 in the BCG vaccine group and 1423 in the placebo group, remained after exclusions. Vaccination occurred at a median (interquartile range) age of 8 (5-11) years; 805 participants (52%) in the BCG group and 710 (50%) in the placebo group were female. At the time of follow-up, 97 participants (7%) in the placebo group and 106 participants (7%) in the BCG vaccine group could not be located; total mortality was 633 participants (44%) in the placebo group and 632 participants (41%) in the BCG group. The overall rate of cancer diagnosis was not significantly different in BCG vaccine vs placebo recipients (hazard ratio, 0.82; 95% CI, 0.66-1.02), including for lymphoma and leukemia. The rate of lung cancer was significantly lower in BCG vs placebo recipients (18.2 vs 45.4 cases per 100 000 person-years; hazard ratio, 0.38; 95% CI, 0.20-0.74; P = .005), controlling for sex, region, alcohol overuse, smoking, and tuberculosis. CONCLUSIONS AND RELEVANCE Childhood BCG vaccination was associated with a lower risk of lung cancer development in American Indian and Alaska Native populations. This finding has potentially important health implications given the high mortality rate associated with lung cancer and the availability of low-cost BCG vaccines.
Collapse
Affiliation(s)
- Nicholas T. Usher
- Infectious Diseases Division, Uniformed Services University of the Health Sciences, Bethesda, Maryland
- College of Agriculture and Life Sciences, Cornell University, Ithaca, New York
| | - Suyoung Chang
- Division of Vaccines and Related Product Applications, US Food and Drug Administration, Silver Spring, Maryland
| | - Robin S. Howard
- Department of Research Programs, Walter Reed National Military Medical Center, Bethesda, Maryland
| | - Adriana Martinez
- Department of Research Programs, Walter Reed National Military Medical Center, Bethesda, Maryland
| | - Lee H. Harrison
- Infectious Diseases Epidemiology Research Unit, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Mathuram Santosham
- Health Systems Program, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
- Center for American Indian Health, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
| | - Naomi E. Aronson
- Infectious Diseases Division, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| |
Collapse
|
23
|
Kanté AM, Exavery A, Jackson EF, Kassimu T, Baynes CD, Hingora A, Phillips JF. The impact of paid community health worker deployment on child survival: the connect randomized cluster trial in rural Tanzania. BMC Health Serv Res 2019; 19:492. [PMID: 31311521 PMCID: PMC6636132 DOI: 10.1186/s12913-019-4203-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 05/30/2019] [Indexed: 11/21/2022] Open
Abstract
Background This paper reports on a rigorously designed non-masked randomized cluster trial of the childhood survival impact of deploying paid community health workers to provide doorstep preventive, promotional, and curative antenatal, newborn, child, and reproductive health care in three rural Tanzanian districts. Methods From August, 2011 to June 2015 ongoing demographic surveillance on 380,000 individuals permitted monitoring of neonatal, infant and under-5 mortality rates for 50 randomly selected intervention and 51 comparison villages. Over the initial 2 years of the project, logistics and supply support systems were managed by the Ifakara Health Institute. In 2013, the experiment transitioned its operational design to logistical support managed by the Ministry of Health and Social Welfare with the goal of enhancing government operational ownership and utilization of results for policy. Results The baseline under 5 mortality rate was 81.3 deaths per 1000 live births with a 95% confidence interval (CI) of 77.2–85.6 in the intervention group and 82.7/1000 (95% CI 78.5–87.1) in the comparison group yielding an adjusted hazard ratio (HR) of 0.99 (95% CI 0.88–1.11, p = 0.867). After 4 years of implementation, the under 5 mortality rate was 73.2/1000 (95% CI 69.3–77.3) in the intervention group and 77.4/1000 (95% CI 73.8–81.1) in the comparison group (adjusted HR 0.95 [95% CI 0.86–1.07], p = 0.443). The intervention had no impact on neonatal mortality in either the first 2 years (HR 1.10 [95% CI 0.89–1.36], p = .392) or last 2 years of implementation (HR 0.98 [95% CI 0.74–1.30], p = .902). Although community health worker deployment significantly reduced mortality among children aged 1–59 months during the first 2 years of implementation (HR 0.85 [95% CI 0.76–0.96], p = 0.008), mortality among post neonates was the same in both groups in years three and four (HR 1.03 [95% CI 0.85–1.24], p = 0.772). Results adjusted for stock-out effects show that diminishing impact was associated with logistics system lapses that constrained worker access to essential drugs and increased post-neonatal mortality risk in the final two project years (HR 1.42 [95% CI 1·07–1·88], p = 0·015). Conclusions Community health worker home-visit deployment had a null effect among neonates, and 2 years of initial impact among children over 1 month of age, but a null effect when tests were based on over 1 month of age data merged for all four project years. The atrophy of under age five effects arose because workers were not continuously equipped with essential medicines in years three and four. Analyses that controlled for stock-out effects suggest that adequately supplied workers had survival effects on children aged 1 to 59 months. Trial registration Registration for trial number ISRCTN96819844 was retrospectively completed on June 21, 2012.
Collapse
Affiliation(s)
- Almamy M Kanté
- Department of International Health, Division of Global Disease Epidemiology and Control, Institute for International Programs, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Amon Exavery
- Ifakara Health Institute, PO Box 78373, Mikocheni, Dar-es-Salaam, Tanzania
| | - Elizabeth F Jackson
- Heilbrunn Department of Population and Family Health, Mailman School of Public Health, Columbia University, 60 Haven Avenue, New York City, NY, 10032, USA
| | - Tani Kassimu
- Ifakara Health Institute, PO Box 78373, Mikocheni, Dar-es-Salaam, Tanzania
| | - Colin D Baynes
- Heilbrunn Department of Population and Family Health, Mailman School of Public Health, Columbia University, 60 Haven Avenue, New York City, NY, 10032, USA
| | - Ahmed Hingora
- Ifakara Health Institute, PO Box 78373, Mikocheni, Dar-es-Salaam, Tanzania
| | - James F Phillips
- Heilbrunn Department of Population and Family Health, Mailman School of Public Health, Columbia University, 60 Haven Avenue, New York City, NY, 10032, USA.
| |
Collapse
|
24
|
In Vitro and In Vivo Antimalarial Activity of LZ1, a Peptide Derived from Snake Cathelicidin. Toxins (Basel) 2019; 11:toxins11070379. [PMID: 31262018 PMCID: PMC6669622 DOI: 10.3390/toxins11070379] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 06/27/2019] [Accepted: 06/28/2019] [Indexed: 01/19/2023] Open
Abstract
Antimalarial drug resistance is an enormous global threat. Recently, antimicrobial peptides (AMPs) are emerging as a new source of antimalarials. In this study, an AMP LZ1 derived from snake cathelicidin was identified with antimalarial activity. In the in vitro antiplasmodial assay, LZ1 showed strong suppression of blood stage Plasmodium falciparum (P. falciparum) with an IC50 value of 3.045 μM. In the in vivo antiplasmodial assay, LZ1 exerted a significant antimalarial activity against Plasmodium berghei (P. berghei) in a dose- and a time- dependent manner. In addition, LZ1 exhibited anti-inflammatory effects and attenuated liver-function impairment during P. berghei infection. Furthermore, by employing inhibitors against glycolysis and oxidative phosphorylation in erythrocytes, LZ1 specifically inhibited adenosine triphosphate (ATP) production in parasite-infected erythrocyte by selectively inhibiting the pyruvate kinase activity. In conclusion, the present study demonstrates that LZ1 is a potential candidate for novel antimalarials development.
