1
|
Sun Z, Wang X, Shi C, Yu T, Xu W, Ji X, Su K, Yan H, Shan Y, Xie T, Xu J, Zhao X, Shan J. TREM2 modulates lipid metabolism to alleviate airway inflammation in asthma: A potential therapeutic target. Int J Biol Macromol 2025; 308:142306. [PMID: 40154695 DOI: 10.1016/j.ijbiomac.2025.142306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 02/23/2025] [Accepted: 03/18/2025] [Indexed: 04/01/2025]
Abstract
Asthma is a chronic inflammatory disease characterized by airway hyperresponsiveness (AHR) and immune cell infiltration. TREM2 (Triggering Receptor Expressed on Myeloid cells 2), known for its role in lipid metabolism and inflammation, was found to be upregulated in asthma. Using Trem2-/- mice, we observed that TREM2 deletion significantly reduces airway inflammation and AHR. This effect is achieved through the modulation of triglyceride (TG) metabolism, leading to increased TG synthesis, decreased lipolysis, and reduced release of pro-inflammatory free fatty acids (FFAs), particularly arachidonic acid. The study reveals a novel role for TREM2 in regulating lipid metabolism in asthma, suggesting that targeting TREM2 may offer new therapeutic opportunities for managing asthma and related inflammatory conditions.
Collapse
Affiliation(s)
- Zhengpeng Sun
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China; Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Xuan Wang
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China; Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Chen Shi
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China; Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Tao Yu
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China; Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Weichen Xu
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China; Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Xinyu Ji
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China; Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Ke Su
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China; Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Hua Yan
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China; Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Yiwen Shan
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China; Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Tong Xie
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China; Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Jianya Xu
- Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China; School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Xia Zhao
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China; Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China.
| | - Jinjun Shan
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China; Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China.
| |
Collapse
|
2
|
Korpi F, Irajian G, Forouhi F, Mohammadian T. A chimeric vaccine targeting Pseudomonas aeruginosa virulence factors protects mice against lethal infection. Microb Pathog 2023; 178:106033. [PMID: 36813005 DOI: 10.1016/j.micpath.2023.106033] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 01/25/2023] [Accepted: 02/12/2023] [Indexed: 02/22/2023]
Abstract
Pseudomonas aeruginosa is an important and hazardous nosocomial pathogen in respiratory tract infections and rapidly achieves antibiotic resistance, so it is necessary to develop an effective vaccine to combat the infection. The Type III secretion system (T3SS) protein P. aeruginosa V-antigen (PcrV), outer membrane protein F (OprF), and two kinds of flagellins (FlaA and FlaB) all play important roles in the pathogenesis of P. aeruginosa lung infection and its spread into deeper tissues. In a mouse acute pneumonia model, the protective effects of a chimer vaccine including PcrV, FlaA, FlaB, and OprF (PABF) protein were investigated. PABF immunization prompted robust opsonophagocytic titer of IgG antibodies and decreased bacterial burden, and improved survival afterward intranasal challenge with ten times 50% lethal doses (LD50) of P. aeruginosa strains, indicating its broad-spectrum immunity. Moreover, these findings showed a promise chimeric vaccine candidate to treat and control P. aeruginosa infections.
