1
|
Devi MB, Bhattacharya A, Kumar A, Singh CT, Das S, Sarma HK, Mukherjee AK, Khan MR. Potential probiotic Lactiplantibacillus plantarum strains alleviate TNF-α by regulating ADAM17 protein and ameliorate gut integrity through tight junction protein expression in in vitro model. Cell Commun Signal 2024; 22:520. [PMID: 39468700 PMCID: PMC11514838 DOI: 10.1186/s12964-024-01900-7] [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: 08/01/2024] [Accepted: 10/19/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Lactiplantibacillus species are extensively studied for their ability to regulate host immune responses and functional therapeutic potentials. Nevertheless, there is a lack of understanding on the mechanisms of interactions with the hosts during immunoregulatory activities. METHODS Two Lactiplantibacillus plantarum strains MKMB01 and MKMB02 were tested for probiotic potential following Indian Council of Medical Research (ICMR) guidelines. Human colorectal adenocarcinoma cells such as HT-29, caco-2, and human monocytic cell THP-1 were also used to study the potential of MKMB01 and MKMB02 in regulating the host immune response when challenged with enteric pathogen Salmonella enterica typhimurium. Cells were pre-treated with MKMB01 and MKMB02 for 4 h and then stimulated with Salmonella. qRT-PCR and ELISA were used to analyze the genes and protein expression. Confocal microscopy and field emission scanning electron microscopy (FESEM) were used to visualize the effects. An Agilent Seahorse XF analyzer was used to determine real-time mitochondrial functioning. RESULTS Both probiotic strains could defend against Salmonella by maintaining gut integrity via expressing tight junction proteins (TJPs), MUC-2, and toll-like receptors (TLRs) negative regulators such as single Ig IL-1-related receptor (SIGIRR), toll-interacting protein (Tollip), interleukin-1 receptor-associated kinase (IRAK)-M, A20, and anti-inflammatory transforming growth factor-β and interleukin-10. Both strains also downregulated the expression of pro-inflammatory cytokines/chemokines interleukin-1β, monocyte chemoattractant protein (MCP)-1, tumor necrosis factor-alpha (TNF-α), interleukin 6, and nitric oxide (NO). Moreover, TNF-α sheddase protein, a disintegrin and metalloproteinase domain 17 (ADAM17), and its regulator iRhom2 were downregulated by both strains. Moreover, the bacteria also ameliorated Salmonella-induced mitochondrial dysfunction by restoring bioenergetic profiles, such as non-mitochondrial respiration, spare respiratory capacity (SRC), basal respiration, adenosine triphosphate (ATP) production, and maximal respiration. CONCLUSIONS MKMB01 and MKMB02 can reduce pathogen-induced gut-associated disorders and therefore should be further explored for their probiotic potential.
Collapse
Affiliation(s)
- M Bidyarani Devi
- Division of Life Sciences, Institute of Advanced Study in Science and Technology (IASST), Guwahati, 781035, Assam, India
- Department of Biotechnology, Gauhati University, Guwahati, 781014, Assam, India
| | - Anupam Bhattacharya
- Division of Life Sciences, Institute of Advanced Study in Science and Technology (IASST), Guwahati, 781035, Assam, India
| | - Arun Kumar
- Division of Life Sciences, Institute of Advanced Study in Science and Technology (IASST), Guwahati, 781035, Assam, India
| | - Chingtham Thanil Singh
- Division of Life Sciences, Institute of Advanced Study in Science and Technology (IASST), Guwahati, 781035, Assam, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Santanu Das
- Division of Life Sciences, Institute of Advanced Study in Science and Technology (IASST), Guwahati, 781035, Assam, India
| | - Hridip Kumar Sarma
- Department of Biotechnology, Gauhati University, Guwahati, 781014, Assam, India
| | - Ashis K Mukherjee
- Division of Life Sciences, Institute of Advanced Study in Science and Technology (IASST), Guwahati, 781035, Assam, India
- Microbial Biotechnology and Protein Research laboratory, Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur, 784028, Assam, India
| | - Mojibur R Khan
- Division of Life Sciences, Institute of Advanced Study in Science and Technology (IASST), Guwahati, 781035, Assam, India.
| |
Collapse
|
2
|
Zhou L, Wang X, Xiao Q, Khan S, Ho WZ. Flagellin Restricts HIV-1 Infection of Macrophages through Modulation of Viral Entry Receptors and CC Chemokines. Viruses 2024; 16:1063. [PMID: 39066226 PMCID: PMC11281555 DOI: 10.3390/v16071063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 06/18/2024] [Accepted: 06/27/2024] [Indexed: 07/28/2024] Open
Abstract
Both bacteria product flagellin and macrophages are implicated in HIV-1 infection/disease progression. However, the impact of their interaction on HIV-1 infection and the associated mechanisms remain to be determined. We thus examined the effect of the flagellins on HIV-1 infection of primary human macrophages. We observed that the pretreatment of macrophages with the flagellins from the different bacteria significantly inhibited HIV-1 infection. The mechanistic investigation showed that the flagellin treatment of macrophages downregulated the major HIV-1 entry receptors (CD4 and CCR5) and upregulated the CC chemokines (MIP-1α, MIP-1β and RANTES), the ligands of CCR5. These effects of the flagellin could be compromised by a toll-like receptor 5 (TLR5) antagonist. Given the important role of flagellin as a vaccine adjuvant in TLR5 activation-mediated immune regulation and in HIV-1 infection of macrophages, future investigations are necessary to determine the in vivo impact of flagellin-TLR5 interaction on macrophage-mediated innate immunity against HIV-1 infection and the effectiveness of flagellin adjuvant-based vaccines studies.
Collapse
Affiliation(s)
| | | | | | | | - Wen-Zhe Ho
- Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
| |
Collapse
|
3
|
Acevedo-Monroy SE, Rocha-Ramírez LM, Martínez Gómez D, Basurto-Alcántara FJ, Medina-Contreras Ó, Hernández-Chiñas U, Quiñones-Peña MA, García-Sosa DI, Ramírez-Lezama J, Rodríguez-García JA, González-Villalobos E, Castro-Luna R, Martínez-Cristóbal L, Eslava-Campos CA. Polyvalent Bacterial Lysate with Potential Use to Treatment and Control of Recurrent Urinary Tract Infections. Int J Mol Sci 2024; 25:6157. [PMID: 38892345 PMCID: PMC11173243 DOI: 10.3390/ijms25116157] [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: 05/01/2024] [Revised: 05/21/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
Overuse of antimicrobials has greatly contributed to the increase in the emergence of multidrug-resistant bacteria, a situation that hinders the control and treatment of infectious diseases. This is the case with urinary tract infections (UTIs), which represent a substantial percentage of worldwide public health problems, thus the need to look for alternatives for their control and treatment. Previous studies have shown the usefulness of autologous bacterial lysates as an alternative for the treatment and control of UTIs. However, a limitation is the high cost of producing individual immunogens. At the same time, an important aspect of vaccines is their immunogenic amplitude, which is the reason why they must be constituted of diverse antigenic components. In the case of UTIs, the etiology of the disease is associated with different bacteria, and even Escherichia coli, the main causal agent of the disease, is made up of several antigenic variants. In this work, we present results on the study of a bacterial lysate composed of 10 serotypes of Escherichia coli and by Klebsiella pneumoniae, Klebsiella aerogenes, Enterococcus faecalis, Proteus mirabilis, Citrobacter freundii, and Staphylococcus haemolyticus. The safety of the compound was tested on cells in culture and in an animal model, and its immunogenic capacity by analysing in vitro human and murine macrophages (cell line J774 A1). The results show that the polyvalent lysate did not cause damage to the cells in culture or alterations in the animal model used. The immunostimulatory activity assay showed that it activates the secretion of TNF-α and IL-6 in human macrophages and TNF-α in murine cells. The obtained results suggest that the polyvalent lysate evaluated can be an alternative for the treatment and control of chronic urinary tract infections, which will reduce the use of antimicrobials.
Collapse
Affiliation(s)
- Salvador Eduardo Acevedo-Monroy
- Laboratorio de Patogenicidad Bacteriana, Unidad de Hemato-Oncología e Investigación, Hospital Infantil de México Federico Gómez/Facultad de Medicina, Universidad Nacional Autónoma de México, Dr. Márquez No. 162, Col Doctores, Alcaldía Cuauhtémoc, Ciudad de México 06720, Mexico or (S.E.A.-M.); (U.H.-C.); (M.A.Q.-P.); (D.I.G.-S.); (J.A.R.-G.)
- Laboratorio de Microbiología Molecular Departamento de Microbiología e Inmunología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Av. Universidad #3000, Colonia, C.U., Coyoacán, Ciudad de México 04510, Mexico
| | - Luz María Rocha-Ramírez
- Unidad de Investigación en Enfermedades Infecciosas, Hospital Infantil de México Federico Gómez. Secretaría de Salud, Dr. Márquez No. 162, Col Doctores, Alcaldía Cuauhtémoc, Ciudad de México 06720, Mexico;
| | - Daniel Martínez Gómez
- Departamento de Producción Agrícola y Animal, Laboratorio de Microbiología Agropecuaria, Universidad Autónoma Metropolitana Xochimilco, Calzada del Hueso 1100, Colonia Villa Quietud, Alcaldía Coyoacán, C.P., Ciudad de México 04960, Mexico;
| | - Francisco Javier Basurto-Alcántara
- Laboratorio de Vacunología y Constatación, Departamento de Microbiología e Inmunología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Av. Universidad #3000, Colonia, C.U., Coyoacán, Ciudad de México 04510, Mexico;
| | - Óscar Medina-Contreras
- Unidad de Investigación Epidemiológica en Endocrinología y Nutrición, Hospital Infantil de México Federico Gómez, Dr. Márquez No. 162, Col. Doctores, Alcaldía Cuauhtémoc, Ciudad de México 06720, Mexico;
| | - Ulises Hernández-Chiñas
- Laboratorio de Patogenicidad Bacteriana, Unidad de Hemato-Oncología e Investigación, Hospital Infantil de México Federico Gómez/Facultad de Medicina, Universidad Nacional Autónoma de México, Dr. Márquez No. 162, Col Doctores, Alcaldía Cuauhtémoc, Ciudad de México 06720, Mexico or (S.E.A.-M.); (U.H.-C.); (M.A.Q.-P.); (D.I.G.-S.); (J.A.R.-G.)
- Unidad Periférica de Investigación Básica y Clínica en Enfermedades Infecciosas; Departamento de Salud Pública, División de Investigación Facultad de Medicina, Universidad Nacional Autónoma de México, Dr. Márquez No. 162, Col Doctores, Alcaldía Cuauhtémoc, Ciudad de México 06720, Mexico
| | - María Alejandra Quiñones-Peña
- Laboratorio de Patogenicidad Bacteriana, Unidad de Hemato-Oncología e Investigación, Hospital Infantil de México Federico Gómez/Facultad de Medicina, Universidad Nacional Autónoma de México, Dr. Márquez No. 162, Col Doctores, Alcaldía Cuauhtémoc, Ciudad de México 06720, Mexico or (S.E.A.-M.); (U.H.-C.); (M.A.Q.-P.); (D.I.G.-S.); (J.A.R.-G.)
- Department of Health & Biomedical Science College of Health Professions, Biomedical Science, The University of Texas Rio Grande Valley, Edinburg, TX 78539, USA
| | - Daniela Itzel García-Sosa
- Laboratorio de Patogenicidad Bacteriana, Unidad de Hemato-Oncología e Investigación, Hospital Infantil de México Federico Gómez/Facultad de Medicina, Universidad Nacional Autónoma de México, Dr. Márquez No. 162, Col Doctores, Alcaldía Cuauhtémoc, Ciudad de México 06720, Mexico or (S.E.A.-M.); (U.H.-C.); (M.A.Q.-P.); (D.I.G.-S.); (J.A.R.-G.)
| | - José Ramírez-Lezama
- Departamento de Patología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico;
| | - José Alejandro Rodríguez-García
- Laboratorio de Patogenicidad Bacteriana, Unidad de Hemato-Oncología e Investigación, Hospital Infantil de México Federico Gómez/Facultad de Medicina, Universidad Nacional Autónoma de México, Dr. Márquez No. 162, Col Doctores, Alcaldía Cuauhtémoc, Ciudad de México 06720, Mexico or (S.E.A.-M.); (U.H.-C.); (M.A.Q.-P.); (D.I.G.-S.); (J.A.R.-G.)
| | - Edgar González-Villalobos
- Laboratorio de Epidemiología Molecular, Departamento de Salud Pública División de Investigación Facultad de Medicina, Universidad Nacional Autónoma de México, Av. Universidad #3000, Colonia, C.U., Coyoacán, Ciudad de México 04510, Mexico;
| | - Raúl Castro-Luna
- Bioterio, Hospital Infantil de México Federico Gómez, Dr. Márquez No. 162, Col Doctores, Alcaldía Cuauhtémoc, Ciudad de México 06720, Mexico; (R.C.-L.); (L.M.-C.)
| | - Leonel Martínez-Cristóbal
- Bioterio, Hospital Infantil de México Federico Gómez, Dr. Márquez No. 162, Col Doctores, Alcaldía Cuauhtémoc, Ciudad de México 06720, Mexico; (R.C.-L.); (L.M.-C.)
| | - Carlos Alberto Eslava-Campos
- Laboratorio de Patogenicidad Bacteriana, Unidad de Hemato-Oncología e Investigación, Hospital Infantil de México Federico Gómez/Facultad de Medicina, Universidad Nacional Autónoma de México, Dr. Márquez No. 162, Col Doctores, Alcaldía Cuauhtémoc, Ciudad de México 06720, Mexico or (S.E.A.-M.); (U.H.-C.); (M.A.Q.-P.); (D.I.G.-S.); (J.A.R.-G.)