Collapse
|
25
|
Dos Santos JC, Vilela Teodoro Silva M, Ribeiro-Dias F, Joosten LAB. Non-specific effects of BCG in protozoal infections: tegumentary leishmaniasis and malaria. Clin Microbiol Infect 2019; 25:1479-1483. [PMID: 31212075 DOI: 10.1016/j.cmi.2019.06.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 05/31/2019] [Accepted: 06/06/2019] [Indexed: 01/01/2023]
Abstract
BACKGROUND Leishmaniasis and malaria are major causes of illness in the poorest countries. In the absence of efficient strategies to prevent infections and to control the transmission of the parasites by insect vectors, treatment relies on drug therapy. Vaccine development continues on several fronts; however none of the candidates developed has so far been shown to provide long-lasting protection at a population level. Because the bacillus Calmette-Guérin (BCG) vaccine can induce heterologous protective effects, we hypothesize that BCG has beneficial effects in the control of tegumentary leishmaniasis (TL) and malaria. AIMS In this review we describe evidence for the protective efficacy of BCG against tegumentary leishmaniasis and malaria in humans. SOURCES Relevant data from peer-reviewed scientific literature published on Pubmed up to January 2019 were examined. CONTENT From experimental animal and various human studies with BCG and one recent randomized malaria trial there is evidence that BCG has beneficial effects in Leishmania spp. and Plasmodium falciparum infections. Although the precise mechanisms remain unknown, BCG can activate innate immune responses, and an increasing body of evidence demonstrates that the induction of trained innate immunity could explain its non-specific protective effects. IMPLICATIONS Despite many years of research to prevent and treat TL and malaria, these diseases remain a public health problem in tropical countries. Future studies are required to examine if BCG vaccination could be used as an effective treatment option.
Collapse
Affiliation(s)
- J C Dos Santos
- Department of Internal Medicine and Radboud Centre of Infectious Diseases (RCI), Radboud University Medical Centre, Nijmegen, the Netherlands; Laboratório de Imunidade Natural (LIN), Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Goiás, Brazil.
| | - M Vilela Teodoro Silva
- Laboratório de Imunidade Natural (LIN), Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Goiás, Brazil
| | - F Ribeiro-Dias
- Laboratório de Imunidade Natural (LIN), Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Goiás, Brazil
| | - L A B Joosten
- Department of Internal Medicine and Radboud Centre of Infectious Diseases (RCI), Radboud University Medical Centre, Nijmegen, the Netherlands
| |
Collapse
|
26
|
Guerra-Maupome M, Vang DX, McGill JL. Aerosol vaccination with Bacille Calmette-Guerin induces a trained innate immune phenotype in calves. PLoS One 2019; 14:e0212751. [PMID: 30794653 PMCID: PMC6386280 DOI: 10.1371/journal.pone.0212751] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 02/10/2019] [Indexed: 01/26/2023] Open
Abstract
Mycobacterium bovis Bacillus Calmette-Guérin (BCG) is a live attenuated vaccine for use against tuberculosis (TB); however, it is known to reduce childhood mortality from infections other than TB. The unspecific protection induced by BCG vaccination has been associated with the induction of memory-like traits of the innate immune system identified as ‘trained’ immunity. In humans and mouse models, in vitro and in vivo BCG training leads to enhanced production of monocyte-derived proinflammatory cytokines in response to secondary unrelated bacterial and fungal pathogens. While BCG has been studied extensively for its ability to induce innate training in humans and mouse models, BCG’s nonspecific protective effects have not been defined in agricultural species. Here, we show that in vitro BCG training induces a functional change in bovine monocytes, characterized by increased transcription of proinflammatory cytokines upon restimulation with the toll-like receptor agonists. Importantly, in vivo, aerosol BCG vaccination in young calves also induced a ‘trained’ phenotype in circulating peripheral blood mononuclear cells (PBMCs), that lead to a significantly enhanced TLR-induced proinflammatory cytokine response and changes in cellular metabolism compared to PBMCs from unvaccinated control calves. Similar to the long-term training effects of BCG reported in humans, our results suggest that in young calves, the effects of BCG induced innate training can last for at least 3 months in circulating immune populations. Interestingly, however, aerosol BCG vaccination did not ‘train’ the innate immune response at the mucosal level, as alveolar macrophages from aerosol BCG vaccinated calves did not mount an enhanced inflammatory response to secondary stimulation, compared to cells isolated from control calves. Together, our results suggest that, like mice and humans, the innate immune system of calves can be ‘trained’; and that BCG vaccination could be used as an immunomodulatory strategy to reduce disease burden in juvenile food animals before the adaptive immune system has fully matured.
Collapse
Affiliation(s)
- Mariana Guerra-Maupome
- Department of Veterinary Microbiology and Preventative Medicine, Iowa State University, Ames, Iowa, United States of America
| | - Dua X. Vang
- Interdepartmental Microbiology Program, Iowa State University, Ames, Iowa, United States of America
| | - Jodi L. McGill
- Department of Veterinary Microbiology and Preventative Medicine, Iowa State University, Ames, Iowa, United States of America
- * E-mail:
| |
Collapse
|
27
|
Messina NL, Zimmermann P, Curtis N. The impact of vaccines on heterologous adaptive immunity. Clin Microbiol Infect 2019; 25:1484-1493. [PMID: 30797062 DOI: 10.1016/j.cmi.2019.02.016] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 02/11/2019] [Accepted: 02/13/2019] [Indexed: 11/26/2022]
Abstract
BACKGROUND Vaccines induce antigen-specific memory in adaptive immune cells that enables long-lived protection against the target pathogen. In addition to this, several vaccines have beneficial effects greater than protection against their target pathogen. These non-specific effects are proposed to be the result of vaccine-induced immunomodulation. In the case of bacille Calmette-Guérin (BCG) vaccine, this involves induction of innate immune memory, termed 'trained immunity', in monocytes and natural killer cells. OBJECTIVES This review discusses current evidence for vaccine-induced immunomodulation of adaptive immune cells and heterologous adaptive immune responses. CONTENT The three vaccines that have been associated with changes in all-cause infant mortality: BCG, diphtheria-tetanus-pertussis (DTP) and measles-containing vaccines (MCV) alter T-cell and B-cell immunity. The majority of studies that investigated non-specific effects of these vaccines on the adaptive immune system report changes in numbers or proportions of adaptive immune cell populations. However, there is also evidence for effects of these vaccines on adaptive immune cell function and responses to heterologous stimuli. There is some evidence that, in addition to BCG, DTP and MCV, other vaccines (that have not been associated with changes in all-cause mortality) may alter adaptive immune responses to unrelated stimuli. IMPLICATIONS This review concludes that vaccines alter adaptive immune cell populations and heterologous immune responses. The non-specific effects differ between various vaccines and their effects on heterologous adaptive immune responses may also involve bystander activation, cross-reactivity and other as yet undefined mechanisms. This has major implications for future vaccine design and vaccination scheduling.