Collapse
Affiliation(s)
- Fatemeh Korpi
- Department of Cell and Molecular Biology, Faculty of Basic Science, Islamic Azad University Shahre Qods Branch, Iran
| | - Gholamreza Irajian
- Department of Cell and Molecular Biology, Faculty of Basic Science, Islamic Azad University Shahre Qods Branch, Iran; Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Fatemeh Forouhi
- Department of Cell and Molecular Biology, Faculty of Basic Science, Islamic Azad University Shahre Qods Branch, Iran
| | - Taher Mohammadian
- Department of Cell and Molecular Biology, Faculty of Basic Science, Islamic Azad University Shahre Qods Branch, Iran
| |
Collapse
|
3
|
Moshref Javadi M, Taghdisi Hosseinzadeh M, Soleimani N, Rommasi F. Evaluating the immunogenicity of gold nanoparticles conjugated RBD with Freund's adjuvant as a potential vaccine against SARS-CoV-2. Microb Pathog 2022; 170:105687. [PMID: 35917987 PMCID: PMC9339102 DOI: 10.1016/j.micpath.2022.105687] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/31/2022] [Accepted: 07/19/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND and Introduction: SARS-CoV-2 is currently considered as the most challenging issue in the field of health and medicine by causing a global pandemic. Vaccines are counted as a promising candidate to terminate this deadly pandemic. Various structural proteins in SARS-CoV-2 have recently drawn attention to be utilized as candidate vaccines to stimulate immune responses against COVID-19. MATERIALS AND METHODS In current study, the RBD protein was cloned and expressed in E. coli host. Then, the expressed RBD protein was purified and its characterizations were evaluated through various methods. Gold nanoparticles, which were utilized as a carrier for candidate Nano-vaccine, were synthesized via oxidation-reduction reaction. While Gold NPs-conjugated RBD was injected into the second treatment group, in the first candidate vaccine, RBD was injected into the first treatment group solely. Complete and Incomplete Freud's Adjuvant were also utilized for both treatment groups to enhance the immune responses against RBD antigen. Immunizations were repeated 2 times in 14-day intervals to boost the immune system of BALB/c mice. The humoral and cell-mediated immune responses were examined through immune and cytokine assays. RESULTS Our outcomes demonstrate that strong short-term humoral immunity (IgM) was induced in both the first and second treatment group, while long-term humoral responses (IgG) were only observed in the second treatment group. While stronger short- and long-term humoral (IgM and IgG, respectively) were observed in the second treatment group, particular cytokines production (TNF-ɑ and IFN-γ) as a marker of cell-mediated responses were significantly higher in the first treatment group. DISCUSSION AND CONCLUSION Our study results show the high potentiality of RBD protein as an appropriate stimulating antigen in vaccine synthesis and testifies RBD-based candidate vaccines to control the COVID-19 pandemic. Our outcomes also recommend that Nano-vaccines can be more suitable candidates when stronger long-term immune responses matter.
Collapse
Affiliation(s)
- Mahtab Moshref Javadi
- Department of Microbiology and Microbial Biotechnology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Mozhgan Taghdisi Hosseinzadeh
- Department of Microbiology and Microbial Biotechnology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Neda Soleimani
- Department of Microbiology and Microbial Biotechnology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran.
| | - Foad Rommasi
- Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| |
Collapse
|
4
|
Tarrah A, Dos Santos Cruz BC, Sousa Dias R, da Silva Duarte V, Pakroo S, Licursi de Oliveira L, Gouveia Peluzio MC, Corich V, Giacomini A, Oliveira de Paula S. Lactobacillus paracasei DTA81, a cholesterol-lowering strain having immunomodulatory activity, reveals gut microbiota regulation capability in BALB/c mice receiving high-fat diet. J Appl Microbiol 2021; 131:1942-1957. [PMID: 33709536 PMCID: PMC8518695 DOI: 10.1111/jam.15058] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 01/02/2021] [Accepted: 03/08/2021] [Indexed: 12/24/2022]
Abstract
Aims In‐vitro/In‐vivo evaluation of cholesterol‐lowering probiotic strain Lactobacillus paracasei DTA81 and the possible connection with the gut microbiota modulation. Methods and Results In the present study, strain DTA81 has been evaluated for the possible influence on blood lipid and glucose concentrations, modulation of the immune system, gastrointestinal survivability and modulation of gut microbiota in BALB/c mice receiving a high‐fat diet. After 6 weeks of treatment, a significant reduction of total cholesterol and fasting blood sugar (FBS) among animals treated with L. paracasei DTA81 has been recorded. Comparison of colon tissue levels of different cytokines revealed a significant reduction of the inflammatory cytokine interleukin‐6. The comparison of gut microbiota using the 16S rRNA approach indicated that the treatment with L. paracasei DTA81 significantly increased the taxa Bacteroidetes and Coprococcus. Moreover, the genome of DTA81 was sequenced for the in‐silico assessment, and the analysis indicated the presence of cholesterol assimilation‐related genes as well as the absence of negative traits such as transmissible antibiotic resistance genes, plasmids and prophage regions. Conclusion The outcome of this study revealed the in‐vitro and in‐vivo properties of L. paracasei DTA81 and the possible mechanism between consumption of this strain, the abundance of Bacteriodetes/Coprococcus taxa, immunomodulatory activity and the subsequent reduction of cholesterol/FBS in BALB/c mice. Significance and Impact of the Study Lactobacillus paracasei DTA81 as a non‐pharmacological potential probiotic supplement can influence metabolic homeostasis in individuals, particularly those adopting high‐fat diets, and it can contribute to reduce coronary heart disease.