- Unidad Periférica de Investigación Básica y Clínica en Enfermedades Infecciosas; Departamento de Salud Pública, División de Investigación Facultad de Medicina, Universidad Nacional Autónoma de México, Dr. Márquez No. 162, Col Doctores, Alcaldía Cuauhtémoc, Ciudad de México 06720, Mexico
| |
Collapse
|
4
|
Salerno-Goncalves R, Chen H, Bafford AC, Izquierdo M, Hormazábal JC, Lagos R, Tettelin H, D’Mello A, Booth JS, Fasano A, Levine MM, Sztein MB. Early host immune responses in a human organoid-derived gallbladder monolayer to Salmonella Typhi strains from patients with acute and chronic infections: a comparative analysis. Front Immunol 2024; 15:1334762. [PMID: 38533492 PMCID: PMC10963533 DOI: 10.3389/fimmu.2024.1334762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 02/26/2024] [Indexed: 03/28/2024] Open
Abstract
Salmonella enterica serovar Typhi (S. Typhi), a human-restricted pathogen, invades the host through the gut to cause typhoid fever. Recent calculations of the typhoid fever burden estimated that more than 10 million new typhoid fever cases occur in low and middle-income countries, resulting in 65,400-187,700 deaths yearly. Interestingly, if not antibiotic-treated, upon the resolution of acute disease, 1%-5% of patients become asymptomatic chronic carriers. Chronically infected hosts are not only critical reservoirs of infection that transmit the disease to naive individuals but are also predisposed to developing gallbladder carcinoma. Nevertheless, the molecular mechanisms involved in the early interactions between gallbladder epithelial cells and S. Typhi remain largely unknown. Based on our previous studies showing that closely related S. Typhi strains elicit distinct innate immune responses, we hypothesized that host molecular pathways activated by S. Typhi strains derived from acutely and chronically infected patients would differ. To test this hypothesis, we used a novel human organoid-derived polarized gallbladder monolayer model, and S. Typhi strains derived from acutely and chronically infected patients. We found that S. Typhi strains derived from acutely and chronically infected patients differentially regulate host mitogen-activated protein kinase (MAPK) and S6 transcription factors. These variations might be attributed to differential cytokine signaling, predominantly via TNF-α and IL-6 production and appear to be influenced by the duration the isolate was subjected to selective pressures in the gallbladder. These findings represent a significant leap in understanding the complexities behind chronic S. Typhi infections in the gallbladder and may uncover potential intervention targets.
Collapse
Affiliation(s)
- Rosângela Salerno-Goncalves
- Center for Vaccine Development and Global Health and Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Haiyan Chen
- Center for Vaccine Development and Global Health and Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Andrea C. Bafford
- Division of General and Oncologic Surgery, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Mariana Izquierdo
- Center for Vaccine Development and Global Health and Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Juan Carlos Hormazábal
- Seccion Bacteriologia, Subdepartamento de Enfermedades Infecciosas, Departamento de Laboratorio Biomédico, Instituto de Salud Pública de Chile (ISP), Santiago, Chile
| | - Rosanna Lagos
- Seccion Bacteriologia, Subdepartamento de Enfermedades Infecciosas, Departamento de Laboratorio Biomédico, Instituto de Salud Pública de Chile (ISP), Santiago, Chile
| | - Hervé Tettelin
- Department of Microbiology and Immunology and Institute for Genome Sciences (IGS), University of Maryland School of Medicine, Baltimore, MD, United States
| | - Adonis D’Mello
- Department of Microbiology and Immunology and Institute for Genome Sciences (IGS), University of Maryland School of Medicine, Baltimore, MD, United States
| | - Jayaum S. Booth
- Center for Vaccine Development and Global Health and Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Alessio Fasano
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Boston, MA, United States
| | - Myron M. Levine
- Center for Vaccine Development and Global Health and Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Marcelo B. Sztein
- Center for Vaccine Development and Global Health and Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
- Program in Oncology, University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, United States
| |
Collapse
|
5
|
Xu H, Xiong S, Chen Y, Ye Q, Guan N, Hu Y, Wu J. Flagella of Tumor-Targeting Bacteria Trigger Local Hemorrhage to Reprogram Tumor-Associated Macrophages for Improved Antitumor Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2303357. [PMID: 37310893 DOI: 10.1002/adma.202303357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 06/12/2023] [Indexed: 06/15/2023]
Abstract
Tumor-associated macrophages (TAMs) exhibit an immunosuppressive M2 phenotype and lead to failure of antitumor therapy. Infiltrated erythrocytes during hemorrhage are recognized as a promising strategy for polarizing TAMs. However, novel materials that precisely induce tumor hemorrhage without affecting normal coagulation still face challenges. Here, tumor-targeting bacteria (flhDC VNP) are genetically constructed to realize precise tumor hemorrhage. FlhDC VNP colonizes the tumor and overexpresses flagella during proliferation. The flagella promote the expression of tumor necrosis factor α, which induces local tumor hemorrhage. Infiltrated erythrocytes during the hemorrhage temporarily polarize macrophages to the M1 subtype. In the presence of artesunate, this short-lived polarization is transformed into a sustained polarization because artesunate and heme form a complex that continuously produces reactive oxygen species. Therefore, the flagella of active tumor-targeting bacteria may open up new strategies for reprogramming TAMs and improving antitumor therapy.
Collapse
Affiliation(s)
- Haiheng Xu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing, 210093, China
- Department of Andrology, Medical School of Nanjing University, Nanjing, 210093, China
- Jiangsu Provincial Key Laboratory for Nano Technology, Nanjing University, Nanjing, 210093, China
- Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing, 210023, China
- Wuxi Xishan NJU Institute of Applied Biotechnology, Anzhen Street, Xishan District, Wuxi, 214101, China
| | - Shuqin Xiong
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing, 210093, China
- Department of Andrology, Medical School of Nanjing University, Nanjing, 210093, China
- Jiangsu Provincial Key Laboratory for Nano Technology, Nanjing University, Nanjing, 210093, China
- Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing, 210023, China
- Wuxi Xishan NJU Institute of Applied Biotechnology, Anzhen Street, Xishan District, Wuxi, 214101, China
| | - Yiyun Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing, 210093, China
- Department of Andrology, Medical School of Nanjing University, Nanjing, 210093, China
- Jiangsu Provincial Key Laboratory for Nano Technology, Nanjing University, Nanjing, 210093, China
- Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing, 210023, China
- Wuxi Xishan NJU Institute of Applied Biotechnology, Anzhen Street, Xishan District, Wuxi, 214101, China
| | - Qingsong Ye
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing, 210093, China
- Department of Andrology, Medical School of Nanjing University, Nanjing, 210093, China
- Jiangsu Provincial Key Laboratory for Nano Technology, Nanjing University, Nanjing, 210093, China
- Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing, 210023, China
- Wuxi Xishan NJU Institute of Applied Biotechnology, Anzhen Street, Xishan District, Wuxi, 214101, China
| | - Nan Guan
- Molecular, Cellular and Development Biology Department, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Yiqiao Hu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing, 210093, China
- Jiangsu Provincial Key Laboratory for Nano Technology, Nanjing University, Nanjing, 210093, China
| | - Jinhui Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing, 210093, China
- Department of Andrology, Medical School of Nanjing University, Nanjing, 210093, China
- Jiangsu Provincial Key Laboratory for Nano Technology, Nanjing University, Nanjing, 210093, China
- Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing, 210023, China
- Wuxi Xishan NJU Institute of Applied Biotechnology, Anzhen Street, Xishan District, Wuxi, 214101, China
| |
Collapse
|
6
|
Piazzesi A, Putignani L. Extremely small and incredibly close: Gut microbes as modulators of inflammation and targets for therapeutic intervention. Front Microbiol 2022; 13:958346. [PMID: 36071979 PMCID: PMC9441770 DOI: 10.3389/fmicb.2022.958346] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 07/25/2022] [Indexed: 11/15/2022] Open
Abstract
Chronic inflammation is a hallmark for a variety of disorders and is at least partially responsible for disease progression and poor patient health. In recent years, the microbiota inhabiting the human gut has been associated with not only intestinal inflammatory diseases but also those that affect the brain, liver, lungs, and joints. Despite a strong correlation between specific microbial signatures and inflammation, whether or not these microbes are disease markers or disease drivers is still a matter of debate. In this review, we discuss what is known about the molecular mechanisms by which the gut microbiota can modulate inflammation, both in the intestine and beyond. We identify the current gaps in our knowledge of biological mechanisms, discuss how these gaps have likely contributed to the uncertain outcome of fecal microbiota transplantation and probiotic clinical trials, and suggest how both mechanistic insight and -omics-based approaches can better inform study design and therapeutic intervention.
Collapse
Affiliation(s)
- Antonia Piazzesi
- Multimodal Laboratory Medicine Research Area, Unit of Human Microbiome, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Lorenza Putignani
- Department of Diagnostic and Laboratory Medicine, Unit of Microbiology and Diagnostic Immunology, Unit of Microbiomics and Multimodal Laboratory Medicine Research Area, Unit of Human Microbiome, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
- *Correspondence: Lorenza Putignani,
| |
Collapse
|
7
|
Díaz-Dinamarca DA, Salazar ML, Castillo BN, Manubens A, Vasquez AE, Salazar F, Becker MI. Protein-Based Adjuvants for Vaccines as Immunomodulators of the Innate and Adaptive Immune Response: Current Knowledge, Challenges, and Future Opportunities. Pharmaceutics 2022; 14:1671. [PMID: 36015297 PMCID: PMC9414397 DOI: 10.3390/pharmaceutics14081671] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/05/2022] [Accepted: 08/07/2022] [Indexed: 12/03/2022] Open
Abstract
New-generation vaccines, formulated with subunits or nucleic acids, are less immunogenic than classical vaccines formulated with live-attenuated or inactivated pathogens. This difference has led to an intensified search for additional potent vaccine adjuvants that meet safety and efficacy criteria and confer long-term protection. This review provides an overview of protein-based adjuvants (PBAs) obtained from different organisms, including bacteria, mollusks, plants, and humans. Notably, despite structural differences, all PBAs show significant immunostimulatory properties, eliciting B-cell- and T-cell-mediated immune responses to administered antigens, providing advantages over many currently adopted adjuvant approaches. Furthermore, PBAs are natural biocompatible and biodegradable substances that induce minimal reactogenicity and toxicity and interact with innate immune receptors, enhancing their endocytosis and modulating subsequent adaptive immune responses. We propose that PBAs can contribute to the development of vaccines against complex pathogens, including intracellular pathogens such as Mycobacterium tuberculosis, those with complex life cycles such as Plasmodium falciparum, those that induce host immune dysfunction such as HIV, those that target immunocompromised individuals such as fungi, those with a latent disease phase such as Herpes, those that are antigenically variable such as SARS-CoV-2 and those that undergo continuous evolution, to reduce the likelihood of outbreaks.
Collapse
Affiliation(s)
- Diego A. Díaz-Dinamarca
- Fundación Ciencia y Tecnología para el Desarrollo (FUCITED), Santiago 7750000, Chile
- Sección de Biotecnología, Departamento Agencia Nacional de Dispositivos Médicos, Innovación y Desarrollo, Instituto de Salud Pública de Chile, Santiago 7750000, Chile
| | - Michelle L. Salazar
- Fundación Ciencia y Tecnología para el Desarrollo (FUCITED), Santiago 7750000, Chile
| | - Byron N. Castillo
- Fundación Ciencia y Tecnología para el Desarrollo (FUCITED), Santiago 7750000, Chile
| | - Augusto Manubens
- Fundación Ciencia y Tecnología para el Desarrollo (FUCITED), Santiago 7750000, Chile
- Biosonda Corporation, Santiago 7750000, Chile
| | - Abel E. Vasquez
- Sección de Biotecnología, Departamento Agencia Nacional de Dispositivos Médicos, Innovación y Desarrollo, Instituto de Salud Pública de Chile, Santiago 7750000, Chile
- Facultad de Ciencias para el Cuidado de la Salud, Universidad San Sebastián, Providencia, Santiago 8320000, Chile
| | - Fabián Salazar
- Fundación Ciencia y Tecnología para el Desarrollo (FUCITED), Santiago 7750000, Chile
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter EX4 4QD, UK
| | - María Inés Becker
- Fundación Ciencia y Tecnología para el Desarrollo (FUCITED), Santiago 7750000, Chile
- Biosonda Corporation, Santiago 7750000, Chile
| |
Collapse
|
8
|
Dai F, Guo M, Shao Y, Li C. Vibrio splendidus flagellin C binds tropomodulin to induce p38 MAPK-mediated p53-dependent coelomocyte apoptosis in Echinodermata. J Biol Chem 2022; 298:102091. [PMID: 35654141 PMCID: PMC9249833 DOI: 10.1016/j.jbc.2022.102091] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/09/2022] [Accepted: 05/13/2022] [Indexed: 12/27/2022] Open
Abstract
As a typical pathogen-associated molecular pattern, bacterial flagellin can bind Toll-like receptor 5 and the intracellular NAIP5 receptor component of the NLRC4 inflammasome to induce immune responses in mammals. However, these flagellin receptors are generally poorly understood in lower animal species. In this study, we found that the isolated flagellum of Vibrio splendidus AJ01 destroyed the integrity of the tissue structure of coelomocytes and promoted apoptosis in the sea cucumber Apostichopus japonicus. To further investigate the molecular mechanism, the novel intracellular LRR domain-containing protein tropomodulin (AjTmod) was identified as a protein that interacts with flagellin C (FliC) with a dissociation constant (Kd) of 0.0086 ± 0.33 μM by microscale thermophoresis assay. We show that knockdown of AjTmod also depressed FliC-induced apoptosis of coelomocytes. Further functional analysis with different inhibitor treatments revealed that the interaction between AjTmod and FliC could specifically activate p38 MAPK, but not JNK or ERK MAP kinases. We demonstrate that the transcription factor p38 is then translocated into the nucleus, where it mediates the expression of p53 to induce coelomocyte apoptosis. Our findings provide the first evidence that intracellular AjTmod serves as a novel receptor of FliC and mediates p53-dependent coelomocyte apoptosis by activating the p38 MAPK signaling pathway in Echinodermata.