Collapse
Affiliation(s)
- N L Messina
- Infectious Diseases Research Group, Murdoch Children's Research Institute, Parkville, VIC, Australia; Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia.
| | - P Zimmermann
- Infectious Diseases Research Group, Murdoch Children's Research Institute, Parkville, VIC, Australia; Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia; Infectious Diseases Unit, The Royal Children's Hospital Melbourne, Parkville, VIC, Australia; Department of Paediatrics, Fribourg Hospital HFR, Fribourg, Switzerland
| | - N Curtis
- Infectious Diseases Research Group, Murdoch Children's Research Institute, Parkville, VIC, Australia; Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia; Infectious Diseases Unit, The Royal Children's Hospital Melbourne, Parkville, VIC, Australia
| |
Collapse
|
28
|
Walk J, de Bree LCJ, Graumans W, Stoter R, van Gemert GJ, van de Vegte-Bolmer M, Teelen K, Hermsen CC, Arts RJW, Behet MC, Keramati F, Moorlag SJCFM, Yang ASP, van Crevel R, Aaby P, de Mast Q, van der Ven AJAM, Stabell Benn C, Netea MG, Sauerwein RW. Outcomes of controlled human malaria infection after BCG vaccination. Nat Commun 2019; 10:874. [PMID: 30787276 PMCID: PMC6382772 DOI: 10.1038/s41467-019-08659-3] [Citation(s) in RCA: 162] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 01/20/2019] [Indexed: 12/05/2022] Open
Abstract
Recent evidence suggests that certain vaccines, including Bacillus-Calmette Guérin (BCG), can induce changes in the innate immune system with non-specific memory characteristics, termed ‘trained immunity’. Here we present the results of a randomised, controlled phase 1 clinical trial in 20 healthy male and female volunteers to evaluate the induction of immunity and protective efficacy of the anti-tuberculosis BCG vaccine against a controlled human malaria infection. After malaria challenge infection, BCG vaccinated volunteers present with earlier and more severe clinical adverse events, and have significantly earlier expression of NK cell activation markers and a trend towards earlier phenotypic monocyte activation. Furthermore, parasitemia in BCG vaccinated volunteers is inversely correlated with increased phenotypic NK cell and monocyte activation. The combined data demonstrate that BCG vaccination alters the clinical and immunological response to malaria, and form an impetus to further explore its potential in strategies for clinical malaria vaccine development. Immune activation induces long-term alterations of setpoints, impacting responses to subsequent unrelated stimuli. Here the authors show that volunteers vaccinated with BCG respond to controlled human malaria infection with increased clinical symptoms and an inverse correlation between immune activation markers and parasitemia.
Collapse
Affiliation(s)
- Jona Walk
- Department of Medical Microbiology, Radboud University Medical Center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands.,Radboud Center for Infectious Diseases, Radboud University Medical Center PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - L Charlotte J de Bree
- Radboud Center for Infectious Diseases, Radboud University Medical Center PO Box 9101, 6500 HB, Nijmegen, The Netherlands.,Department of Internal Medicine, Radboud University Medical Center PO Box 9101, 6500 HB, Nijmegen, The Netherlands.,Research Center for Vitamins and Vaccines, Bandim Health Project, Statens Serum Institut, 2300, Copenhagen, Denmark.,Odense Patient Data Explorative Network, University of Southern Denmark/Odense University Hospital, 5000, Odense, Denmark
| | - Wouter Graumans
- Department of Medical Microbiology, Radboud University Medical Center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands.,Radboud Center for Infectious Diseases, Radboud University Medical Center PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Rianne Stoter
- Department of Medical Microbiology, Radboud University Medical Center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands.,Radboud Center for Infectious Diseases, Radboud University Medical Center PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Geert-Jan van Gemert
- Department of Medical Microbiology, Radboud University Medical Center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands.,Radboud Center for Infectious Diseases, Radboud University Medical Center PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Marga van de Vegte-Bolmer
- Department of Medical Microbiology, Radboud University Medical Center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands.,Radboud Center for Infectious Diseases, Radboud University Medical Center PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Karina Teelen
- Department of Medical Microbiology, Radboud University Medical Center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands.,Radboud Center for Infectious Diseases, Radboud University Medical Center PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Cornelus C Hermsen
- Department of Medical Microbiology, Radboud University Medical Center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands.,Radboud Center for Infectious Diseases, Radboud University Medical Center PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Rob J W Arts
- Radboud Center for Infectious Diseases, Radboud University Medical Center PO Box 9101, 6500 HB, Nijmegen, The Netherlands.,Department of Internal Medicine, Radboud University Medical Center PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Marije C Behet
- Department of Medical Microbiology, Radboud University Medical Center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands.,Radboud Center for Infectious Diseases, Radboud University Medical Center PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Farid Keramati
- Department of Molecular Biology, Faculty of Science, Radboud university, 6525 GA, Nijmegen, The Netherlands
| | - Simone J C F M Moorlag
- Radboud Center for Infectious Diseases, Radboud University Medical Center PO Box 9101, 6500 HB, Nijmegen, The Netherlands.,Department of Internal Medicine, Radboud University Medical Center PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Annie S P Yang
- Department of Medical Microbiology, Radboud University Medical Center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands.,Radboud Center for Infectious Diseases, Radboud University Medical Center PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Reinout van Crevel
- Radboud Center for Infectious Diseases, Radboud University Medical Center PO Box 9101, 6500 HB, Nijmegen, The Netherlands.,Department of Internal Medicine, Radboud University Medical Center PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Peter Aaby
- Research Center for Vitamins and Vaccines, Bandim Health Project, Statens Serum Institut, 2300, Copenhagen, Denmark
| | - Quirijn de Mast
- Radboud Center for Infectious Diseases, Radboud University Medical Center PO Box 9101, 6500 HB, Nijmegen, The Netherlands.,Department of Internal Medicine, Radboud University Medical Center PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - André J A M van der Ven
- Radboud Center for Infectious Diseases, Radboud University Medical Center PO Box 9101, 6500 HB, Nijmegen, The Netherlands.,Department of Internal Medicine, Radboud University Medical Center PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Christine Stabell Benn
- Research Center for Vitamins and Vaccines, Bandim Health Project, Statens Serum Institut, 2300, Copenhagen, Denmark.,Odense Patient Data Explorative Network, University of Southern Denmark/Odense University Hospital, 5000, Odense, Denmark
| | - Mihai G Netea
- Radboud Center for Infectious Diseases, Radboud University Medical Center PO Box 9101, 6500 HB, Nijmegen, The Netherlands.,Department of Internal Medicine, Radboud University Medical Center PO Box 9101, 6500 HB, Nijmegen, The Netherlands.,Department for Genomics & Immunoregulation, Life and Medical Sciences Institute (LIMES), University of Bonn, 53115, Bonn, Germany
| | - Robert W Sauerwein
- Department of Medical Microbiology, Radboud University Medical Center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands. .,Radboud Center for Infectious Diseases, Radboud University Medical Center PO Box 9101, 6500 HB, Nijmegen, The Netherlands.