Collapse
Affiliation(s)
- A Tarrah
- Department of Agronomy Food Natural Resources Animals and Environment, University of Padova, Viale dell'Universitá, Italy
| | - B C Dos Santos Cruz
- Department of Nutrition and Health, Federal University of Viçosa, Viçosa, Minas Gerais, Brazil
| | - R Sousa Dias
- Department of General Biology, Federal University of Viçosa, Viçosa, Minas Gerais, Brazil
| | - V da Silva Duarte
- Department of Agronomy Food Natural Resources Animals and Environment, University of Padova, Viale dell'Universitá, Italy
| | - S Pakroo
- Department of Agronomy Food Natural Resources Animals and Environment, University of Padova, Viale dell'Universitá, Italy
| | - L Licursi de Oliveira
- Department of General Biology, Federal University of Viçosa, Viçosa, Minas Gerais, Brazil
| | - M C Gouveia Peluzio
- Department of Nutrition and Health, Federal University of Viçosa, Viçosa, Minas Gerais, Brazil
| | - V Corich
- Department of Agronomy Food Natural Resources Animals and Environment, University of Padova, Viale dell'Universitá, Italy
| | - A Giacomini
- Department of Agronomy Food Natural Resources Animals and Environment, University of Padova, Viale dell'Universitá, Italy
| | - S Oliveira de Paula
- Department of General Biology, Federal University of Viçosa, Viçosa, Minas Gerais, Brazil
| |
Collapse
|
5
|
Hashemi FB, Behrouz B, Irajian G, Laghaei P, Korpi F, Fatemi MJ. A trivalent vaccine consisting of "flagellin A+B and pilin" protects against Pseudomonas aeruginosa infection in a murine burn model. Microb Pathog 2019; 138:103697. [PMID: 31465785 DOI: 10.1016/j.micpath.2019.103697] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 02/22/2019] [Accepted: 08/26/2019] [Indexed: 10/26/2022]
Abstract
Pseudomonas aeruginosa is a common nosocomial pathogen in burn patients, and rapidly achieves antibiotic resistance, and thus, developing an effective vaccine is critically important for combating P. aeruginosa infection. Flagella and pili play important roles in colonization of P. aeruginosa at the burn wound site and its subsequent dissemination to deeper tissue and organs. In the present study, we evaluated protective efficacy of a trivalent vaccine containing flagellins A and B (FlaA + FlaB) + pilin (PilA) in a murine burn model of infection. "FlaA + FlaB + PilA" induced greater protection in P. aeruginosa murine burn model than the single components alone, and it showed broad immune protection against P. aeruginosa strains. Immunization with "FlaA + FlaB + PilA" induced strong opsonophagocytic antibodies and resulted in reduced bacterial loads, systemic IL-12/IL-10 cytokine expression, and increased survival after challenge with three times lethal dose fifty (LD50) of P. eruginosa strains. Moreover, the protective efficacy of "FlaA + FlaB + PilA" vaccination was largely attributed to specific antibodies. Taken together, these data further confirm that the protective effects of "FlaA + FlaB + PilA" vaccine significantly enhance efficacy compared with antibodies against either mono or divalent antigen, and that the former broadens the coverage against P. eruginosa strains that express two of the three antigens.