Collapse
Affiliation(s)
- Fa Dai
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, Ningbo, China; State-Province Joint Laboratory of Marine Biotechnology and Engineering, Ningbo University, Ningbo, China
| | - Ming Guo
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, Ningbo, China; State-Province Joint Laboratory of Marine Biotechnology and Engineering, Ningbo University, Ningbo, China
| | - Yina Shao
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, Ningbo, China; State-Province Joint Laboratory of Marine Biotechnology and Engineering, Ningbo University, Ningbo, China
| | - Chenghua Li
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, Ningbo, China; State-Province Joint Laboratory of Marine Biotechnology and Engineering, Ningbo University, Ningbo, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, PR China.
| |
Collapse
|
9
|
Takahashi R, Radcliff FJ, Proft T, Tsai CJ. Pilus proteins from
Streptococcus pyogenes
stimulate innate immune responses through Toll‐like receptor 2. Immunol Cell Biol 2022; 100:174-185. [PMID: 35124861 PMCID: PMC9303359 DOI: 10.1111/imcb.12523] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/27/2022] [Accepted: 02/02/2022] [Indexed: 11/28/2022]
Abstract
The group A Streptococcus (GAS) pilus is a long, flexible, hair‐like structure anchored to the cell surface that facilitates the adherence of GAS to host cells, thus playing a critical role in initiating infections. Because of its important role in GAS virulence, the pilus has become an attractive target for vaccine development. While current research mainly focuses on pilus function and its potential as a vaccine component, there is a lack of knowledge on how the host immune system recognizes and responds to this abundant surface structure. Here we show that both assembled GAS pili and individual pilus proteins induce a potent release of the proinflammatory cytokines tumor necrosis factor and interleukin‐8. We further show that the surface‐exposed backbone pilin and ancillary pilin 1 subunits are Toll‐like receptor 2 (TLR2) agonists. Using reporter cell lines coexpressing human TLR2 in combination with either TLR1 or TLR6, we determined that activation was mediated by the TLR2/TLR6 heterodimer. Finally, we used solid‐phase and flow cytometry binding assays to illustrate a direct interaction between the pilus subunits and TLR2. These results provide further support for the suitability of the pilus as a vaccine component and opens potential avenues for using GAS pili as an adjuvant or immune‐modulation agent.
Collapse
Affiliation(s)
- Risa Takahashi
- Department of Molecular Medicine and Pathology, School of Medical Sciences The University of Auckland Auckland New Zealand
| | - Fiona J Radcliff
- Department of Molecular Medicine and Pathology, School of Medical Sciences The University of Auckland Auckland New Zealand
- Maurice Wilkins Centre for Biomolecular Discoveries The University of Auckland Auckland New Zealand
| | - Thomas Proft
- Department of Molecular Medicine and Pathology, School of Medical Sciences The University of Auckland Auckland New Zealand
- Maurice Wilkins Centre for Biomolecular Discoveries The University of Auckland Auckland New Zealand
| | - Catherine J‐Y Tsai
- Department of Molecular Medicine and Pathology, School of Medical Sciences The University of Auckland Auckland New Zealand
- Maurice Wilkins Centre for Biomolecular Discoveries The University of Auckland Auckland New Zealand
| |
Collapse
|
10
|
Zhao BR, Wang XX, Wang XW. Shoc2 recognizes bacterial flagellin and mediates antibacterial Erk/Stat signaling in an invertebrate. PLoS Pathog 2022; 18:e1010253. [PMID: 35073369 PMCID: PMC8812994 DOI: 10.1371/journal.ppat.1010253] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 02/03/2022] [Accepted: 01/06/2022] [Indexed: 11/18/2022] Open
Abstract
Flagellin is a key bacterial virulence factor that can stimulate molecular immune signaling in both animals and plants. The detailed mechanisms of recognizing flagellin and mounting an efficient immune response have been uncovered in vertebrates; however, whether invertebrates can discriminate flagellin remains largely unknown. In the present study, the homolog of human SHOC2 leucine rich repeat scaffold protein in kuruma shrimp (Marsupenaeus japonicus), designated MjShoc2, was found to interact with Vibrio anguillarum flagellin A (FlaA) using yeast two-hybrid and pull-down assays. MjShoc2 plays a role in antibacterial response by mediating the FlaA-induced expression of certain antibacterial effectors, including lectin and antimicrobial peptide. FlaA challenge, via MjShoc2, led to phosphorylation of extracellular regulated kinase (Erk), and the subsequent activation of signal transducer and activator of transcription (Stat), ultimately inducing the expression of effectors. Therefore, by establishing the FlaA/MjShoc2/Erk/Stat signaling axis, this study revealed a new antibacterial strategy in shrimp, and provides insights into the flagellin sensing mechanism in invertebrates. Flagellin sensing has been proven as a general antibacterial strategy. Recognition of bacterial flagellin by the transmembrane receptor toll like receptor 5 (TLR5) leads to the activation of nuclear factor kappa B (NF-κB) pathway and induction of proinflammatory cytokines, while recognition by the intracellular nucleotide-binding leucine-rich (NLR) receptor leads to caspase-activation and cytokines-expression. Although flagellin is an effective immune stimulator that induces antimicrobial peptides in Drosophila and in crustaceans, how an invertebrate host senses flagellin and mounts an immune response is poorly understood. Here, we used the flagellin (FlaA) from Vibrio anguillarum, a pathogen of shrimp, as a bait protein to screen a yeast two-hybrid library derived from kuruma shrimp (Marsupenaeus japonicus). We found a scaffold protein, MjShoc2, able to interact with FlaA. We also found that FlaA could effectively induce the expression of certain recognized antibacterial effectors in shrimp depending on MjShoc2. We revealed that extracellular regulated kinase (Erk) phosphorylation occurred downstream of FlaA/MjShoc2, and led to signal transducer and activator of transcription (Stat) activation, resulting in transcription of certain effectors. Therefore our study provides new insights into the FlaA-induced molecular immunity in invertebrates.
Collapse
Affiliation(s)
- Bao-Rui Zhao
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Xin-Xin Wang
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Xian-Wei Wang
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
- * E-mail:
| |
Collapse
|
11
|
Dey TK, Bose P, Paul S, Karmakar BC, Saha RN, Gope A, Koley H, Ghosh A, Dutta S, Dhar P, Mukhopadhyay AKKUMAR. Protective efficacy of fish oil nanoemulsion against non-typhoidal Salmonella mediated mucosal inflammation and loss of barrier function. Food Funct 2022; 13:10083-10095. [DOI: 10.1039/d1fo04419b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Non-typhoidal Salmonella serotypes are well-adapted to utilize the inflammation for colonization in mammalian gut mucosa and bring down the integrity of the epithelial barrier in mammalian intestine. The present study...
Collapse
|
12
|
Cage Environment Regulates Gut Microbiota Independent of Toll-Like Receptors. Infect Immun 2021; 89:e0018721. [PMID: 33941577 DOI: 10.1128/iai.00187-21] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The gut microbiome orchestrates epithelial homeostasis and both local and remote immunological responses. Critical to these regulatory interactions are innate immune receptors termed Toll-like receptors (TLRs). Studies to date have implicated innate immunity and Toll-like receptors in shaping key features of the gut microbiome. However, a variety of biological and environmental variables are also implicated in determining gut microbiota composition. In this report, we hypothesized that cohousing and environment dominated the regulation of the gut microbiota in animal models independent of innate immunity. To determine the importance of these variables, innate immunity, or environment in shaping gut microbiota, we used a randomized cohousing strategy and transgenic TLR-deficient mice. We have found that mice cohoused together by genotype exhibited limited changes over time in the composition of the gut microbiota. However, for mice randomized to cage, we report extensive changes in the gut microbiota, independent of TLR function, whereby the fecal microbiota of TLR-deficient mice converges with that of wild-type mice. TLR5-deficient mice in these experiments exhibit greater susceptibility to comparative changes in the microbiota than other TLR-deficient mice and wild-type mice. Our work has broad implications for the study of innate immunity and host-microbiota interactions. Given the profound impact that gut dysbiosis may have on immunity, this report highlights the potential impact of cohousing on the gut microbiota and indices of inflammation as outcomes in biological models of infectious or inflammatory disease.
Collapse
|
13
|
Maiti S, Howlader DR, Halder P, Bhaumik U, Dutta M, Dutta S, Koley H. Bivalent non-typhoidal Salmonella outer membrane vesicles immunized mice sera confer passive protection against gastroenteritis in a suckling mice model. Vaccine 2020; 39:380-393. [PMID: 33303233 DOI: 10.1016/j.vaccine.2020.11.040] [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: 06/08/2020] [Revised: 11/06/2020] [Accepted: 11/15/2020] [Indexed: 11/28/2022]
Abstract
Invasive non-typhoidal Salmonella (iNTS) serovars, especially Salmonella Typhimurium (ST) and Salmonella Enteritidis (SE), cause gastroenteritis worldwide. Due to the emergence of multi-drug resistance in iNTS, a broad-spectrum vaccine is urgently needed for the prevention of iNTS infection. Currently, there is no effective licensed vaccine against iNTS available in the market. We have formulated an outer membrane vesicles (OMVs) based bivalent immunogen as a vaccine candidate to generate broad-spectrum protective immunity against both recently circulating prevalent ST and SE. We have isolated OMVs from ST and SE and formulated the immunogen by mixing both OMVs (1:1 ratio). Three doses of bivalent immunogen significantly induced humoral immune responses against lipopolysaccharides (LPSs) and outer membrane proteins (OMPs) as well as a cell-mediated immune response in adult mice. We also observed that proteins of OMVs act as an adjuvant for generation of high levels of anti-LPS antibodies through T cell activation. We then characterized the one-day old suckling mice model for both ST and SE mediated gastroenteritis and used the model for a passive protection study. In the passive protection study, we found the passive transfer of bivalent OMVs immunized sera significantly reduced ST and SE mediated colonization and gastroenteritis symptoms in the colon of suckling mice compared to non-immunized sera recipients. The overall study demonstrated that OMVs based bivalent vaccine could generate broad-spectrum immunity against prevalent iNTS mediated gastroenteritis. This study also established the suckling mice model as a suitable animal model for vaccine study against iNTS mediated gastroenteritis.
Collapse
Affiliation(s)
- Suhrid Maiti
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, P-33 CIT Road, Scheme-XM, Beliaghata, Kolkata 700010, India
| | - Debaki Ranjan Howlader
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, P-33 CIT Road, Scheme-XM, Beliaghata, Kolkata 700010, India
| | - Prolay Halder
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, P-33 CIT Road, Scheme-XM, Beliaghata, Kolkata 700010, India
| | - Ushasi Bhaumik
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, P-33 CIT Road, Scheme-XM, Beliaghata, Kolkata 700010, India
| | - Moumita Dutta
- Division of Electron Microscopy, ICMR-National Institute of Cholera and Enteric Diseases, P-33 CIT Road, Scheme-XM, Beliaghata, Kolkata 700010, India
| | - Shanta Dutta
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, P-33 CIT Road, Scheme-XM, Beliaghata, Kolkata 700010, India
| | - Hemanta Koley
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, P-33 CIT Road, Scheme-XM, Beliaghata, Kolkata 700010, India.
| |
Collapse
|
14
|
Kang W, Park A, Huh JW, You G, Jung DJ, Song M, Lee HK, Kim YM. Flagellin-Stimulated Production of Interferon-β Promotes Anti-Flagellin IgG2c and IgA Responses. Mol Cells 2020; 43:251-263. [PMID: 32131150 PMCID: PMC7103879 DOI: 10.14348/molcells.2020.2300] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 12/25/2019] [Accepted: 12/30/2019] [Indexed: 12/14/2022] Open
Abstract
Flagellin, a major structural protein of the flagellum found in all motile bacteria, activates the TLR5- or NLRC4 inflammasomedependent signaling pathway to induce innate immune responses. Flagellin can also serve as a specific antigen for the adaptive immune system and stimulate anti-flagellin antibody responses. Failure to recognize commensal-derived flagellin in TLR5-deficient mice leads to the reduction in antiflagellin IgA antibodies at steady state and causes microbial dysbiosis and mucosal barrier breach by flagellated bacteria to promote chronic intestinal inflammation. Despite the important role of anti-flagellin antibodies in maintaining the intestinal homeostasis, regulatory mechanisms underlying the flagellin-specific antibody responses are not well understood. In this study, we show that flagellin induces interferon-β (IFN-β) production and subsequently activates type I IFN receptor signaling in a TLR5- and MyD88-dependent manner in vitro and in vivo . Internalization of TLR5 from the plasma membrane to the acidic environment of endolysosomes was required for the production of IFN-β, but not for other proinflammatory cytokines. In addition, we found that antiflagellin IgG2c and IgA responses were severely impaired in interferon-alpha receptor 1 (IFNAR1)-deficient mice, suggesting that IFN-β produced by the flagellin stimulation regulates anti-flagellin antibody class switching. Our findings shed a new light on the regulation of flagellin-mediated immune activation and may help find new strategies to promote the intestinal health and develop mucosal vaccines.