| |
Collapse
|
29
|
Bivona G, Agnello L, Lo Sasso B, Scazzone C, Butera D, Gambino CM, Iacolino G, Bellia C, Ciaccio M. Vitamin D in malaria: more hypotheses than clues. Heliyon 2019; 5:e01183. [PMID: 30793054 PMCID: PMC6370580 DOI: 10.1016/j.heliyon.2019.e01183] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 01/22/2019] [Accepted: 01/25/2019] [Indexed: 12/15/2022] Open
Abstract
Vitamin D is a secosteroid hormone regulating calcium and phosphate metabolism, immune response and brain development. Low blood 25(OH)D levels have been reported in patients affected by infectious diseases caused by parasites, including malaria. Despite the high effectiveness of antimalarials, malaria is burdened with high morbidity and mortality, and the search for additional therapies is rapidly growing. Furthermore, available preventive measures have proved to be barely effective so far. Finding new prevention and therapy tools is a matter of urgency. Studies on animal models and humans have hypothesized some mechanisms by which the hormone can influence malaria pathogenesis, and the role of Vitamin D supplementation in preventing and treating this disease has been suggested. Few studies on the association between Vitamin D and malaria are available and disagreeing results have been reported. Studies in humans reporting an association between low 25(OH)D circulating levels and Malaria have a small sample size and observational study-set. Randomized controlled trials are needed in order to understand if Vitamin D administration might play a role in preventing and treating malaria.
Collapse
Affiliation(s)
- Giulia Bivona
- Section of Clinical Biochemistry and Clinical Molecular Medicine, Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Italy
| | - Luisa Agnello
- Section of Clinical Biochemistry and Clinical Molecular Medicine, Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Italy
| | - Bruna Lo Sasso
- Section of Clinical Biochemistry and Clinical Molecular Medicine, Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Italy
| | - Concetta Scazzone
- Section of Clinical Biochemistry and Clinical Molecular Medicine, Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Italy
| | - Daniela Butera
- Section of Clinical Biochemistry and Clinical Molecular Medicine, Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Italy
| | - Caterina Maria Gambino
- Section of Clinical Biochemistry and Clinical Molecular Medicine, Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Italy
| | - Giorgia Iacolino
- Section of Clinical Biochemistry and Clinical Molecular Medicine, Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Italy
| | - Chiara Bellia
- Section of Clinical Biochemistry and Clinical Molecular Medicine, Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Italy
| | - Marcello Ciaccio
- Section of Clinical Biochemistry and Clinical Molecular Medicine, Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Italy
- Department and U.O.C. Laboratory Medicine, University Hospital “Paolo Giaccone” of Palermo, Italy
| |
Collapse
|
30
|
Parra M, Yang J, Weitner M, Derrick S, Yang A, Schmidt T, Singh B, Moreno A, Akkoyunlu M. TACI Contributes to Plasmodium yoelii Host Resistance by Controlling T Follicular Helper Cell Response and Germinal Center Formation. Front Immunol 2018; 9:2612. [PMID: 30473702 PMCID: PMC6237915 DOI: 10.3389/fimmu.2018.02612] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 10/23/2018] [Indexed: 01/22/2023] Open
Abstract
The delay in parasite-specific B cell development leaves people in malaria endemic areas vulnerable to repeated Plasmodium infections. Here, we investigated the role of transmembrane activator and calcium-modulator and cyclophilin ligand interactor (TACI), a molecule involved in the generation of antigen-specific antibody secreting cells, in host response to non-lethal Plasmodium yoelii infection. We found that TACI deficiency not only resulted in higher peak parasitemia levels in P. yoelii challenged mice, but also led to a delay in parasite clearance and anti-P. yoelii Merozoite Surface Protein 1(C-terminal 19-kDa fragment [rMSP-119]) protein and anti-rMSP-119 and anti-P. yoelii IgG antibody development. There was also a delay in the generation of splenic high affinity antibody secreting cells that recognize rMSP-119 protein as compared to wild-type mice. Interestingly, coinciding with the delay in parasite clearance there was a delay in the resolution of T follicular helper (TFH) cell and germinal center (GC) B cell responses in TACI -/- mice. The persistence of TFH and GC B cells is likely a result of enhanced interaction between TFH and GC B cells because inducible costimulator ligand (ICOSL) expression was significantly higher on TACI -/- GC B cells than wild-type cells. The difference in the kinetics of GC reaction appeared to also impact the emergence of plasma cells (PC) because there was a delay in the generation of TACI -/- mice PC. Nevertheless, following the recovery from P. yoelii infection, TACI -/- and wild-type mice were both protected from a rechallenge infection. Establishment of protective B cell response was responsible for the resolution of parasitemia because B cells purified from recovered TACI -/- or wild-type mice were equally protective when introduced to naïve wild-type mice prior to P. yoelii challenge. Thus, despite the increased susceptibility of TACI -/- mice to P. yoelii infection and a delay in the development of protective antibody levels, TACI -/- mice are able to clear the infection and resist rechallenge infection.
Collapse
Affiliation(s)
- Marcela Parra
- US Food and Drug Administration, Division of Bacterial Allergenic and Parasitic Diseases, Center for Biologics Evaluation and Research, Silver Spring, MD, United States
| | - Jiyeon Yang
- US Food and Drug Administration, Division of Bacterial Allergenic and Parasitic Diseases, Center for Biologics Evaluation and Research, Silver Spring, MD, United States
| | - Megan Weitner
- US Food and Drug Administration, Division of Bacterial Allergenic and Parasitic Diseases, Center for Biologics Evaluation and Research, Silver Spring, MD, United States
| | - Steven Derrick
- US Food and Drug Administration, Division of Bacterial Allergenic and Parasitic Diseases, Center for Biologics Evaluation and Research, Silver Spring, MD, United States
| | - Amy Yang
- US Food and Drug Administration, Division of Bacterial Allergenic and Parasitic Diseases, Center for Biologics Evaluation and Research, Silver Spring, MD, United States
| | - Thomas Schmidt
- US Food and Drug Administration, Division of Bacterial Allergenic and Parasitic Diseases, Center for Biologics Evaluation and Research, Silver Spring, MD, United States
| | - Balwan Singh
- Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States
| | - Alberto Moreno
- Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States.,Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, United States
| | - Mustafa Akkoyunlu
- US Food and Drug Administration, Division of Bacterial Allergenic and Parasitic Diseases, Center for Biologics Evaluation and Research, Silver Spring, MD, United States
| |
Collapse
|
31
|
Arama C, Quin JE, Kouriba B, Östlund Farrants AK, Troye-Blomberg M, Doumbo OK. Epigenetics and Malaria Susceptibility/Protection: A Missing Piece of the Puzzle. Front Immunol 2018; 9:1733. [PMID: 30158923 PMCID: PMC6104485 DOI: 10.3389/fimmu.2018.01733] [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: 05/08/2018] [Accepted: 07/12/2018] [Indexed: 12/22/2022] Open
Abstract
A better understanding of stable changes in regulation of gene expression that result from epigenetic events is of great relevance in the development of strategies to prevent and treat infectious diseases. Histone modification and DNA methylation are key epigenetic mechanisms that can be regarded as marks, which ensure an accurate transmission of the chromatin states and gene expression profiles over generations of cells. There is an increasing list of these modifications, and the complexity of their action is just beginning to be understood. It is clear that the epigenetic landscape plays a fundamental role in most biological processes that involve the manipulation and expression of DNA. Although the molecular mechanism of gene regulation is relatively well understood, the hierarchical order of events and dependencies that lead to protection against infection remain largely unknown. In this review, we propose that host epigenetics is an essential, though relatively under studied, factor in the protection or susceptibility to malaria.