Collapse
Affiliation(s)
- Farhad B Hashemi
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Bahador Behrouz
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Burn Research Center, Hazrat Fatima Hospital, Iran University of Medical Sciences, Tehran, Iran.
| | - Gholamreza Irajian
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Parisa Laghaei
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Korpi
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Javad Fatemi
- Burn Research Center, Hazrat Fatima Hospital, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
6
|
TSLP-activated dendritic cells induce T helper type 2 inflammation in Aspergillus fumigatus keratitis. Exp Eye Res 2018; 171:120-130. [DOI: 10.1016/j.exer.2018.03.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 03/08/2018] [Accepted: 03/13/2018] [Indexed: 11/19/2022]
|
7
|
Dendritic cells that highly express SOCS1 induce T-cell hypo-responsiveness and prolong islet allograft survival. Cell Immunol 2017; 314:36-41. [DOI: 10.1016/j.cellimm.2017.01.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Revised: 01/07/2017] [Accepted: 01/27/2017] [Indexed: 11/17/2022]
|
8
|
Banadkoki AZ, Keshavarzmehr M, Afshar Z, Aleyasin N, Fatemi MJ, Behrouz B, Hashemi FB. Protective effect of pilin protein with alum+naloxone adjuvant against acute pulmonary Pseudomonas aeruginosa infection. Biologicals 2016; 44:367-73. [PMID: 27427517 DOI: 10.1016/j.biologicals.2016.06.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Revised: 05/29/2016] [Accepted: 06/21/2016] [Indexed: 11/26/2022] Open
Abstract
Pseudomonas aeruginosa is an important opportunistic human pathogen that causes a wide variety of severe nosocomial infections. Type IV pili of P. aeruginosa are made up of polymerized pilin that aids in bacterial adhesion, biofilm formation and twitching motility. The aim of this study was to evaluate the efficacy of alum and naloxone (alum+NLX) as an adjuvant for P. aeruginosa recombinant PilA (r-PilA) as a vaccine candidate in the improvement of humoral and cellular immunity. Primary immunization with r-PilA in combination with alum+NLX followed by two booster shots was sufficient to generate robust cellular and humoral responses, which were Th1 and Th2 type responses consisting of IgG1 and IgG2a subtypes. Analysis of the cytokine response among immunized mice showed an increased production of IL-4, INF-γ and IL-17 by splenocytes upon stimulation by r-PilA. These sera were also able to reduce bacterial load in the lung tissue of challenged mice. The reduction of systemic bacterial spread resulted in increased survival rates in challenged immunized mice. In conclusion, immunization with r-PilA combined with alum+NLX evokes cellular and humoral immune responses, which play an important role in providing protection against acute P. aeruginosa lung infection among immunized mice.
Collapse
Affiliation(s)
- Abbas Zare Banadkoki
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Quality Control Department of Iran Gelatin Capsule Mfg. Co., Tehran, Iran
| | | | - Zahra Afshar
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Neda Aleyasin
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Javad Fatemi
- Burn Research Center, Hazrat Fatima Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Bahador Behrouz
- Burn Research Center, Hazrat Fatima Hospital, Iran University of Medical Sciences, Tehran, Iran; Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Farhad B Hashemi
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
9
|
Immunogenicity and protective efficacy of Pseudomonas aeruginosa type a and b flagellin vaccines in a burned mouse model. Mol Immunol 2016; 74:71-81. [DOI: 10.1016/j.molimm.2016.04.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2015] [Revised: 04/07/2016] [Accepted: 04/14/2016] [Indexed: 12/31/2022]
|
10
|
Korpi F, Hashemi FB, Irajian G, Fatemi MJ, Laghaei P, Behrouz B. Flagellin and pilin immunization against multi-drug resistant Pseudomonas aeruginosa protects mice in the burn wound sepsis model. Immunol Lett 2016; 176:8-17. [PMID: 27210422 DOI: 10.1016/j.imlet.2016.04.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 03/26/2016] [Accepted: 04/02/2016] [Indexed: 01/15/2023]
Abstract
Pseudomonas aeruginosa is a formidable pathogen and a major threat to burn patients. Antimicrobial therapy is often unsuccessful because P. aeruginosa can develop multi-drug resistance; thus, immunotherapy and vaccine can be a rational alternative. Flagella and type IV pili have been identified as important virulence factors in the colonization and pathogenesis of P. aeruginosa in burn wound infections. Immunogenicity and efficacy of mixed recombinant full-length type b flagellin (r-b-flagellin) and recombinant PilA (r-PilA) as candidate vaccines were assessed by measuring humoral and cellular responses, using an experimental burned mouse model. Primary immunization with "r-b-flagellin+r-PilA" followed by two booster shots was sufficient to generate a robust humoral response, which was predominantly a Th2 response consisting mainly of subtype IgG1 and low levels of IgG2a. Analysis of the cytokine response among immunized mice showed an increased production of IL-4, INF-γ and IL-17 by splenocytes upon stimulation by "r-b-flagellin+r-PilA". Opsonophagocytosis assays confirmed the enhanced killing of bacteria by anti "r-b-flagellin+r-PilA" immune sera. These antibodies were also able to reduce bacterial load in the site of original infection into the liver and spleen of challenged mice. The reduction of systemic bacterial spread resulted in an increased survival rate of challenged immunized mice. In conclusion, immunization with "r-b-flagellin+r-PilA" proteins provides a better protective response against P. aeruginosa infection in the burn mouse model.