Collapse
Affiliation(s)
- Wondae Kang
- Division of Integrative Biosciences & Biotechnology, Pohang University of Science and Technology, Pohang 37673, Korea
| | - Areum Park
- Division of Integrative Biosciences & Biotechnology, Pohang University of Science and Technology, Pohang 37673, Korea
| | - Ji-Won Huh
- Division of Integrative Biosciences & Biotechnology, Pohang University of Science and Technology, Pohang 37673, Korea
| | - Gihoon You
- Division of Integrative Biosciences & Biotechnology, Pohang University of Science and Technology, Pohang 37673, Korea
| | - Da-Jung Jung
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea
| | - Manki Song
- International Vaccine Institute, Seoul 08826, Korea
| | - Heung Kyu Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea
| | - You-Me Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea
| |
Collapse
|
15
|
Wemyss MA, Pearson JS. Host Cell Death Responses to Non-typhoidal Salmonella Infection. Front Immunol 2019; 10:1758. [PMID: 31402916 PMCID: PMC6676415 DOI: 10.3389/fimmu.2019.01758] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 07/11/2019] [Indexed: 12/20/2022] Open
Abstract
Salmonella enterica subsp. enterica serovar Typhimurium (S. Typhimurium) is a Gram-negative bacterium with a broad host range that causes non-typhoidal salmonellosis in humans. S. Typhimurium infects epithelial cells and macrophages in the small intestine where it replicates in a specialized intracellular niche called the Salmonella-containing vacuole (SCV) and promotes inflammation of the mucosa to induce typically self-limiting gastroenteritis. Virulence and spread of the bacterium is determined in part by the host individual's ability to limit the infection through innate immune responses at the gastrointestinal mucosa, including programmed cell death. S. Typhimurium however, has evolved a myriad of mechanisms to counteract or exploit host responses through the use of Type III Secretion Systems (T3SS), which allow the translocation of virulence (effector) proteins into the host cell for the benefit of optimal bacterial replication and dissemination. T3SS effectors have been found to interact with apoptotic, necroptotic, and pyroptotic cell death cascades, interfering with both efficient clearance of the bacteria and the recruitment of neutrophils or dendritic cells to the area of infection. The interplay of host inflammation, programmed cell death responses, and bacterial defenses in the context of non-typhoidal Salmonella (NTS) infection is a continuing area of interest within the field, and as such has been reviewed here.
Collapse
Affiliation(s)
- Madeleine A Wemyss
- Department of Molecular and Translational Research, Monash University, Clayton, VIC, Australia.,Department of Microbiology, Monash University, Clayton, VIC, Australia
| | - Jaclyn S Pearson
- Department of Molecular and Translational Research, Monash University, Clayton, VIC, Australia.,Department of Microbiology, Monash University, Clayton, VIC, Australia
| |
Collapse
|
16
|
Dos Reis EC, Leal VNC, Soares JLDS, Fernandes FP, Souza de Lima D, de Alencar BC, Pontillo A. Flagellin/NLRC4 Pathway Rescues NLRP3-Inflammasome Defect in Dendritic Cells From HIV-Infected Patients: Perspective for New Adjuvant in Immunocompromised Individuals. Front Immunol 2019; 10:1291. [PMID: 31244842 PMCID: PMC6579915 DOI: 10.3389/fimmu.2019.01291] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 05/21/2019] [Indexed: 12/27/2022] Open
Abstract
Introduction: NLRP3 inflammasome plays a key role in dendritic cells (DC) activation in response to vaccine adjuvants, however we previously showed that it is not properly activated in DC from HIV-infected patients (HIV-DC), explaining, at least in part, the poor response to immunization of these patients. Taking in account that several cytoplasmic receptors are able to activate inflammasome, and that bacterial components are considered as a novel and efficient adjuvant, we postulated that bacterial flagellin (FLG), a natural ligand of NAIP/NLRC4 inflammasome, could rescue the activation of the complex in HIV-DC. Objective: Demonstrate that FLG is able to activate monocyte-derived dendritic cells from HIV-infected individuals better than LPS, and to what extent the entity of inflammasome activation differs between DC from HIV-infected patients and healthy donors. Methods: Monocyte-derived dendritic cells from HIV-infected patients (HIV-DC) and healthy donors (HD-DC) were stimulated with FLG, and inflammasome as well as DC activation (phenotypic profile, cytokine production, autologous lymphocytes activation) were compared. Chemical and genetic inhibitors were used to depict the relative contribution of NLRC4 and NLRP3 in HIV/HD-DC response to FLG. Results: FLG properly activates HD-DC and HIV-DC. FLG induces higher inflammasome activation than LPS in HIV-DC. FLG acts through NLRC4 and NLRP3 in HD-DC, but at a lesser extent in HIV-DC due to intrinsic NLRP3 defect. Conclusions: FLG by-passes NLRP3 defect in HIV-DC, through the activation of NAIP/NLRC4 inflammasome, indicating possible future use of the bacterial component as an efficient adjuvant in immunocompromised individuals.
Collapse
Affiliation(s)
- Edione Cristina Dos Reis
- Laboratory of Immunogenetics, Department of Immunology, Institute of Biomedical Sciences/ICB, University of São Paulo/USP, São Paulo, Brazil
| | - Vinícius Nunes Cordeiro Leal
- Laboratory of Immunogenetics, Department of Immunology, Institute of Biomedical Sciences/ICB, University of São Paulo/USP, São Paulo, Brazil
| | - Jaíne Lima da Silva Soares
- Laboratory of Immunogenetics, Department of Immunology, Institute of Biomedical Sciences/ICB, University of São Paulo/USP, São Paulo, Brazil
| | - Fernanda Pereira Fernandes
- Laboratory of Immunogenetics, Department of Immunology, Institute of Biomedical Sciences/ICB, University of São Paulo/USP, São Paulo, Brazil
| | - Dhêmerson Souza de Lima
- Laboratory of Immunogenetics, Department of Immunology, Institute of Biomedical Sciences/ICB, University of São Paulo/USP, São Paulo, Brazil
| | - Bruna Cunha de Alencar
- Laboratory of Immune System Cell Biology, Department of Immunology, Institute of Biomedical Sciences/ICB, University of São Paulo/USP, São Paulo, Brazil
| | - Alessandra Pontillo
- Laboratory of Immunogenetics, Department of Immunology, Institute of Biomedical Sciences/ICB, University of São Paulo/USP, São Paulo, Brazil
| |
Collapse
|
17
|
Khani MH, Bagheri M, Zahmatkesh A, Moradi Bidhendi S. Immunostimulatory effects of truncated and full-length flagellin recombinant proteins. Microb Pathog 2018; 127:190-197. [PMID: 30528248 DOI: 10.1016/j.micpath.2018.12.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Revised: 12/03/2018] [Accepted: 12/03/2018] [Indexed: 02/04/2023]
Abstract
Problems regarding purification efficacy in recombinant technologies is due to the protein structure. Experimental manipulation of genes and the subsequent proteins may overcome this issue. In order to improve production efficacy and maintain immunestimulatory effect of flagellin, the Toll-like Receptor 5 (TLR5) ligand and a potent adjuvant, we performed a bioinformatic study to find the best model for FliC manipulation. Truncated modified FliC (tmFliC) and full length FliC (flFliC) genes were cloned and expressed in pET-21a vector and protein purification was carried out using an improved His-Tag method. Polyclonal antibodies were generated against flFliC and tmFliC in New Zealand white rabbits. IgG response to the recombinant proteins was determined by ELISA. Cross-reactivity assay was performed by ELISA for all proteins and bacteria. Immunogenicity of tmFliC and flFliC was evaluated in chicken cells, and expression level of tumor necrotic factor-α (TNF-α) and interleukin-6 (IL-6) were relatively analyzed by Real-Time-PCR. Results showed high purification efficacy for tmFliC. Antibody titer of tmFliC was significantly higher than that of flFliC. In addition, the cross-reactivity assay for both proteins and Salmonella was positive which indicates similar epitopic regions. Stimulation of both FliCs significantly increased TNF-α and IL-6 expression in peripheral blood mononuclear cells (PBMCs) and splenocytes, with higher effect observed with flFliC. IL-8 protein level increased after 6 and 24 h stimulation with different concentrations of tmFliC and flFliC. These results suggest that the aimed gene modification in fliC gene produces a bioactive immunostimulant type of flagellin which upregulates TLR5 downstream genes as well as in flFliC.
Collapse
Affiliation(s)
- Mohammad-Hosein Khani
- Department of Genomics and Genetic Engineering, Razi Vaccine and Serum Research Institute (RVSRI), Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Masoumeh Bagheri
- Department of Genomics and Genetic Engineering, Razi Vaccine and Serum Research Institute (RVSRI), Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran.
| | - Azadeh Zahmatkesh
- Department of Genomics and Genetic Engineering, Razi Vaccine and Serum Research Institute (RVSRI), Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Soheila Moradi Bidhendi
- Department of Genomics and Genetic Engineering, Razi Vaccine and Serum Research Institute (RVSRI), Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| |
Collapse
|
18
|
Pompilio A, Crocetta V, Di Bonaventura G. Stenotrophomonas maltophilia mutant lacking flagella remains virulent in DBA/2N mice but is less efficient in stimulating TNF-α expression. FEMS Microbiol Lett 2018; 365:5075579. [DOI: 10.1093/femsle/fny205] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 08/15/2018] [Indexed: 12/17/2022] Open
Affiliation(s)
- Arianna Pompilio
- Department of Medical, Oral and Biotechnological Sciences, ‘G. d’Annunzio’ University of Chieti-Pescara, Via Vestini 31, Chieti 66100, Italy
- Center of Excellence on Aging, “G. d'Annunzio” University Foundation, Via L. Polacchi 11, Chieti 66100, Italy
| | - Valentina Crocetta
- Department of Medical, Oral and Biotechnological Sciences, ‘G. d’Annunzio’ University of Chieti-Pescara, Via Vestini 31, Chieti 66100, Italy
- Center of Excellence on Aging, “G. d'Annunzio” University Foundation, Via L. Polacchi 11, Chieti 66100, Italy
| | - Giovanni Di Bonaventura
- Department of Medical, Oral and Biotechnological Sciences, ‘G. d’Annunzio’ University of Chieti-Pescara, Via Vestini 31, Chieti 66100, Italy
- Center of Excellence on Aging, “G. d'Annunzio” University Foundation, Via L. Polacchi 11, Chieti 66100, Italy
| |
Collapse
|
19
|
V T Nair D, Kollanoor-Johny A. Effect of Propionibacterium freudenreichii on Salmonella multiplication, motility, and association with avian epithelial cells1. Poult Sci 2018; 96:1376-1386. [PMID: 27738122 DOI: 10.3382/ps/pew367] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 09/01/2016] [Indexed: 11/20/2022] Open
Abstract
We investigated the effects of a probiotic bacterium, Propionibacterium freudenreichii, on Salmonella multiplication, motility, and association to and invasion of avian epithelial cells in vitro. Two subspecies of P. freudenreichii (P. freudenreichii subsp. freudenreichii and P. freudenreichii subsp. shermanii) were tested against 3 Salmonella serotypes in poultry, namely, S. Enteritidis, S. Typhimurium, and S. Heidelberg, using co-culture-, motility, multiplication, cell association, and invasion assays. Both strains of P. freudenreichii were effective in reducing or inhibiting multiplication of all 3 Salmonella serotypes in co-culture and turkey cecal contents (P ≤ 0.05). P. freudenreichii significantly reduced Salmonella motility (P ≤ 0.05). Cell culture studies revealed that P. freudenreichii associated with the avian epithelial cells effectively and reduced S. Enteritidis, S. Heidelberg, and S. Typhimurium cell association in the range of 1.0 to 1.6 log10 CFU/mL, and invasion in the range of 1.3 to 1.5 log10 CFU/mL (P ≤ 0.05), respectively. Our current in vitro results indicate the potential of P. freudenreichii against Salmonella in poultry. Follow-up in vivo studies are underway to evaluate this possibility.
Collapse
|
20
|
Kollanoor Johny A, Frye JG, Donoghue A, Donoghue DJ, Porwollik S, McClelland M, Venkitanarayanan K. Gene Expression Response of Salmonella enterica Serotype Enteritidis Phage Type 8 to Subinhibitory Concentrations of the Plant-Derived Compounds Trans-Cinnamaldehyde and Eugenol. Front Microbiol 2017; 8:1828. [PMID: 29018419 PMCID: PMC5623010 DOI: 10.3389/fmicb.2017.01828] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 09/06/2017] [Indexed: 11/13/2022] Open
Abstract
Background:Salmonella Enteritidis phage type 8 (PT8) is a major poultry-associated Salmonella strain implicated in foodborne outbreaks in the United States. We previously reported that two plant-derived compounds generally recognized as safe (GRAS), trans-cinnamaldehyde (TC), and eugenol (EG), significantly reduced S. Enteritidis colonization in broiler and layer chickens. To elucidate potential PT8 genes affected by TC and EG during colonization, a whole-genome microarray analysis of the bacterium treated with TC and EG was conducted. Results:S. Enteritidis PT8 was grown in Luria-Bertani broth at 37°C to an OD600 of ~0.5. Subinhibitory concentrations (SICs; concentration that does not inhibit bacterial growth) of TC (0.01%; 0.75 mM) or EG (0.04%; 2.46 mM) were then added to the culture. S. Enteritidis PT8 RNA was extracted before and 30 min after TC or EG addition. Labeled cDNA from three replicate experiments was subsequently hybridized to a microarray of over 99% of S. Enteritidis PT4 genes, and the hybridization signals were quantified. The plant-derived compounds down-regulated (P < 0.005) expression of S. Enteritidis PT8 genes involved in flagellar motility, regulation of the Salmonella Pathogenicity Island 1, and invasion of intestinal epithelial cells. TC and EG also suppressed transcription of genes encoding multiple transport systems and outer membrane proteins. Moreover, several metabolic and biosynthetic pathways in the pathogen were down-regulated during exposure to the plant-derived compounds. Both TC and EG stimulated the transcription of heat shock genes, such as dnaK, dnaJ, ibpB, and ibpA in S. Enteritidis PT8 (P < 0.005). The results obtained from microarray were validated using a quantitative real-time PCR. Conclusion: The plant-derived compounds TC and EG exert antimicrobial effects on S. Enteritidis PT8 by affecting multiple genes, including those associated with virulence, colonization, cell membrane composition, and transport systems.