Collapse
Affiliation(s)
- Charles Arama
- Malaria Research and Training Centre, Department of Epidemiology of Parasitic Diseases, International Center of Excellence in Research, University of Sciences, Technique and Technology of Bamako, Bamako, Mali
| | - Jaclyn E Quin
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Bourèma Kouriba
- Malaria Research and Training Centre, Department of Epidemiology of Parasitic Diseases, International Center of Excellence in Research, University of Sciences, Technique and Technology of Bamako, Bamako, Mali
| | | | - Marita Troye-Blomberg
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Ogobara K Doumbo
- Malaria Research and Training Centre, Department of Epidemiology of Parasitic Diseases, International Center of Excellence in Research, University of Sciences, Technique and Technology of Bamako, Bamako, Mali
| |
Collapse
|
32
|
de Bree LCJ, Koeken VACM, Joosten LAB, Aaby P, Benn CS, van Crevel R, Netea MG. Non-specific effects of vaccines: Current evidence and potential implications. Semin Immunol 2018; 39:35-43. [PMID: 30007489 DOI: 10.1016/j.smim.2018.06.002] [Citation(s) in RCA: 164] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 06/05/2018] [Accepted: 06/08/2018] [Indexed: 12/17/2022]
Abstract
Besides protection against specific microorganisms, vaccines can induce heterologous or non-specific effects (NSE). Epidemiological data suggest that vaccination with live-attenuated vaccines such as Bacillus Calmette-Guérin (BCG), measles vaccine, and oral polio vaccine results in increased overall childhood survival, and several of these observations have been confirmed in randomized trials. Immunological mechanisms mediating NSE include heterologous lymphocyte effects and induction of innate immune memory (trained immunity). Trained immunity induces long-term functional upregulation of innate immune cells through epigenetic and metabolic reprogramming. An overview of the epidemiological evidence of non-specific effects of vaccines and the latest insights regarding the biological mechanisms behind this phenomenon is presented, and future research priorities and potential implications are discussed.
Collapse
Affiliation(s)
- L C J de Bree
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, The Netherlands; Research Center for Vitamins and Vaccines, Bandim Health Project, Statens Serum Institut, Copenhagen, Denmark; Odense Patient Data Explorative Network, University of Southern Denmark, Odense University Hospital, Odense, Denmark
| | - Valerie A C M Koeken
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Leo A B Joosten
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Peter Aaby
- Research Center for Vitamins and Vaccines, Bandim Health Project, Statens Serum Institut, Copenhagen, Denmark; Odense Patient Data Explorative Network, University of Southern Denmark, Odense University Hospital, Odense, Denmark
| | - Christine Stabell Benn
- Research Center for Vitamins and Vaccines, Bandim Health Project, Statens Serum Institut, Copenhagen, Denmark; Odense Patient Data Explorative Network, University of Southern Denmark, Odense University Hospital, Odense, Denmark
| | - Reinout van Crevel
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, The Netherlands; Department for Genomics & Immunoregulation, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany.
| |
Collapse
|
33
|
de Bree LCJ, Marijnissen RJ, Kel JM, Rosendahl Huber SK, Aaby P, Benn CS, Wijnands MVW, Diavatopoulos DA, van Crevel R, Joosten LAB, Netea MG, Dulos J. Bacillus Calmette-Guérin-Induced Trained Immunity Is Not Protective for Experimental Influenza A/Anhui/1/2013 (H7N9) Infection in Mice. Front Immunol 2018; 9:869. [PMID: 29760700 PMCID: PMC5936970 DOI: 10.3389/fimmu.2018.00869] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 04/09/2018] [Indexed: 12/17/2022] Open
Abstract
Avian influenza A of the subtype H7N9 has been responsible for almost 1,600 confirmed human infections and more than 600 deaths since its first outbreak in 2013. Although sustained human-to-human transmission has not been reported yet, further adaptations to humans in the viral genome could potentially lead to an influenza pandemic, which may have severe consequences due to the absence of pre-existent immunity to this strain at population level. Currently there is no influenza A (H7N9) vaccine available. Therefore, in case of a pandemic outbreak, alternative preventive approaches are needed, ideally even independent of the type of influenza virus outbreak. Bacillus Calmette-Guérin (BCG) is known to induce strong heterologous immunological effects, and it has been shown that BCG protects against non-related infection challenges in several mouse models. BCG immunization of mice as well as human induces trained innate immune responses, resulting in increased cytokine responses upon subsequent ex vivo peripheral blood mononuclear cell restimulation. We investigated whether BCG (Statens Serum Institut-Denmark)-induced trained immunity may protect against a lethal avian influenza A/Anhui/1/2013 (H7N9) challenge. Here, we show that isolated splenocytes as well as peritoneal macrophages of BCG-immunized BALB/c mice displayed a trained immunity phenotype resulting in increased innate cytokine responses upon ex vivo restimulation. However, after H7N9 infection, no significant differences were found between the BCG immunized and the vehicle control group at the level of survival, weight loss, pulmonary influenza A nucleoprotein staining, or histopathology. In conclusion, BCG-induced trained immunity did not result in protection in an oseltamivir-sensitive influenza A/Anhui/1/2013 (H7N9) challenge mouse model.