Collapse
Affiliation(s)
- Fatemeh Korpi
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Farhad B Hashemi
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Gholamreza Irajian
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Mohammad Javad Fatemi
- Burn Research Center, Motahari Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Parisa Laghaei
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Bahador Behrouz
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Burn Research Center, Motahari Hospital, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
11
|
Behrouz B, Mahdavi M, Amirmozafari N, Fatemi MJ, Irajian G, Bahroudi M, Hashemi FB. Immunogenicity of Pseudomonas aeruginosa recombinant b-type fagellin as a vaccine candidate: Protective efficacy in a murine burn wound sepsis model. Burns 2016:S0305-4179(16)30028-6. [PMID: 27156804 DOI: 10.1016/j.burns.2016.03.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 03/16/2016] [Accepted: 03/23/2016] [Indexed: 01/09/2023]
Abstract
Pseudomonas aeruginosa (PA) is a formidable opportunistic pathogen among patients with burn wound infections. Antimicrobial therapy is often unsuccessful because PA can develop multi-drug resistance; thus, immunotherapy can be a rational alternative. The goal of this study was to evaluate the immunogenicity recombinant type b flagellin (r-b-flagellin) as a potential vaccine against P. aeruginosa in a mouse model for burn wound sepsis. Primary immunization with r-b-flagellin (10μg) followed by two booster shots was sufficient to generate a robust humoral response, which was predominantly a T helper 2 (Th2) type response consisting mainly of subtype IgG1 and low levels of IgG2a. Analysis of the Th1-Th2 response among immunized mice showed an increased production of IL-4, INF-γ and IL-17 by splenocytes upon stimulation by r-b-flagellin. Opsono-phagocytosis assays confirmed the enhanced killing of bacteria by anti r-b-flagellin immune sera. These antibodies were also able to inhibit motility of P. aeruginosa and afforded protection to immunized mice by reducing bacterial load in the site of original infection into the liver of challenged mice. The reduction of systemic bacterial spread resulted in an increase in the survival rate of challenged immunized mice. In conclusion, immunization of mice with r-b-flagellin protein increased the level of humoral and cellular immune response and led to an efficacious protection against P. aeruginosa infection in the burn mouse model.
Collapse
Affiliation(s)
- Bahador Behrouz
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehdi Mahdavi
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
| | - Nour Amirmozafari
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Javad Fatemi
- Department of Plastic and Reconstructive Surgery, Burn Research Center, Hazrat Fatima Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Gholamreza Irajian
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mahbubeh Bahroudi
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Farhad B Hashemi
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
12
|
Kwak BR, Bäck M, Bochaton-Piallat ML, Caligiuri G, Daemen MJAP, Davies PF, Hoefer IE, Holvoet P, Jo H, Krams R, Lehoux S, Monaco C, Steffens S, Virmani R, Weber C, Wentzel JJ, Evans PC. Biomechanical factors in atherosclerosis: mechanisms and clinical implications. Eur Heart J 2014; 35:3013-20, 3020a-3020d. [PMID: 25230814 DOI: 10.1093/eurheartj/ehu353] [Citation(s) in RCA: 339] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Blood vessels are exposed to multiple mechanical forces that are exerted on the vessel wall (radial, circumferential and longitudinal forces) or on the endothelial surface (shear stress). The stresses and strains experienced by arteries influence the initiation of atherosclerotic lesions, which develop at regions of arteries that are exposed to complex blood flow. In addition, plaque progression and eventually plaque rupture is influenced by a complex interaction between biological and mechanical factors-mechanical forces regulate the cellular and molecular composition of plaques and, conversely, the composition of plaques determines their ability to withstand mechanical load. A deeper understanding of these interactions is essential for designing new therapeutic strategies to prevent lesion development and promote plaque stabilization. Moreover, integrating clinical imaging techniques with finite element modelling techniques allows for detailed examination of local morphological and biomechanical characteristics of atherosclerotic lesions that may be of help in prediction of future events. In this ESC Position Paper on biomechanical factors in atherosclerosis, we summarize the current 'state of the art' on the interface between mechanical forces and atherosclerotic plaque biology and identify potential clinical applications and key questions for future research.