Collapse
Affiliation(s)
- Anup Kollanoor Johny
- Department of Animal Science, University of Minnesota, Saint Paul, MN, United States
| | - Jonathan G Frye
- Bacterial Epidemiology and Antimicrobial Resistance Research Unit, USDA-ARS, Richard B. Russell Research Center, Athens, GA, United States
| | - Annie Donoghue
- Poultry Production and Product Safety Research Unit, USDA, Fayetteville, AR, United States
| | - Dan J Donoghue
- Department of Poultry Science, University of Arkansas, Fayetteville, AR, United States
| | - Steffen Porwollik
- Department of Microbiology and Molecular Genetics, University of California, Irvine, Irvine, CA, United States
| | - Michael McClelland
- Department of Microbiology and Molecular Genetics, University of California, Irvine, Irvine, CA, United States
| | | |
Collapse
|
21
|
Bacterial flagellin-a potent immunomodulatory agent. Exp Mol Med 2017; 49:e373. [PMID: 28860663 PMCID: PMC5628280 DOI: 10.1038/emm.2017.172] [Citation(s) in RCA: 232] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 04/23/2017] [Accepted: 05/01/2017] [Indexed: 01/07/2023] Open
Abstract
Flagellin is a subunit protein of the flagellum, a whip-like appendage that enables bacterial motility. Traditionally, flagellin was viewed as a virulence factor that contributes to the adhesion and invasion of host cells, but now it has emerged as a potent immune activator, shaping both the innate and adaptive arms of immunity during microbial infections. In this review, we summarize our understanding of bacterial flagellin and host immune system interactions and the role flagellin as an adjuvant, anti-tumor and radioprotective agent, and we address important areas of future research interests.
Collapse
|
22
|
Lalsiamthara J, Lee JH. Brucella lipopolysaccharide reinforced Salmonella delivering Brucella immunogens protects mice against virulent challenge. Vet Microbiol 2017. [DOI: 10.1016/j.vetmic.2017.05.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
23
|
Chaichana P, Chantratita N, Brod F, Koosakulnirand S, Jenjaroen K, Chumseng S, Sumonwiriya M, Burtnick MN, Brett PJ, Teparrukkul P, Limmathurotsakul D, Day NPJ, Dunachie SJ, West TE. A nonsense mutation in TLR5 is associated with survival and reduced IL-10 and TNF-α levels in human melioidosis. PLoS Negl Trop Dis 2017; 11:e0005587. [PMID: 28475641 PMCID: PMC5435357 DOI: 10.1371/journal.pntd.0005587] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 05/17/2017] [Accepted: 04/21/2017] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Melioidosis, caused by the flagellated bacterium Burkholderia pseudomallei, is a life-threatening and increasingly recognized emerging disease. Toll-like receptor (TLR) 5 is a germline-encoded pattern recognition receptor to bacterial flagellin. We evaluated the association of a nonsense TLR5 genetic variant that truncates the receptor with clinical outcomes and with immune responses in melioidosis. METHODOLOGY/PRINCIPAL FINDINGS We genotyped TLR5 c.1174C>T in 194 acute melioidosis patients in Thailand. Twenty-six (13%) were genotype CT or TT. In univariable analysis, carriage of the c.1174C>T variant was associated with lower 28-day mortality (odds ratio (OR) 0.21, 95% confidence interval (CI) 0.05-0.94, P = 0.04) and with lower 90-day mortality (OR 0.25, 95% CI 0.07-086, P = 0.03). In multivariable analysis adjusting for age, sex, diabetes and renal disease, the adjusted OR for 28-day mortality in carriers of the variant was 0.24 (95% CI 0.05-1.08, P = 0.06); and the adjusted OR for 90-day mortality was 0.27 (95% CI 0.08-0.97, P = 0.04). c.1174C>T was associated with a lower rate of bacteremia (P = 0.04) and reduced plasma levels of IL-10 (P = 0.049) and TNF-α (P < 0.0001). We did not find an association between c.1174C>T and IFN-γ ELISPOT (T-cell) responses (P = 0.49), indirect haemagglutination titers or IgG antibodies to bacterial flagellin during acute melioidosis (P = 0.30 and 0.1, respectively). CONCLUSIONS/SIGNIFICANCE This study independently confirms the association of TLR5 c.1174C>T with protection against death in melioidosis, identifies lower bacteremia, IL-10 and TNF-α production in carriers of the variant with melioidosis, but does not demonstrate an association of the variant with acute T-cell IFN-γ response, indirect haemagglutination antibody titer, or anti-flagellin IgG antibodies.
Collapse
Affiliation(s)
- Panjaporn Chaichana
- Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
| | - Narisara Chantratita
- Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Florian Brod
- Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
- Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | - Kemajittra Jenjaroen
- Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
| | - Suchintana Chumseng
- Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
| | | | - Mary N. Burtnick
- Department of Microbiology and Immunology, University of South Alabama, Mobile, AL, United States of America
| | - Paul J. Brett
- Department of Microbiology and Immunology, University of South Alabama, Mobile, AL, United States of America
| | | | - Direk Limmathurotsakul
- Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
- Department of Tropical Hygiene, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Center for Tropical Medicine and Global Health, University of Oxford, Oxford, United Kingdom
| | - Nicholas P. J. Day
- Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
- Center for Tropical Medicine and Global Health, University of Oxford, Oxford, United Kingdom
| | - Susanna J. Dunachie
- Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
- Center for Tropical Medicine and Global Health, University of Oxford, Oxford, United Kingdom
| | - T. Eoin West
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- International Respiratory and Severe Illness Center, University of Washington, Seattle, Washington, United States of America
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Washington, Seattle, Washington, United States of America
| |
Collapse
|
24
|
Lee SJ, Birhanu BT, Awji EG, Kim MH, Park JY, Suh JW, Park SC. BaeR protein acts as an activator of nuclear factor-kappa B and Janus kinase 2 to induce inflammation in murine cell lines. Can J Microbiol 2016; 62:753-61. [PMID: 27374640 DOI: 10.1139/cjm-2016-0057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
BaeR, a response regulator protein, takes part in multidrug efflux, bacterial virulence activity, and other biological functions. Recently, BaeR was shown to induce inflammatory responses by activating the mitogen-activated protein kinases (MAPKs). In this study, we investigated additional pathways used by BaeR to induce an inflammatory response. BaeR protein was purified from Salmonella enterica Paratyphi A and subcloned into a pPosKJ expression vector. RAW 264.7 cells were treated with BaeR, and RNA was extracted by TRIzol reagent for RT-PCR. Cytokine gene expression was analyzed by using the comparative cycle threshold method, while western blotting and ELISA were used to assess protein expression. We confirmed that BaeR activates nuclear factor-kappa B (NF-κB), thereby inducing an inflammatory response and increases the production of interleukins (IL-)1β and IL-6. During this process, the Janus kinase 2 (JAK2)-STAT1 signaling pathway was activated, resulting in an increase in the release of interferons I and II. Additionally, COX-2 was activated and its expression increased with time. In conclusion, BaeR induced an inflammatory response through activation of NF-κB in addition to the MAPKs. Furthermore, activation of the JAK2-STAT1 pathway and COX-2 facilitated the cytokine binding activity, suggesting an additional role for BaeR in the modulation of the immune system of the host and the virulence activity of the pathogen.
Collapse
Affiliation(s)
- Seung-Jin Lee
- a Laboratory of Veterinary Pharmacokinetics and Pharmacodynamics, College of Veterinary Medicine, Kyungpook National University, Daegu 702-701, Republic of Korea
| | - Biruk Tesfaye Birhanu
- a Laboratory of Veterinary Pharmacokinetics and Pharmacodynamics, College of Veterinary Medicine, Kyungpook National University, Daegu 702-701, Republic of Korea
| | - Elias Gebru Awji
- b COPD Program, Lovelace Respiratory Research Institute, Albuquerque, NM 87108, USA
| | - Myung Hee Kim
- c Infection and Immunity Research Center, Korea Research Institute of Bioscience and Biotechnology, KRIBB, Daejeon 305-806, Republic of Korea
| | - Ji-Yong Park
- d Cleanbio Research Institute, Daejeon 301-212, Korea
| | - Joo-Won Suh
- e Center for Nutraceutical and Pharmaceutical Materials, Division of Bioscience and Bioinformatics, Science campus, Myongji University, 449-728 Yongin, Gyeonggi, Republic of Korea
| | - Seung-Chun Park
- a Laboratory of Veterinary Pharmacokinetics and Pharmacodynamics, College of Veterinary Medicine, Kyungpook National University, Daegu 702-701, Republic of Korea
| |
Collapse
|
25
|
Toll-Like Receptor Activation by Generalized Modules for Membrane Antigens from Lipid A Mutants of Salmonella enterica Serovars Typhimurium and Enteritidis. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2016; 23:304-14. [PMID: 26865597 PMCID: PMC4820502 DOI: 10.1128/cvi.00023-16] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 01/30/2016] [Indexed: 01/16/2023]
Abstract
Invasive nontyphoidal Salmonella (iNTS) disease is a neglected disease with high mortality in children and HIV-positive individuals in sub-Saharan Africa, caused primarily by Africa-specific strains of Salmonella enterica serovars Typhimurium and Enteritidis. A vaccine using GMMA (generalized modules for membrane antigens) from S. Typhimurium and S. Enteritidis containing lipid A modifications to reduce potential in vivo reactogenicity is under development. GMMA with penta-acylated lipid A showed the greatest reduction in the level of cytokine release from human peripheral blood monocytes from that for GMMA with wild-type lipid A. Deletion of the lipid A modification genes msbB and pagP was required to achieve pure penta-acylation. Interestingly, ΔmsbB ΔpagP GMMA from S. Enteritidis had a slightly higher stimulatory potential than those from S. Typhimurium, a finding consistent with the higher lipopolysaccharide (LPS) content and Toll-like receptor 2 (TLR2) stimulatory potential of the former. Also, TLR5 ligand flagellin was found in Salmonella GMMA. No relevant contribution to the stimulatory potential of GMMA was detected even when the flagellin protein FliC from S. Typhimurium was added at a concentration as high as 10% of total protein, suggesting that flagellin impurities are not a major factor for GMMA-mediated immune stimulation. Overall, the stimulatory potential of S. Typhimurium and S. Enteritidis ΔmsbB ΔpagP GMMA was close to that of Shigellasonnei GMMA, which are currently in phase I clinical trials.
Collapse
|
26
|
Rady HF, Dai G, Huang W, Shellito JE, Ramsay AJ. Flagellin Encoded in Gene-Based Vector Vaccines Is a Route-Dependent Immune Adjuvant. PLoS One 2016; 11:e0148701. [PMID: 26844553 PMCID: PMC4742079 DOI: 10.1371/journal.pone.0148701] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 01/20/2016] [Indexed: 12/21/2022] Open
Abstract
Flagellin has been tested as a protein-based vaccine adjuvant, with the majority of studies focused on antibody responses. Here, we evaluated the adjuvant activity of flagellin for both cellular and humoral immune responses in BALB/c mice in the setting of gene-based immunization, and have made several novel observations. DNA vaccines and adenovirus (Ad) vectors were engineered to encode mycobacterial protein Ag85B, with or without flagellin of Salmonella typhimurium (FliC). DNA-encoded flagellin given IM enhanced splenic CD4+ and CD8+ T cell responses to co-expressed vaccine antigen, including memory responses. Boosting either IM or intranasally with Ad vectors expressing Ag85B without flagellin led to durable enhancement of Ag85B-specific antibody and CD4+ and CD8+ T cell responses in both spleen and pulmonary tissues, correlating with significantly improved protection against challenge with pathogenic aerosolized M. tuberculosis. However, inclusion of flagellin in both DNA prime and Ad booster vaccines induced localized pulmonary inflammation and transient weight loss, with route-dependent effects on vaccine-induced T cell immunity. The latter included marked reductions in levels of mucosal CD4+ and CD8+ T cell responses following IM DNA/IN Ad mucosal prime-boosting, although antibody responses were not diminished. These findings indicate that flagellin has differential and route-dependent adjuvant activity when included as a component of systemic or mucosally-delivered gene-based prime-boost immunization. Clear adjuvant activity for both T and B cell responses was observed when flagellin was included in the DNA priming vaccine, but side effects occurred when given in an Ad boosting vector, particularly via the pulmonary route.