Collapse
Affiliation(s)
- L Charlotte J de Bree
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands.,Radboud Centre for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, Netherlands.,Research Center for Vitamins and Vaccines, Bandim Health Project, Statens Serum Institut, Copenhagen, Denmark.,Odense Patient Data Explorative Network, University of Southern Denmark, Odense University Hospital, Odense, Denmark
| | | | - Junda M Kel
- Department of Immunology, Triskelion B.V., Zeist, Netherlands
| | | | - Peter Aaby
- Research Center for Vitamins and Vaccines, Bandim Health Project, Statens Serum Institut, Copenhagen, Denmark
| | - Christine Stabell Benn
- Research Center for Vitamins and Vaccines, Bandim Health Project, Statens Serum Institut, Copenhagen, Denmark.,Odense Patient Data Explorative Network, University of Southern Denmark, Odense University Hospital, Odense, Denmark
| | | | - Dimitri A Diavatopoulos
- Radboud Centre for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, Netherlands.,Laboratory of Pediatric Infectious Diseases, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Reinout van Crevel
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands.,Radboud Centre for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, Netherlands
| | - Leo A B Joosten
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands.,Radboud Centre for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, Netherlands
| | - Mihai G Netea
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands.,Radboud Centre for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, Netherlands.,Department for Genomics and Immunoregulation, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| | | |
Collapse
|
34
|
Trained Immunity and Susceptibility to HIV. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2017; 24:CVI.00509-16. [PMID: 27847369 DOI: 10.1128/cvi.00509-16] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
In this issue of Clinical and Vaccine Immunology, K. Jensen et al. (Clin Vaccine Immunol 24:e00360-16, 2017, https://doi.org/10.1128/CVI.00360-16) describe a dual-purpose attenuated Mycobacterium tuberculosis-simian immunodeficiency virus vaccine (AMTB-SIV). Interestingly, immunized infant macaques required fewer oral exposures to SIV to become infected relative to nonimmunized animals. The authors hypothesized that augmented susceptibility to SIV was due to activation of CD4+ T cells through trained immunity. This commentary explores the possible relationship between trained immunity, enhanced CD4 T cell responses, and increased susceptibility to human immunodeficiency virus (HIV).
Collapse
|
35
|
Kandasamy R, Voysey M, McQuaid F, de Nie K, Ryan R, Orr O, Uhlig U, Sande C, O'Connor D, Pollard AJ. Non-specific immunological effects of selected routine childhood immunisations: systematic review. BMJ 2016; 355:i5225. [PMID: 27737830 PMCID: PMC5063033 DOI: 10.1136/bmj.i5225] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
OBJECTIVE To identify and characterise non-specific immunological effects after routine childhood vaccines against BCG, measles, diphtheria, pertussis, and tetanus. DESIGN Systematic review of randomised controlled trials, cohort studies, and case-control studies. DATA SOURCES Embase, PubMed, Cochrane library, and Trip searched between 1947 and January 2014. Publications submitted by a panel of experts in the specialty were also included. ELIGIBILITY CRITERIA FOR SELECTING STUDIES All human studies reporting non-specific immunological effects after vaccination with standard childhood immunisations. Studies using recombinant vaccines, no vaccine at all, or reporting only vaccine specific outcomes were excluded. The primary aim was to systematically identify, assemble, and review all available studies and data on the possible non-specific or heterologous immunological effects of BCG; measles; mumps, measles, and rubella (MMR); diphtheria; tetanus; and pertussis vaccines. RESULTS The initial search yielded 11 168 references; 77 manuscripts met the inclusion criteria for data analysis. In most included studies (48%) BCG was the vaccine intervention. The final time point of outcome measurement was primarily performed (70%) between one and 12 months after vaccination. There was a high risk of bias in the included studies, with no single study rated low risk across all assessment criteria. A total of 143 different immunological variables were reported, which, in conjunction with differences in measurement units and summary statistics, created a high number of combinations thus precluding any meta-analysis. Studies that compared BCG vaccinated with unvaccinated groups showed a trend towards increased IFN-γ production in vitro in the vaccinated groups. Increases were also observed for IFN-γ measured after BCG vaccination in response to in vitro stimulation with microbial antigens from Candida albicans, tetanus toxoid, Staphylococcus aureas, lipopolysaccharide, and hepatitis B. Cohort studies of measles vaccination showed an increase in lymphoproliferation to microbial antigens from tetanus toxoid and C albicans Increases in immunogenicity to heterologous antigens were noted after diphtheria-tetanus (herpes simplex virus and polio antibody titres) and diphtheria-tetanus-pertussis (pneumococcus serotype 14 and polio neutralising responses) vaccination. CONCLUSIONS The papers reporting non-specific immunological effects had heterogeneous study designs and could not be conventionally meta-analysed, providing a low level of evidence quality. Some studies, such as BCG vaccine studies examining in vitro IFN-γ responses and measles vaccine studies examining lymphoproliferation to microbial antigen stimulation, showed a consistent direction of effect suggestive of non-specific immunological effects. The quality of the evidence, however, does not provide confidence in the nature, magnitude, or timing of non-specific immunological effects after vaccination with BCG, diphtheria, pertussis, tetanus, or measles containing vaccines nor the clinical importance of the findings.
Collapse
Affiliation(s)
- Rama Kandasamy
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Churchill Hospital, Oxford OX3 7LE, UK NIHR Oxford Biomedical Research Centre, Oxford, OX3 7LE, UK
| | - Merryn Voysey
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Churchill Hospital, Oxford OX3 7LE, UK NIHR Oxford Biomedical Research Centre, Oxford, OX3 7LE, UK Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, OX2 6GG, UK
| | - Fiona McQuaid
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Churchill Hospital, Oxford OX3 7LE, UK NIHR Oxford Biomedical Research Centre, Oxford, OX3 7LE, UK
| | - Karlijn de Nie
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Churchill Hospital, Oxford OX3 7LE, UK NIHR Oxford Biomedical Research Centre, Oxford, OX3 7LE, UK
| | - Rebecca Ryan
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Churchill Hospital, Oxford OX3 7LE, UK NIHR Oxford Biomedical Research Centre, Oxford, OX3 7LE, UK
| | - Olivia Orr
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Churchill Hospital, Oxford OX3 7LE, UK NIHR Oxford Biomedical Research Centre, Oxford, OX3 7LE, UK
| | - Ulrike Uhlig
- Department of Paediatrics, Children's Hospital Oxford, Oxford University Hospitals NHS Trust, Oxford, OX3 9DU, UK
| | - Charles Sande
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Churchill Hospital, Oxford OX3 7LE, UK NIHR Oxford Biomedical Research Centre, Oxford, OX3 7LE, UK
| | - Daniel O'Connor
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Churchill Hospital, Oxford OX3 7LE, UK NIHR Oxford Biomedical Research Centre, Oxford, OX3 7LE, UK
| | - Andrew J Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Churchill Hospital, Oxford OX3 7LE, UK NIHR Oxford Biomedical Research Centre, Oxford, OX3 7LE, UK
| |
Collapse
|
36
|
Induction of Unconventional T Cells by a Mutant Mycobacterium bovis BCG Strain Formulated in Cationic Liposomes Correlates with Protection against Mycobacterium tuberculosis Infections of Immunocompromised Mice. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2016; 23:638-47. [PMID: 27226281 DOI: 10.1128/cvi.00232-16] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 05/18/2016] [Indexed: 02/08/2023]
Abstract
Earlier studies aimed at defining protective immunity induced by Mycobacterium bovis BCG immunization have largely focused on the induction of antituberculosis CD4(+) and CD8(+) T cell responses. Here we describe a vaccine consisting of a BCGΔmmaA4 deletion mutant formulated in dimethyl dioctadecyl-ammonium bromide (DDA) with d-(+)-trehalose 6,6'-dibehenate (TDB) (DDA/TDB) adjuvant (A4/Adj) that protected TCRδ(-/-) mice depleted of CD4(+), CD8(+), and NK1.1(+) T cells against an aerosol challenge with M. tuberculosis These mice were significantly protected relative to mice immunized with a nonadjuvanted BCGΔmmaA4 (BCG-A4) mutant and nonvaccinated controls at 2 months and 9 months postvaccination. In the absence of all T cells following treatment with anti-Thy1.2 antibody, the immunized mice lost the ability to control the infection. These results indicate that an unconventional T cell population was mediating protection in the absence of CD4(+), CD8(+), NK1.1(+), and TCRγδ T cells and could exhibit memory. Focusing on CD4(-) CD8(-) double-negative (DN) T cells, we found that these cells accumulated in the lungs postchallenge significantly more in A4/Adj-immunized mice and induced significantly greater frequencies of pulmonary gamma interferon (IFN-γ)-producing cells than were seen in the nonvaccinated or nonadjuvanted BCG control groups. Moreover, pulmonary DN T cells from the A4/Adj group exhibited significantly higher IFN-γ integrated median fluorescence intensity (iMFI) values than were seen in the control groups. We also showed that enriched DN T cells from mice immunized with A4/Adj could control mycobacterial growth in vitro significantly better than naive whole-spleen cells. These results suggest that formulating BCG in DDA/TDB adjuvant confers superior protection in immunocompromised mice and likely involves the induction of long-lived memory DN T cells.