Collapse
Affiliation(s)
- Brenda R Kwak
- Department of Pathology and Immunology, University of Geneva, CMU, Rue Michel-Servet 1, CH-1211 Geneva, Switzerland
| | | | | | | | | | | | - Imo E Hoefer
- University Medical Center Urecht, Utrecht, The Netherlands
| | | | | | | | | | | | | | | | | | | | - Paul C Evans
- Department of Cardiovascular Science, Medical School, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK
| |
Collapse
|
13
|
Differential Effects of Naja naja atra Venom on Immune Activity. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2014; 2014:287631. [PMID: 25024726 PMCID: PMC4082923 DOI: 10.1155/2014/287631] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Revised: 05/15/2014] [Accepted: 05/21/2014] [Indexed: 11/21/2022]
Abstract
Previous studies reported that Naja naja atra venom (NNAV) inhibited inflammation and adjuvant arthritis. Here we investigated the role of NNAV in regulation of immune responses in mice. Oral administration of NNAV to normal mice showed significant increase in natural killer cell activity, B lymphocyte proliferation stimulated by lipopolysaccharides, and antibody production in response to sheep red blood cells. Meanwhile, NNAV markedly decreased T lymphocyte proliferation stimulated by concanavalin A, arrested the cell cycle at G0/G1 phase, and suppressed CD4 and CD8 T cell divisions. Furthermore, NNAV inhibited the dinitrofluorobenzene-induced delayed-type hypersensitivity reaction. This modulation of immune responses may be partly attributed to the selective increase in Th1 and Th2 cytokines (IFN-γ, IL-4) secretion and inhibition of Th17 cytokine (IL-17) production. In dexamethasone-induced immunosuppressed mice, NNAV restored the concentration of serum IgG and IgM, while decreasing the percentage of CD4 and CD8 T-cell subsets. These results indicate that NNAV enhances the innate and humoral immune responses while inhibiting CD4 Th17 and CD8 T cell actions, suggesting that NNAV could be a potential therapeutic agent for autoimmune diseases.
Collapse
|
14
|
Arab S, Mojarrad M, Motamedi M, Mirzaei R, Modarressi MH, Hadjati J. Tumour regression induced by co-administration of MIP-3α and CpG in an experimental model of colon carcinoma. Scand J Immunol 2013; 78:28-34. [PMID: 23672351 DOI: 10.1111/sji.12058] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Accepted: 03/25/2013] [Indexed: 01/19/2023]
Abstract
CCL20/macrophage inflammatory protein-3α (MIP-3α) represents one of the potent chemoattractive proteins for dendritic cells (DCs). Herein, we investigated whether in vivo genetic modification of tumour cells aimed at intratumoural production of MIP-3α might lead to accumulation of DCs in tumour tissue. Mice injected with CT26, received recombinant adenovirus (Ad) vectors (AdMIP-3α) expressing MIP-3α protein. This was complemented by injections of CpG. Interestingly, MIP-3α gene therapy combined with CpG injections resulted in specific cytotoxicity. This was associated with significant suppression of tumour growth rate. These findings demonstrate the potential of strategies that utilize in vivo overexpression of chemokines.