Collapse
Affiliation(s)
- Hamada F. Rady
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, United States of America
- Louisiana Vaccine Center, Louisiana State University Health Sciences Center, New Orleans, Louisiana, United States of America
| | - Guixiang Dai
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, United States of America
- Louisiana Vaccine Center, Louisiana State University Health Sciences Center, New Orleans, Louisiana, United States of America
| | - Weitao Huang
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, United States of America
| | - Judd E. Shellito
- Department of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana, United States of America
- Louisiana Vaccine Center, Louisiana State University Health Sciences Center, New Orleans, Louisiana, United States of America
| | - Alistair J. Ramsay
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, United States of America
- Department of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana, United States of America
- Louisiana Vaccine Center, Louisiana State University Health Sciences Center, New Orleans, Louisiana, United States of America
- * E-mail:
| |
Collapse
|
27
|
King JE, Roberts IS. Bacterial Surfaces: Front Lines in Host-Pathogen Interaction. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 915:129-56. [PMID: 27193542 DOI: 10.1007/978-3-319-32189-9_10] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
All bacteria are bound by at least one membrane that acts as a barrier between the cell's interior and the outside environment. Surface components within and attached to the cell membrane are essential for ensuring that the overall homeostasis of the cell is maintained. However, many surface components of the bacterial cell also have an indispensable role mediating interactions of the bacteria with their immediate environment and as such are essential to the pathogenesis of infectious disease. During the course of an infection, bacterial pathogens will encounter many different ecological niches where environmental conditions such as salinity, temperature, pH, and the availability of nutrients fluctuate. It is the bacterial cell surface that is at the front-line of these host-pathogen interactions often protecting the bacterium from hostile surroundings but at the same time playing a critical role in the adherence to host tissues promoting colonization and subsequent infection. To deal effectively with the changing environments that pathogens may encounter in different ecological niches within the host many of the surface components of the bacterial cell are subject to phenotypic variation resulting in heterogeneous subpopulations of bacteria within the clonal population. This dynamic phenotypic heterogeneity ensures that at least a small fraction of the population will be adapted for a particular circumstance should it arise. Diversity within the clonal population has often been masked by studies on entire bacterial populations where it was often assumed genes were expressed in a uniform manner. This chapter, therefore, aims to highlight the non-uniformity in certain cell surface structures and will discuss the implication of this heterogeneity in bacterial-host interaction. Some of the recent advances in studying bacterial surface structures at the single cell level will also be reviewed.
Collapse
Affiliation(s)
- Jane E King
- Faculty of Life Sciences, University of Manchester, Manchester, M13 9PT, UK
| | - Ian S Roberts
- Faculty of Life Sciences, University of Manchester, Manchester, M13 9PT, UK.
| |
Collapse
|
28
|
Bao W, Wang Y, Fu Y, Jia X, Li J, Vangan N, Bao L, Hao H, Wang Z. mTORC1 Regulates Flagellin-Induced Inflammatory Response in Macrophages. PLoS One 2015; 10:e0125910. [PMID: 25942007 PMCID: PMC4420466 DOI: 10.1371/journal.pone.0125910] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Accepted: 03/25/2015] [Indexed: 11/28/2022] Open
Abstract
Bacterial flagellin triggers inflammatory responses. Phosphoinositide 3-kinase (PI3K) and mammalian target of rapamycin (mTOR) regulate the production of pro- and anti-inflammatory cytokines that are induced by extrinsic antigens, but the function of mTORC1 in flagellin-induced inflammatory response is unknown. The purpose of this study was to examine the role and the mechanism of PI3K/Akt/mTOR pathway in flagellin-induced cytokine expression in mouse macrophages. We observed that flagellin upregulated TNF-α time- and dose-dependently. Flagellin stimulated rapid (<15 min) PI3K/Akt/mTOR phosphorylation that was mediated by TLR5. Inhibition of PI3K with LY294002 and wortmannin, and of mTORC1 with rapamycin decreased flagellin-induced TNF-α and IL-6 expression and cell proliferation. The activation of NF-κB p65 and STAT3 was regulated by mTORC1 via degradation of IκBα and phosphorylation of STAT3 in response to flagellin, respectively. Thus, the PI3K/Akt/mTORC1 pathway regulates the innate immune response to bacterial flagellin. Rapamycin is potential therapy that can regulate host defense against pathogenic infections.
Collapse
Affiliation(s)
- Wenlei Bao
- College of Life Science, Inner Mongolia University, Hohhot, China
| | - Yanfeng Wang
- College of Life Science, Inner Mongolia University, Hohhot, China
| | - Yuting Fu
- College of Life Science, Inner Mongolia University, Hohhot, China
| | - Xiaoyang Jia
- College of Life Science, Inner Mongolia University, Hohhot, China
| | - Jiaxin Li
- College of Life Science, Inner Mongolia University, Hohhot, China
| | | | - Lili Bao
- College of Life Science, Inner Mongolia University, Hohhot, China
- College of Basic Medical Science, Inner Mongolia Medical University, Hohhot, China
| | - Huifang Hao
- College of Life Science, Inner Mongolia University, Hohhot, China
| | - Zhigang Wang
- College of Life Science, Inner Mongolia University, Hohhot, China
- * E-mail:
| |
Collapse
|
29
|
Haque SS. Immunological Evaluation of Formulated Drugs against Typhoid. Open Access Maced J Med Sci 2014. [DOI: 10.3889/oamjms.2014.069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
OBJECTIVES: Typhoid fever an important causes of illness and death, particularly among children and adolescents in south-central and Southeast Asia, where enteric fever is associated with poor sanitation and unsafe food and water. Cell-mediated immunity (CMI) plays an important role for the survival of the host in experimental salmonellosis. Nitric oxide (NO) is the one of the product of macrophages activated by cytokines, microbial compounds or both, is derived from the amino acid L-arginine by the enzymatic activity of inducible nitric oxide synthase (iNOS or NOS2) which acts as antimicrobial molecule.AIM: The aim was to examine the induction of DTH reaction in the animals treated with L-Arginine, ciprofloxacin and their combination followed by immunization with S. typhimurium cell lysate  as assessed by the footpad swelling test.RESULTS: The results of the present study showed that the induction of DTH reaction in the animals treated with L-Arginine, ciprofloxacin and their combination followed by immunization with S. typhimurium cell lysate using an antigen revealed that the treatment with combination increased foot pad swelling significantly as compared to saline treated control animals at 48 hour which was followed by a decrease of the swelling at 72 hour.CONCLUSIONS: Animal treated with L-arginine, ciprofloxacin and their combination showed increased cell mediated immune responses as evident by DTH response whereas groups (B+S) shows decreases CMI responses.
Collapse
|
30
|
Tarahomjoo S. Utilizing bacterial flagellins against infectious diseases and cancers. Antonie van Leeuwenhoek 2013; 105:275-88. [PMID: 24276957 DOI: 10.1007/s10482-013-0075-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Accepted: 11/12/2013] [Indexed: 12/31/2022]
Abstract
The flagellum is the organelle providing motility to bacterial cells and its activity is coupled to the cellular chemotaxis machinery. The flagellar filament is the largest portion of the flagellum, which consists of repeating subunits of the protein flagellin. Receptors of the innate immune system including Toll like receptor 5, ICE protease activating factor, and neuronal apoptosis inhibitory protein 5 signal in response to bacterial flagellins. In addition to inducing innate immune responses, bacterial flagellins mediate the development of adaptive immune responses to both flagellins and coadministered antigens. Therefore, these proteins have intensively been investigated for the vaccine development and the immunotherapy. This review describes the utilization of bacterial flagellins for the construction of vaccines against infectious diseases and cancer immunotherapy. Furthermore, the key factors affecting the performance of these systems are highlighted.
Collapse
Affiliation(s)
- Shirin Tarahomjoo
- Department of Biotechnology, Razi Vaccine and Serum Research Institute, 31975/148, Karaj, Iran,
| |
Collapse
|
31
|
Lee SJ, Gebru Awji E, Kim MH, Park SC. BaeR protein from Salmonella enterica serovar Paratyphi A induces inflammatory response in murine and human cell lines. Microbes Infect 2013; 15:951-7. [PMID: 24055826 DOI: 10.1016/j.micinf.2013.09.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Revised: 08/20/2013] [Accepted: 09/09/2013] [Indexed: 10/26/2022]
Abstract
BaeR is the response regulator of the two-component system, BaeSR, found in Escherichia coli (E. coli) and Salmonella. Several biological functions of BaeR, related to multidrug efflux and bacterial virulence, have been described. Herein, we report a putative function of BaeR during inflammatory response of the host by using BaeR protein of Salmonella enterica Paratyphi A (S. Paratyphi A) origin overexpressed in E. coli, and RAW 264.7 and THP-1 cells as in vitro models. BaeR (3 μg/ml) upregulated iNOS mRNA expression in both cell lines, and induced significant production of NO. Greater than ten-fold (TNF-α), 24-fold (IL-1β) and 156-fold (IL-6) increases in mRNA expression levels were observed in THP-1 cells treated with BaeR, compared to untreated controls. Furthermore, an eight-fold (IL-1β), 12-fold (IL-6) and 41-fold (TNF-α) higher protein concentrations were observed in RAW 264.7 cells stimulated with BaeR, compared to control cells. Immunoblot analysis showed BaeR-induced phosphorylation of the MAPKs (ERK 1/2, JNK and p38 MAPK) in RAW 264.7 cells. Pharmacological inhibition of the three MAPKs using specific inhibitors resulted in significant reduction of BaeR-induced NO production and iNOS mRNA expression by inhibitors of JNK and p38 MAPK. Also, all inhibitors of the MAPKs significantly attenuated BaeR-induced IL-1β, IL-6 and TNF-α at both transcript and protein levels with different degrees of inhibition. Taken together, our data suggest that BaeR is a putative inducer of inflammatory response and the MAPKs are involved in the process.
Collapse
Affiliation(s)
- Seung Jin Lee
- Laboratory of Veterinary Pharmacokinetics & Pharmacodynamics, College of Veterinary Medicine, Kyungpook National University, Daegu 702-701, Republic of Korea
| | | | | | | |
Collapse
|
32
|
Innate immune detection of flagellin positively and negatively regulates salmonella infection. PLoS One 2013; 8:e72047. [PMID: 23977202 PMCID: PMC3747147 DOI: 10.1371/journal.pone.0072047] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2013] [Accepted: 07/06/2013] [Indexed: 11/19/2022] Open
Abstract
Salmonella enterica serovar Typhimurium is a flagellated bacterium and one of the leading causes of gastroenteritis in humans. Bacterial flagellin is required for motility and also a prime target of the innate immune system. Innate immune recognition of flagellin is mediated by at least two independent pathways, TLR5 and Naip5-Naip6/NlrC4/Caspase-1. The functional significance of each of the two independent flagellin recognition systems for host defense against wild type Salmonella infection is complex, and innate immune detection of flagellin contributes to both protection and susceptibility. We hypothesized that efficient modulation of flagellin expression in vivo permits Salmonella to evade innate immune detection and limit the functional role of flagellin-specific host innate defenses. To test this hypothesis, we used Salmonella deficient in the anti-sigma factor flgM, which overproduce flagella and are attenuated in vivo. In this study we demonstrate that flagellin recognition by the innate immune system is responsible for the attenuation of flgM− S. Typhimurium, and dissect the contribution of each flagellin recognition pathway to bacterial clearance and inflammation. We demonstrate that caspase-1 controls mucosal and systemic infection of flgM− S. Typhimurium, and also limits intestinal inflammation and injury. In contrast, TLR5 paradoxically promotes bacterial colonization in the cecum and systemic infection, but attenuates intestinal inflammation. Our results indicate that Salmonella evasion of caspase-1 dependent flagellin recognition is critical for establishing infection and that evasion of TLR5 and caspase-1 dependent flagellin recognition helps Salmonella induce intestinal inflammation and establish a niche in the inflamed gut.
Collapse
|
33
|
Terron-Exposito R, Dudognon B, Galindo I, Quetglas JI, Coll JM, Escribano JM, Gomez-Casado E. Antibodies against Marinobacter algicola and Salmonella typhimurium flagellins do not cross-neutralize TLR5 activation. PLoS One 2012; 7:e48466. [PMID: 23155384 PMCID: PMC3498291 DOI: 10.1371/journal.pone.0048466] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Accepted: 09/25/2012] [Indexed: 11/18/2022] Open
Abstract
Flagellins evoke strong innate and adaptive immune responses. These proteins may play a key role as radioprotectors, exert antitumoral activity in certain types of tumor and reduce graft-versus-host disease in allogeneic hematopoietic stem cell transplant recipients. Notwithstanding, flagellins are highly immunogenic, and repeated use leads to their neutralization by systemic antibodies. This neutralization is not prevented by using functional deleted flagellins. These observations led us to explore the possibility of preventing initial neutralization by means of another functional flagellin that does not belong to common pathogenic bacteria but that has the capacity to activate TLR5. Here we characterized the functional capacity of the two-phase Marinobacter algicola (MA)-derived flagellins (F and FR) as systemic and mucosal adjuvants and compared their performance with that of Salmonella typhimurium (STF) flagellins (FljB and FliC). We also report for the first time on the in vitro and in vivo capacity of various flagellins to trigger TLR5 activation in the presence of species-specific anti-flagellin antibodies, the cross-neutralization mediated by these antibodies, and the sequential use of these flagellins for TLR5 activation. Our results showed that MA flagellins behave in a similar way to STF ones, inducing pro-inflammatory cytokines (IL8, CCL20, CCL2) and evoking a strong in vivo antibody response against a model epitope. More importantly, MA flagellins were fully functional, in vitro or in vivo, in the presence of a high concentration of neutralizing anti-flagellin STF antibodies, and STF flagellin was not inhibited by neutralizing anti-flagellin MA antibodies. The use of active flagellins from distinct bacteria could be a useful approach to prevent systemic neutralization of this group of adjuvants and to facilitate the rational design of flagellin-based vaccines and/or other therapeutic treatments (against ischemia, acute renal failure, tumors, ionizing radiations and also to improve the outcome of bone marrow transplants).