Collapse
|
37
|
Unravelling the nature of non-specific effects of vaccines-A challenge for innate immunologists. Semin Immunol 2016; 28:377-83. [PMID: 27354354 DOI: 10.1016/j.smim.2016.05.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 05/13/2016] [Accepted: 05/17/2016] [Indexed: 01/29/2023]
Abstract
Epidemiological observations have shown that vaccines can influence morbidity and mortality more than can be ascribed to target-disease immunity. A growing number of immunological studies have helped identify possible biological mechanisms to explain these so-called nonspecific effects (NSE) of vaccines, including heterologous T-cell reactivity and innate immune memory or 'trained innate immunity', which involves epigenetic reprogramming of innate immune cells. Here, we review the epidemiological evidence for NSE as well as human, animal and in vitro immunological data that could explain these NSE, and discuss priorities for future epidemiologic and immunologic studies to further unravel the biology and optimize the benefits of current and new vaccines.
Collapse
|
38
|
Blank J, Eggers L, Behrends J, Jacobs T, Schneider BE. One Episode of Self-Resolving Plasmodium yoelii Infection Transiently Exacerbates Chronic Mycobacterium tuberculosis Infection. Front Microbiol 2016; 7:152. [PMID: 26913029 PMCID: PMC4753732 DOI: 10.3389/fmicb.2016.00152] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 01/27/2016] [Indexed: 11/13/2022] Open
Abstract
Malaria and tuberculosis (Tb) are two of the main causes of death from infectious diseases globally. The pathogenic agents, Plasmodium parasites and Mycobacterium tuberculosis, are co-endemic in many regions in the world, however, compared to other co-infections like HIV/Tb or helminth/Tb, malaria/Tb has been given less attention both in clinical and immunological studies. Due to the lack of sufficient human data, the impact of malaria on Tb and vice versa is difficult to estimate but co-infections are likely to occur very frequently. Due to its immunomodulatory properties malaria might be an underestimated risk factor for latent or active Tb patients particularly in high-endemic malaria settings were people experience reinfections very frequently. In the present study, we used the non-lethal strain of Plasmodium yoelii to investigate, how one episode of self-resolving malaria impact on a chronic M. tuberculosis infection. P. yoelii co-infection resulted in exacerbation of Tb disease as demonstrated by increased pathology and cellular infiltration of the lungs which coincided with elevated levels of pro- and anti-inflammatory mediators. T cell responses were not impaired in co-infected mice but enhanced and likely contributed to increased cytokine production. We found a slight but statistically significant increase in M. tuberculosis burden in co-infected animals and increased lung CFU was positively correlated with elevated levels of TNFα but not IL-10. Infection with P. yoelii induced the recruitment of a CD11c+ population into lungs and spleens of M. tuberculosis infected mice. CD11c+ cells isolated from P. yoelii infected spleens promoted survival and growth of M. tuberculosis in vitro. 170 days after P. yoelii infection changes in immunopathology and cellular immune responses were no longer apparent while M. tuberculosis numbers were still slightly higher in lungs, but not in spleens of co-infected mice. In conclusion, one episode of P. yoelii co-infection transiently exacerbated disease severity but had no long-term consequences on disease progression and survival of M. tuberculosis infected mice.
Collapse
Affiliation(s)
- Jannike Blank
- Division of Coinfection, Priority Research Area Infections, Research Center Borstel Borstel, Germany
| | - Lars Eggers
- Division of Coinfection, Priority Research Area Infections, Research Center Borstel Borstel, Germany
| | - Jochen Behrends
- Fluorescence Cytometry Core Facility, Research Center Borstel Borstel, Germany
| | - Thomas Jacobs
- Department of Immunology, Bernhard Nocht Institute for Tropical Medicine Hamburg, Germany
| | - Bianca E Schneider
- Division of Coinfection, Priority Research Area Infections, Research Center Borstel Borstel, Germany
| |
Collapse
|
39
|
Muraille E. The Unspecific Side of Acquired Immunity Against Infectious Disease: Causes and Consequences. Front Microbiol 2016; 6:1525. [PMID: 26793171 PMCID: PMC4707229 DOI: 10.3389/fmicb.2015.01525] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 12/18/2015] [Indexed: 11/13/2022] Open
Abstract
Acquired immunity against infectious disease (AIID) has long been considered as strictly dependent on the B and T lymphocytes of the adaptive immune system. Consequently, AIID has been viewed as highly specific to the antigens expressed by pathogens. However, a growing body of data motivates revision of this central paradigm of immunology. Unrelated past infection, vaccination, and chronic infection have been found to induce cross-protection against numerous pathogens. These observations can be partially explained by the poly-specificity of antigenic T and B receptors, the Mackaness effect and trained immunity. In addition, numerous studies highlight the importance of microbiota composition on resistance to infectious disease via direct competition or modulation of the immune response. All of these data support the idea that a non-negligible part of AIID in nature can be nonspecific to the pathogens encountered and even of the antigens expressed by pathogens. As this protection may be dependent on the private T and B repertoires produced by the random rearrangement of genes, past immune history, chronic infection, and microbiota composition, it is largely unpredictable at the individual level. However, we can reasonably expect that a better understanding of the underlying mechanisms will allow us to statistically predict cross-protection at the population level. From an evolutionary perspective, selection of immune mechanisms allowing for partially nonspecific AIID would appear to be advantageous against highly polymorphic and rapidly evolving pathogens. This new emerging paradigm may have several important consequences on our understanding of individual infectious disease susceptibility and our conception of tolerance, vaccination and therapeutic strategies against infection and cancer. It also underscores the importance of viewing the microbiota and persisting infectious agents as integral parts of the immune system.