Collapse
Affiliation(s)
- S Arab
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | | | | | | |
Collapse
|
15
|
Cavel O, Shomron O, Shabtay A, Vital J, Trejo-Leider L, Weizman N, Krelin Y, Fong Y, Wong RJ, Amit M, Gil Z. Endoneurial macrophages induce perineural invasion of pancreatic cancer cells by secretion of GDNF and activation of RET tyrosine kinase receptor. Cancer Res 2012; 72:5733-43. [PMID: 22971345 DOI: 10.1158/0008-5472.can-12-0764] [Citation(s) in RCA: 129] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Perineural invasion of cancer cells (CPNI) is found in most patients with pancreatic adenocarcinomas (PDA), prostate, or head and neck cancers. These patients undergo palliative rather than curative treatment due to dissemination of cancer along nerves, well beyond the extent of any local invasion. Although CPNI is a common source of distant tumor spread and a cause of significant morbidity, its exact mechanism is undefined. Immunohistochemical analysis of specimens excised from patients with PDAs showed a significant increase in the number of endoneurial macrophages (EMΦ) that lie around nerves invaded by cancer compared with normal nerves. Video microscopy and time-lapse analysis revealed that EMΦs are recruited by the tumor cells in response to colony-stimulated factor-1 secreted by invading cancer cells. Conditioned medium (CM) of tumor-activated EMΦs (tEMΦ) induced a 5-fold increase in migration of PDA cells compared with controls. Compared with resting EMΦs, tEMΦs secreted higher levels of glial-derived neurotrophic factor (GDNF), inducing phosphorylation of RET and downstream activation of extracellular signal-regulated kinases (ERK) in PDA cells. Genetic and pharmacologic inhibition of the GDNF receptors GFRA1 and RET abrogated the migratory effect of EMΦ-CM and reduced ERK phosphorylation. In an in vivo CPNI model, CCR2-deficient mice that have reduced macrophage recruitment and activation showed minimal nerve invasion, whereas wild-type mice developed complete sciatic nerve paralysis due to massive CPNI. Taken together, our results identify a paracrine response between EMΦs and PDA cells that orchestrates the formation of cancer nerve invasion.
Collapse
Affiliation(s)
- Oren Cavel
- The Laboratory for Applied Cancer Research, Department of Pathology, Tel Aviv Medical Center, Tel Aviv University, Tel Aviv, Israel
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Peluso L, de Luca C, Bozza S, Leonardi A, Giovannini G, Lavorgna A, De Rosa G, Mascolo M, Ortega De Luna L, Catania MR, Romani L, Rossano F. Protection against Pseudomonas aeruginosa lung infection in mice by recombinant OprF-pulsed dendritic cell immunization. BMC Microbiol 2010; 10:9. [PMID: 20070893 PMCID: PMC2820439 DOI: 10.1186/1471-2180-10-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2009] [Accepted: 01/13/2010] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The Pseudomonas aeruginosa major constitutive outer membrane porin protein F (OprF) has been shown to be a protective antigen and was previously used to activate an immunological response in a mouse model of lung pneumonia. The purpose of our study was to demonstrate the ability of mouse dendritic cells pulsed with purified or recombinant OprF to protect mice against P. aeruginosa infection and inflammation.Both native (n-OprF), isolated and purified from PAO1 bacterial strain, and recombinant (histidin-conjugated) OprF (His-OprF), obtained by cloning of the oprF gene into the pET28a expression vector, were used to stimulate dendritic cells in vitro before adoptive transfer into prospective recipient mice with P. aeruginosa pulmonary infection. RESULTS Similar to n-OprF, His-OprF activated dendritic cells in vitro, inducing the costimulatory molecule expression as well as cytokine production. Upon adoptive transfer in vivo, porin-pulsed dendritic cells (DCs) induced Th1-mediated resistance to infection and associated inflammatory pathology caused by either the PAO1 strain or a clinically-isolated mucoid strain. CONCLUSIONS This study highlights the pivotal contribution of DCs to vaccine-induced protection against P. aeruginosa infection and associated inflammation.