Collapse
Affiliation(s)
- Raul Terron-Exposito
- Department of Biotechnology, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, INIA, Madrid, Spain
| | - Benoit Dudognon
- Alternative Gene Expression S. L. (ALGENEX S. L.), Madrid, Spain
| | - Inmaculada Galindo
- Department of Biotechnology, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, INIA, Madrid, Spain
| | - Jose I. Quetglas
- Division of Gene Therapy, Centro de Investigación en Medicina Aplicada, CIMA, Pamplona, Spain
| | - Julio M. Coll
- Department of Biotechnology, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, INIA, Madrid, Spain
| | - Jose M. Escribano
- Department of Biotechnology, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, INIA, Madrid, Spain
| | - Eduardo Gomez-Casado
- Department of Biotechnology, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, INIA, Madrid, Spain
- * E-mail:
| |
Collapse
|
34
|
Jindal G, Tewari R, Gautam A, Pandey SK, Rishi P. Immunological characterization of recombinant Salmonella enterica serovar Typhi FliC protein expressed in Escherichia coli. AMB Express 2012; 2:55. [PMID: 23067582 PMCID: PMC3502275 DOI: 10.1186/2191-0855-2-55] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Accepted: 09/29/2012] [Indexed: 11/17/2022] Open
Abstract
Like any other enteric pathogen, Salmonella also encounters acidic stress in the stomach as well as within the host macrophage milieu. However, the pathogen is reported to combat this stress through acid tolerance response (ATR), expressing a number of genes and eventually the proteins. Recently, an acid induced outer membrane phenotype encoded by fliC gene in Salmonella enterica serovar Typhi has been identified. In the present study, fliC gene was cloned to study its biological implications. The recombinant FliC (rFliC) protein was observed to stimulate the production of antibodies. These antibodies could also recognize the FliC protein (antigen) in the clinical samples i.e. blood samples from typhoid patents as well as healthy blood samples spiked with serovar Typhi. Moreover, the rFliC also reacted with the sera from patients suffering with typhoid fever indicating its in-vivo immunogenicity. Ex-vivo study revealed that rFliC has the potential to stimulate the macrophages to generate higher levels of inflammatory mediators such as malondialdehyde (MDA) and nitrite. The inflammatory potential of FliC was also confirmed in-vivo, by the paw oedema test as well as by flicking response of the inflamed paw indicating hyperalgesia occurring during inflammatory response. The findings of the present study indicate that acid induced FliC might be one of the factors enhancing the virulence of serovar Typhi under the host acidic conditions and may prove to be helpful in designing the prophylactic measures.
Collapse
|
35
|
Impact of a mutator phenotype on motility and cell adherence in Salmonella Heidelberg. Vet Microbiol 2012; 159:99-106. [DOI: 10.1016/j.vetmic.2012.03.025] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2011] [Revised: 03/13/2012] [Accepted: 03/16/2012] [Indexed: 02/04/2023]
|
36
|
Role of antigens and virulence factors of Salmonella enterica serovar Typhi in its pathogenesis. Microbiol Res 2012; 167:199-210. [DOI: 10.1016/j.micres.2011.08.001] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2011] [Revised: 08/05/2011] [Accepted: 08/08/2011] [Indexed: 12/25/2022]
|
37
|
Escherichia coli flagellin stimulates pro-inflammatory immune response. World J Microbiol Biotechnol 2012; 28:2139-46. [DOI: 10.1007/s11274-012-1019-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2011] [Accepted: 02/01/2012] [Indexed: 10/14/2022]
|
38
|
Liu B, Hu B, Zhou Z, Guo D, Guo X, Ding P, Feng L, Wang L. A novel non-homologous recombination-mediated mechanism for Escherichia coli unilateral flagellar phase variation. Nucleic Acids Res 2012; 40:4530-8. [PMID: 22287625 PMCID: PMC3378880 DOI: 10.1093/nar/gks040] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Flagella contribute to the virulence of bacteria through chemotaxis, adhesion to and invasion of host surfaces. Flagellar phase variation is believed to facilitate bacterial evasion of the host immune response. In this study, the flnA gene that encodes Escherichia coli H17 flagellin was examined by whole genome sequencing and genetic deletion analysis. Unilateral flagellar phase variation has been reported in E. coli H3, H47 and H17 strains, although the mechanism for phase variation in the H17 strain has not been previously understood. Analysis of phase variants indicated that the flagellar phase variation in the H17 strain was caused by the deletion of an ∼35 kb DNA region containing the flnA gene from diverse excision sites. The presence of covalently closed extrachromosomal circular forms of this excised 35 kb region was confirmed by the two-step polymerase chain reaction. The deletion and complementation test revealed that the Int1157 integrase, a tyrosine recombinase, mediates the excision of this region. Unlike most tyrosine recombinases, Int1157 is suggested to recognize diverse sites and mediate recombination between non-homologous DNA sequences. This is the first report of non-homologous recombination mediating flagellar phase variation.
Collapse
Affiliation(s)
- Bin Liu
- TEDA School of Biological Sciences and Biotechnology, Nankai University, 23 Hongda Street, TEDA, Tianjin Key Laboratory of Microbial Functional Genomics, Tianjin Research Center for Functional Genomics and Biochip, Tianjin 300457 and The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Tianjin, P. R. China
| | - Bo Hu
- TEDA School of Biological Sciences and Biotechnology, Nankai University, 23 Hongda Street, TEDA, Tianjin Key Laboratory of Microbial Functional Genomics, Tianjin Research Center for Functional Genomics and Biochip, Tianjin 300457 and The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Tianjin, P. R. China
| | - Zhemin Zhou
- TEDA School of Biological Sciences and Biotechnology, Nankai University, 23 Hongda Street, TEDA, Tianjin Key Laboratory of Microbial Functional Genomics, Tianjin Research Center for Functional Genomics and Biochip, Tianjin 300457 and The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Tianjin, P. R. China
| | - Dan Guo
- TEDA School of Biological Sciences and Biotechnology, Nankai University, 23 Hongda Street, TEDA, Tianjin Key Laboratory of Microbial Functional Genomics, Tianjin Research Center for Functional Genomics and Biochip, Tianjin 300457 and The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Tianjin, P. R. China
| | - Xi Guo
- TEDA School of Biological Sciences and Biotechnology, Nankai University, 23 Hongda Street, TEDA, Tianjin Key Laboratory of Microbial Functional Genomics, Tianjin Research Center for Functional Genomics and Biochip, Tianjin 300457 and The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Tianjin, P. R. China
| | - Peng Ding
- TEDA School of Biological Sciences and Biotechnology, Nankai University, 23 Hongda Street, TEDA, Tianjin Key Laboratory of Microbial Functional Genomics, Tianjin Research Center for Functional Genomics and Biochip, Tianjin 300457 and The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Tianjin, P. R. China
| | - Lu Feng
- TEDA School of Biological Sciences and Biotechnology, Nankai University, 23 Hongda Street, TEDA, Tianjin Key Laboratory of Microbial Functional Genomics, Tianjin Research Center for Functional Genomics and Biochip, Tianjin 300457 and The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Tianjin, P. R. China
| | - Lei Wang
- TEDA School of Biological Sciences and Biotechnology, Nankai University, 23 Hongda Street, TEDA, Tianjin Key Laboratory of Microbial Functional Genomics, Tianjin Research Center for Functional Genomics and Biochip, Tianjin 300457 and The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Tianjin, P. R. China
- *To whom correspondence should be addressed. Tel: +86 22 66229588; Fax: +86 22 66229596;
| |
Collapse
|
39
|
Letran SE, Lee SJ, Atif SM, Flores-Langarica A, Uematsu S, Akira S, Cunningham AF, McSorley SJ. TLR5-deficient mice lack basal inflammatory and metabolic defects but exhibit impaired CD4 T cell responses to a flagellated pathogen. THE JOURNAL OF IMMUNOLOGY 2011; 186:5406-12. [PMID: 21451112 DOI: 10.4049/jimmunol.1003576] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
TLR5-deficient mice have been reported to develop spontaneous intestinal inflammation and metabolic abnormalities. However, we report that TLR5-deficient mice from two different animal colonies display no evidence of basal inflammatory disease, metabolic abnormalities, or enhanced resistance to Salmonella infection. In contrast, the absence of TLR5 hindered the initial activation and clonal expansion of intestinal flagellin-specific CD4 T cells following oral Salmonella infection. Together, these data demonstrate that a basal inflammatory phenotype is not a consistent feature of TLR5-deficient mice and document a novel role for TLR5 in the rapid targeting of flagellin by intestinal pathogen-specific CD4 T cells.
Collapse
Affiliation(s)
- Shirdi E Letran
- Center for Infectious Diseases and Microbiology Translational Research, McGuire Translational Research Facility, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Cho HY, Choi EK, Lee SW, Kim KH, Park SJ, Lee CK, Lee SW. All-trans retinoic acid induces TLR-5 expression and cell differentiation and promotes flagellin-mediated cell functions in human THP-1 cells. Immunol Lett 2011; 136:97-107. [PMID: 21237205 DOI: 10.1016/j.imlet.2011.01.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2010] [Revised: 12/16/2010] [Accepted: 01/03/2011] [Indexed: 11/18/2022]
Abstract
Toll-like receptor 5 (TLR-5), which is expressed on macrophages and dendritic cells (DCs), is a crucial cell surface molecule that senses microbial-associated molecular patterns and initiates host innate immune responses upon infection with invaders that express flagellin. Little information is known about the induction factors and mechanisms of TLR-5 expression. In this study, we demonstrate that all-trans retinoic acid (ATRA) significantly up-regulated TLR-5 expression in human macrophage THP-1 cells by co-activating NF-κB and the RARα receptor and inducing the differentiation of CD11b(-)CD11c(-) THP-1 cells to CD11b(+)CD11c(low) cells. Furthermore, when stimulated with flagellin, ATRA-induced THP-1 cells expressed multiple cytokines, including TNF-α, IL-1beta, and IL-12p40, and several co-stimulatory molecules, such as CD40, CD80, CD86, and MHC class I and II. We also showed that when ATRA-induced THP-1 cells were stimulated with flagellin, the cells displayed an allostimulatory capacity rather than phagocytic activity. Taken together, our findings suggest that ATRA is a crucial immunostimulatory cofactor that induces the activation of macrophages and their subsequent differentiation into dendritic-like cells.
Collapse
Affiliation(s)
- Hae-Yun Cho
- Advanced Research Center for Multiple Myeloma, Department of Microbiology, College of Medicine, Inje University, Busan 614-735, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
41
|
Mizel SB, Bates JT. Flagellin as an adjuvant: cellular mechanisms and potential. THE JOURNAL OF IMMUNOLOGY 2010; 185:5677-82. [PMID: 21048152 DOI: 10.4049/jimmunol.1002156] [Citation(s) in RCA: 301] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Flagellin is a potent activator of a broad range of cell types involved in innate and adaptive immunity. An increasing number of studies have demonstrated the effectiveness of flagellin as an adjuvant, as well as its ability to promote cytokine production by a range of innate cell types, trigger a generalized recruitment of T and B lymphocytes to secondary lymphoid sites, and activate TLR5(+)CD11c(+) cells and T lymphocytes in a manner that is distinct from cognate Ag recognition. The plasticity of flagellin has allowed for the generation of a range of flagellin-Ag fusion proteins that have proven to be effective vaccines in animal models. This review summarizes the state of our current understanding of the adjuvant effect of flagellin and addresses important areas of current and future research interest.
Collapse
Affiliation(s)
- Steven B Mizel
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA.
| | | |
Collapse
|
42
|
Dilmaghani M, Ahmadi M, Zahraei Salehi T, Talebi A, Darvishzadeh R. PCR-Restriction Fragment Length Polymorphism analysis of fljB gene in Salmonella enterica subspecies enterica serovar Typhimurium isolated from avians. IRANIAN JOURNAL OF MICROBIOLOGY 2010; 2:178-84. [PMID: 22347569 PMCID: PMC3279790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND AND OBJECTIVES Economic constraint of diseases arising from Salmonella Typhimurium causes the study of this zoonotic organism more important. Most studies on identification and characterization of S. Typhimurium are conducted at DNA level. Flagellin genes (fliC and fljB genes encoding phase-1 and phase-2 flagella, respectively) are useful as a model system for studying genetic differentiation. The objectives of the present study were to identify the polymorphism of fljB among avians in different regions by the PCR-RFLP method. MATERIALS AND METHODS Fifty-two S. Typhimurium isolates out of 1,870 intestine samples were identified using culture and serotyping as well as multiplex-PCR (broiler (n=13), layer (n=12), duck (n=5), goose (n=5), sparrow (n=8), canary (n=3), pigeon (n=5) and casco parrot (n=1)). Amplification of fljB gene was performed and amplified products subjected to restriction digestion with Hha I enzyme. RESULTS Two RFLP patterns generated DNA fragments between approximately 50 to 800 bps. Pattern A was observed in 33 (63.46%) and pattern B in 19 (36.54%) of isolates. Salmonella Typhimurium recovered from 13 broilers (ten with pattern A and 3 with pattern B) and 8 sparrow (three with pattern A and 5 with pattern B) showed both A and B patterns. Twelve layers, 5 pigeons and 3 canaries showed pattern A and 5 ducks, 5 geese and one casco parrot showed pattern B. None of these patterns was allotted for a special region. CONCLUSION The results of the present study showed that fljB gene is highly conserved among avians in different geographical regions, suggesting not only the importance of fljB gene in survival of organism in different environmental conditions but also the relation between proteins encoded by fljB gene and serotyping scheme.