Collapse
Affiliation(s)
- Eric Muraille
- Laboratoire de Parasitologie, Faculté de Médecine, Université Libre de BruxellesBruxelles, Belgium
| |
Collapse
|
40
|
Mycobacterium tuberculosis Coinfection Has No Impact on Plasmodium berghei ANKA-Induced Experimental Cerebral Malaria in C57BL/6 Mice. Infect Immun 2015; 84:502-10. [PMID: 26644378 DOI: 10.1128/iai.01290-15] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 11/23/2015] [Indexed: 11/20/2022] Open
Abstract
Cerebral malaria (CM) is the most severe complication of human infection with Plasmodium falciparum. The mechanisms predisposing to CM are still not fully understood. Proinflammatory immune responses are required for the control of blood-stage malaria infection but are also implicated in the pathogenesis of CM. A fine balance between pro- and anti-inflammatory immune responses is required for parasite clearance without the induction of host pathology. The most accepted experimental model to study human CM is Plasmodium berghei ANKA (PbANKA) infection in C57BL/6 mice that leads to the development of a complex neurological syndrome which shares many characteristics with the human disease. We applied this model to study the outcome of PbANKA infection in mice previously infected with Mycobacterium tuberculosis, the causative agent of tuberculosis. Tuberculosis is coendemic with malaria in large regions in the tropics, and mycobacteria have been reported to confer some degree of unspecific protection against rodent Plasmodium parasites in experimental coinfection models. We found that concomitant M. tuberculosis infection did not change the clinical course of PbANKA-induced experimental cerebral malaria (ECM) in C57BL/6 mice. The immunological environments in spleen and brain did not differ between singly infected and coinfected animals; instead, the overall cytokine and T cell responses in coinfected mice were comparable to those in animals solely infected with PbANKA. Our data suggest that M. tuberculosis coinfection is not able to change the outcome of PbANKA-induced disease, most likely because the inflammatory response induced by the parasite rapidly dominates in mice previously infected with M. tuberculosis.
Collapse
|
41
|
Freyne B, Marchant A, Curtis N. BCG-associated heterologous immunity, a historical perspective: intervention studies in animal models of infectious diseases. Trans R Soc Trop Med Hyg 2015; 109:52-61. [PMID: 25573109 DOI: 10.1093/trstmh/tru197] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The WHO Special Advisory Group of Experts (SAGE) review of the available epidemiological and trial evidence in humans concluded that bacillus Calmette-Guérin (BCG) vaccination leads to beneficial heterologous ('non-specific') effects, specifically on all-cause mortality. Randomized controlled trials showing this beneficial effect suggest improved survival is the result of enhanced protection against infection. This paper reviews the available evidence for the attenuating effects of BCG vaccine on experimental infections in animal models, including protection from bacteria, viruses, parasites and fungi. The reviewed studies suggest that BCG activates multiple immune pathways and that the basis for BCG-associated heterologous immunity may vary by pathogen. Modern immunological and molecular methods, exemplified by 'vaccinomics', are well placed to further investigate the basis of BCG's heterologous effects using a systems biology approach.
Collapse
Affiliation(s)
- Bridget Freyne
- Department of Paediatrics, The University of Melbourne, The Royal Children's Hospital Melbourne, Parkville, Vic 3052, Australia Infectious Diseases Unit, The Royal Children's Hospital Melbourne, Parkville, Vic 3052, Australia Infectious Diseases & Microbiology Research Group, Murdoch Children's Research Institute, Parkville, Vic 3052, Australia
| | - Arnaud Marchant
- Institute for Medical Immunology, Université Libre de Bruxelles, 6041 Charleroi, Belgium
| | - Nigel Curtis
- Department of Paediatrics, The University of Melbourne, The Royal Children's Hospital Melbourne, Parkville, Vic 3052, Australia Infectious Diseases Unit, The Royal Children's Hospital Melbourne, Parkville, Vic 3052, Australia Infectious Diseases & Microbiology Research Group, Murdoch Children's Research Institute, Parkville, Vic 3052, Australia
| |
Collapse
|
42
|
Blok BA, Arts RJW, van Crevel R, Benn CS, Netea MG. Trained innate immunity as underlying mechanism for the long-term, nonspecific effects of vaccines. J Leukoc Biol 2015; 98:347-56. [DOI: 10.1189/jlb.5ri0315-096r] [Citation(s) in RCA: 148] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 06/19/2015] [Indexed: 12/31/2022] Open
|
43
|
Cusick SE, Opoka RO, Lund TC, John CC, Polgreen LE. Vitamin D insufficiency is common in Ugandan children and is associated with severe malaria. PLoS One 2014; 9:e113185. [PMID: 25470777 PMCID: PMC4254466 DOI: 10.1371/journal.pone.0113185] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Accepted: 10/21/2014] [Indexed: 12/13/2022] Open
Abstract
Vitamin D plays an increasingly recognized role in the innate and adaptive immune response to infection. Based on demonstrated roles in up-regulating innate immunity, decreasing inflammation, and reducing the severity of disease in illnesses such as tuberculosis and influenza, we hypothesized that poor vitamin D status would be associated with severe malaria. We measured 25-hydroxyvitamin D [25(OH)D] by immunoassay in a sample of Ugandan children aged 18 months-12 years with severe malaria (cerebral malaria or severe malarial anemia, n = 40) and in healthy community children (n = 20). Ninety-five percent of children with severe malaria (n = 38) and 80% of control children (n = 16) were vitamin D-insufficient [plasma 25(OH)D <30 ng/mL]. Mean plasma 25(OH)D levels were significantly lower in children with severe malaria than in community children (21.2 vs. 25.3 ng/mL, p = 0.03). Logistic regression revealed that for every 1 ng/mL increase in plasma 25(OH)D, the odds of having severe malaria declined by 9% [OR = 0.91 (95% CI: 0.84, 1.0)]. These preliminary results suggest that vitamin D insufficiency may play a role in the development of severe malaria. Further prospective studies in larger cohorts are indicated to confirm the relationship of vitamin D levels to severity of malaria infection and to investigate causality.
Collapse
Affiliation(s)
- Sarah E Cusick
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Robert O Opoka
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, United States of America; Department of Paediatrics, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Troy C Lund
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Chandy C John
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Lynda E Polgreen
- Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor, University of California Los Angeles, Los Angeles, California, United States of America
| |
Collapse
|
44
|
BCG-induced protection: Effects on innate immune memory. Semin Immunol 2014; 26:512-7. [DOI: 10.1016/j.smim.2014.09.006] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 09/17/2014] [Accepted: 09/21/2014] [Indexed: 01/23/2023]
|