Collapse
Affiliation(s)
- M Dilmaghani
- Department of Microbiology, Faculty of Veterinary Medicine, University of Urmia,Urmia, Iran
| | - M Ahmadi
- Department of Microbiology, Faculty of Veterinary Medicine, University of Urmia,Urmia, Iran
| | - T Zahraei Salehi
- Department of Microbiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - A Talebi
- Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Urmia,Urmia, Iran
| | - R Darvishzadeh
- Department of Agronomy and Plant breeding University of Urmia, Urmia, Iran
| |
Collapse
|
43
|
Braga CJM, Massis LM, Sbrogio-Almeida ME, Alencar BCG, Bargieri DY, Boscardin SB, Rodrigues MM, Ferreira LCS. CD8+ T cell adjuvant effects of Salmonella FliCd flagellin in live vaccine vectors or as purified protein. Vaccine 2009; 28:1373-82. [PMID: 19932669 DOI: 10.1016/j.vaccine.2009.11.003] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2009] [Revised: 11/01/2009] [Accepted: 11/03/2009] [Indexed: 12/21/2022]
Abstract
Salmonella flagellin, the flagellum structural subunit, has received particular interest as a vaccine adjuvant conferring enhanced immunogenity to soluble proteins or peptides, both for activation of antibody and cellular immune responses. In the present study, we evaluated the Salmonella enterica FliCd flagellin as a T cell vaccine adjuvant using as model the 9-mer (SYVPSAEQI) synthetic H2(d)-restricted CD8(+) T cell-specific epitope (CS(280-288)) derived from the Plasmodium yoelii circumsporozoite (CS) protein. The FliCd adjuvant effects were determined under two different conditions: (i) as recombinant flagella, expressed by orally delivered live S. Dublin vaccine strains expressing the target CS(280-288) peptide fused at the central hypervariable domain, and (ii) as purified protein in acellular vaccines in which flagellin was administered to mice either as a recombinant protein fused or admixed with the target CS(280-288) peptide. The results showed that CS(280-288)-specific cytotoxic CD8(+) T cells were primed when BALB/c mice were orally inoculated with the expressing the CS(280-288) epitope S. Dublin vaccine strain. In contrast, mice immunized with purified FliCd admixed with the CS(280-288) peptide and, to a lesser extent, fused with the target peptide developed specific cytotoxic CD8(+) T cell responses without the need of a heterologous booster immunization. The CD8(+) T cell adjuvant effects of flagellin, either fused or not with the target peptide, correlated with the in vivo activation of CD11c(+) dendritic cells. Taken together, the present results demonstrate that Salmonella flagellins are flexible adjuvant and induce adaptative immune responses when administered by different routes or vaccine formulations.
Collapse
Affiliation(s)
- Catarina J M Braga
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, Av. Prof. Lineu Prestes, 1374, São Paulo, SP 05008-000, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Hu Y, Wang Y, Ding L, Lu P, Atkinson S, Chen S. Positive regulation of flhDC expression by OmpR in Yersinia pseudotuberculosis. Microbiology (Reading) 2009; 155:3622-3631. [DOI: 10.1099/mic.0.030908-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
OmpR has been demonstrated to negatively regulate the expression of the flagellar master operon flhDC in a wide variety of bacterial species. Here we report the positive regulation of flhDC expression by OmpR in Yersinia pseudotuberculosis. A σ
70-dependent promoter was identified by primer extension analysis and an active region with two conserved OmpR-binding sites around the flhDC promoter was confirmed. To confirm the regulation of flhDC expression by OmpR, flhDC as well as the downstream flagellar genes fliA, flgD, flgA, flgM, fliC and flaA were fused to lacZ, and decreased expression of all these genes in an ompR mutant (ΔompR) was detected. Furthermore, ΔompR was defective in bacterial motility and flagella synthesis. This defect was due to the low level of expression of flhDC in ΔompR since overproduction of FlhDC in ΔompR restored bacterial motility. The importance of two conserved OmpR-binding sites around the flhDC promoter region in the regulation of flhDC expression by OmpR was demonstrated by the fact that mutation of either one or both sites significantly decreased the promoter activity in the wild-type but not in ΔompR. The binding of OmpR to these two sites was also demonstrated by DNA mobility shift assay. The possible mechanism underlying this positive regulation in Y. pseudotuberculosis is discussed. To our knowledge, this is the first report to demonstrate that OmpR positively regulates flhDC expression.
Collapse
Affiliation(s)
- Yangbo Hu
- Graduate School of the Chinese Academy of Sciences, Beijing 100049, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, the Chinese Academy of Sciences, Wuhan 430071, China
| | - Yao Wang
- State Key Laboratory of Virology, Wuhan Institute of Virology, the Chinese Academy of Sciences, Wuhan 430071, China
| | - Lisha Ding
- Graduate School of the Chinese Academy of Sciences, Beijing 100049, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, the Chinese Academy of Sciences, Wuhan 430071, China
| | - Pei Lu
- Graduate School of the Chinese Academy of Sciences, Beijing 100049, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, the Chinese Academy of Sciences, Wuhan 430071, China
| | - Steve Atkinson
- Institute of Infection, Immunity and Inflammation, Centre for Biomolecular Sciences, University of Nottingham, Nottingham NG7 2UH, UK
| | - Shiyun Chen
- State Key Laboratory of Virology, Wuhan Institute of Virology, the Chinese Academy of Sciences, Wuhan 430071, China
| |
Collapse
|
45
|
Ruan P, Xia XP, Sun D, Ojcius DM, Mao YF, Yue WY, Yan J. Recombinant SpaO and H1a as immunogens for protection of mice from lethal infection with Salmonella paratyphi A: implications for rational design of typhoid fever vaccines. Vaccine 2009; 26:6639-44. [PMID: 18834913 DOI: 10.1016/j.vaccine.2008.09.030] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2008] [Revised: 07/30/2008] [Accepted: 09/11/2008] [Indexed: 11/29/2022]
Abstract
The Vi capsular polysaccharide vaccine is one of two vaccines against typhoid recommended worldwide and is the vaccine generally used in China. However, in recent years a Salmonella paratyphi A strain that is naturally devoid of capsule has caused frequent outbreaks of typhoid fever in Southern China, leading to the need for identification of additional antigens that could be incorporated into new vaccines. SpaO acts as a major invasion factor of Salmonella enterica spp. and H1a is the unique flagellin subunit ofS. paratyphi A. In this study, the two prokaryotic recombinant antigens, rSpaO and rH1a, were expressed and their immunogenicity was demonstrated by the slide agglutination test and Western blot assays. Using PCR and sequencing analysis as well as ELISA, we find that the spaO and h1a genes are widely distributed in 196 S. paratyphi A isolates (97.5 and 100%, respectively), with high expression frequencies for the SpaO (98.0%) and H1a (100%) antigens. The two genes also show high sequence conservation (similarities from 99.31 to 99.88% for both genes). In sera from 172 paratyphoid A patients, anti-SpaO and anti-H1a IgGs were detectable by ELISA, in 94.8 and 98.8% of patients, respectively. Furthermore, 41.7-66.7% of mice immunized with rSpaO or rH1a alone were protected against subsequent infection, and the protection rate rose to 75.0-91.7% in mice co-immunized with the two antigens. As the spaO and h1a genes of S. paratyphi A are sequence conserved, extensively distributed and highly expressed, the rSpaO and rH1a immunogens should be considered in the development of novel vaccines to prevent S. paratyphi A-caused typhoid fever.
Collapse
Affiliation(s)
- Ping Ruan
- Division of Basic Medical Microbiology, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Zhejiang, Hangzhou 310003, PR China
| | | | | | | | | | | | | |
Collapse
|
46
|
Incorporation of membrane-anchored flagellin into influenza virus-like particles enhances the breadth of immune responses. J Virol 2008; 82:11813-23. [PMID: 18786995 DOI: 10.1128/jvi.01076-08] [Citation(s) in RCA: 107] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
We have designed a membrane-anchored form of the Toll-like receptor 5 ligand flagellin, the major proinflammatory determinant of enteropathogenic Salmonella, which was found to be glycosylated and expressed on cell surfaces. A chimeric influenza virus-like particle (cVLP) vaccine candidate containing A/PR8/34 (H(1)N(1)) hemagglutinin (HA), matrix protein (M1), and the modified flagellin as a molecular adjuvant was produced. The immunogenicity, including the serum antibody levels and cellular immune responses, and the protective efficacy against homologous and heterologous live virus challenge of the resulting VLPs were tested after intramuscular administration in a mouse model. The results demonstrated that flagellin-containing VLPs elicited higher specific immunoglobulin G (IgG) responses than standard HA and M1 VLPs, indicating the adjuvant effect of flagellin. Enhanced IgG2a and IgG2b but not IgG1 responses were observed with flagellin-containing VLPs, illuminating the activation of Th1 class immunity. The adjuvant effects of flagellin were also reflected by enhanced specific cellular responses revealed by the secretion of cytokines by freshly isolated splenocyte cultures when stimulated with pools of major histocompatibility complex class I or II peptides. When immunized mice were challenged with homologous live PR8 virus, complete protection was observed for both the standard and cVLP groups. However, when a heterosubtypic A/Philippines (H(3)N(2)) virus was used for challenge, all of the standard VLP group lost at least 25% of body weight, reaching the experimental endpoint. In contrast, for the cVLP group, 67% of mice survived the challenge infection. These results reveal that cVLPs designed by incorporating flagellin as a membrane-anchored adjuvant induce enhanced cross-protective heterosubtypic immune responses. They also indicate that such cVLP vaccines are a promising new approach for protection against pandemic influenza viruses.
Collapse
|
47
|
Sanders CJ, Moore DA, Williams IR, Gewirtz AT. Both radioresistant and hemopoietic cells promote innate and adaptive immune responses to flagellin. THE JOURNAL OF IMMUNOLOGY 2008; 180:7184-92. [PMID: 18490717 DOI: 10.4049/jimmunol.180.11.7184] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The TLR5 agonist flagellin induces innate and adaptive immune responses in a MyD88-dependent manner and is under development as a vaccine adjuvant. In vitro studies indicate that, compared with other bacteria-derived adjuvants, flagellin is a very potent activator of proinflammatory gene expression and cytokine production from cells of nonhemopoietic origin. However, the role of nonhemopoietic cells in promoting flagellin-induced immune responses in vivo remains unclear. To investigate the relative contributions of the nonhemopoietic (radioresistant) and the hemopoietic (radiosensitive) compartments, we measured both innate and adaptive immune responses of flagellin-treated MyD88 radiation bone marrow chimeras. We observed that radiosensitive and radioresistant cells played distinct roles in the innate response to flagellin, with the radiosensitive cells producing the majority of the TNF-alpha, IL-12, and IL-6 cytokines and the radioresistant cells most of the KC, IP-10, and MCP-1 cytokines. Direct activation of either compartment alone by flagellin initiated dendritic cell costimulatory molecule up-regulation and induced a significant humoral immune response to the protein itself as well as to coinjected OVA. However, robust humoral responses were only observed when MyD88 was present in both cell compartments. Further studies revealed that hemopoietic and nonhemopoietic expression of the cytokines TNF-alpha and IL-6, but not IL-1, played an important role in promoting flagellin-induced Ab responses. Thus, in vivo both radioresistant and hemopoietic cells play key nonredundant roles in mediating innate and adaptive immune responses to flagellin.
Collapse
|
48
|
Bandyopadhaya A, Sarkar M, Chaudhuri K. IL-1β expression in Int407 is induced by flagellin of Vibrio cholerae through TLR5 mediated pathway. Microb Pathog 2008; 44:524-36. [DOI: 10.1016/j.micpath.2008.01.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2007] [Revised: 01/10/2008] [Accepted: 01/11/2008] [Indexed: 02/05/2023]
|
49
|
Le Moigne V, Robreau G, Mahana W. Flagellin as a good carrier and potent adjuvant for Th1 response: Study of mice immune response to the p27 (Rv2108) Mycobacterium tuberculosis antigen. Mol Immunol 2008; 45:2499-507. [DOI: 10.1016/j.molimm.2008.01.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2007] [Revised: 01/04/2008] [Accepted: 01/08/2008] [Indexed: 10/22/2022]
|
50
|
Abstract
Flagella are locomotive organelles present on a wide range of bacteria and are important for the pathogenesis of many species. Cells of the innate immune system lack memory per se, but recognize conserved pathogen-associated molecular patterns (PAMPs) through a family of type I membrane receptors known as Toll-like receptors (TLRs). Flagellin, the major structural component of flagella, is a highly conserved protein recognized in hosts by TLR5. Signaling of flagellin via TLR5/TLR4 heteromeric complexes enhances the diversity of the response, likely by engaging MyD88-independent adaptors to activate the interferon pathway. Flagellin is a potent immune activator, stimulating diverse biologic effects that mediate both innate inflammatory responses as well as the development of adaptive immunity. Binding of flagellin to the extracellular domain of TLR5 rapidly induces a signal cascade that culminates in the production of proinflammatory mediators such as cytokines, chemokines, and costimulatory molecules. This review focuses on the mechanisms of action of flagellin and its effects on both innate and adaptive immunity.
Collapse
Affiliation(s)
- Anna N Honko
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, USA
| | | |
Collapse
|