Collapse
Affiliation(s)
- Lucia Peluso
- Department of Cellular and Molecular Biology and Pathology L, Califano, University of Naples Federico II, Via S Pansini 5, 80131 Naples, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Damayanti T, Kikuchi T, Zaini J, Daito H, Kanehira M, Kohu K, Ishii N, Satake M, Sugamura K, Nukiwa T. Serial OX40 engagement on CD4+ T cells and natural killer T cells causes allergic airway inflammation. Am J Respir Crit Care Med 2009; 181:688-98. [PMID: 20019337 DOI: 10.1164/rccm.200910-1598oc] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
RATIONALE OX40-OX40 ligand (OX40L) interactions have been proposed to support induction of allergic airway inflammation, which may be attributable to OX40 signaling in CD4(+) helper T cells for adaptive immune responses. However, a possible involvement of natural killer T (NKT) cells in the pathogenesis suggests that the underlying mechanisms are not yet fully elucidated. OBJECTIVES We aimed to characterize the OX40-modulated cellular contribution to allergic airway inflammation in a mouse model of house dust mite (HDM) allergen exposure. METHODS Mice were sensitized to HDM and, 3 weeks later, challenged with HDM on three consecutive days through the airways. Two days after the last exposure, bronchoalveolar lavage fluids and blood samples and lung tissues were evaluated for the airway inflammation. MEASUREMENTS AND MAIN RESULTS The development of HDM-induced eosinophilic airway inflammation was dependent on OX40 of both CD4(+) T cells and NKT cells; OX40 engagement on CD4(+) T cells in the sensitization led to pulmonary OX40L augmentation after the allergen challenge, which stimulated pulmonary NKT cells through OX40 to provide the pathogenic cytokine milieu. This was ablated by OX40L blockade by inhalation of the neutralizing antibody during the challenge, suggesting the therapeutic potential of targeting pulmonary OX40-OX40L interactions. Moreover, OX40 expression in CD4(+) T cells, but not in NKT cells, was reciprocally regulated by the helper T cell type 1-skewing transcription factor Runx3. CONCLUSIONS OX40 on not only CD4(+) T cells but also NKT cells is involved in allergic airway inflammation. Notably, pulmonary blockade of OX40 ligation on NKT cells has therapeutic implications.
Collapse
Affiliation(s)
- Triya Damayanti
- Department of Pulmonary Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Macrophages are an abundant component of myeloma microenvironment and protect myeloma cells from chemotherapy drug-induced apoptosis. Blood 2009; 114:3625-8. [PMID: 19710503 DOI: 10.1182/blood-2009-05-220285] [Citation(s) in RCA: 243] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Multiple myeloma remains an incurable disease. One of the major problems is that myeloma cells develop drug resistance on interaction with bone marrow stromal cells. In this study, we examined the effects of macrophages (Mvarphis), a type of stromal cells, on myeloma cell survival and response to chemotherapy. We showed that Mvarphi, in particular tumor-associated Mvarphi, is a protector of myeloma cells. The protective effect was dependent on direct contact between Mvarphis and myeloma cells. Mvarphis protected both myeloma cell lines and primary myeloma cells from spontaneous and chemotherapy drug-induced apoptosis by attenuating the activation and cleavage of caspase-dependent apoptotic signaling. These findings are clinically relevant because we found that CD68+ Mvarphis heavily infiltrate the bone marrow of patients with myeloma but not the bone marrow of control patients. Thus, our results indicate that Mvarphis may contribute to myeloma cell survival and resistance to chemotherapeutic treatment in vivo.
Collapse
|
19
|
A fusion protein vaccine containing OprF epitope 8, OprI, and type A and B flagellins promotes enhanced clearance of nonmucoid Pseudomonas aeruginosa. Infect Immun 2009; 77:2356-66. [PMID: 19349426 DOI: 10.1128/iai.00054-09] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Although chronic Pseudomonas aeruginosa infection is the major cause of morbidity and mortality in cystic fibrosis (CF) patients, there is no approved vaccine for human use against P. aeruginosa. The goal of this study was to establish whether a multivalent vaccine containing P. aeruginosa type A and B flagellins as well as the outer membrane proteins OprF and OprI would promote enhanced clearance of P. aeruginosa. Intramuscular immunization with flagellins and OprI (separate) or OprI-flagellin fusion proteins generated significant antiflagellin immunoglobulin G (IgG) responses. However, only the fusions of OprI with type A and type B flagellins generated OprI-specific IgG. Immunization with a combination of OprF epitope 8 (OprF(311-341)), OprI, and flagellins elicited high-affinity IgG antibodies specific to flagellins, OprI, and OprF that individually promoted extensive deposition of C3 on P. aeruginosa. Although these antibodies exhibited potent antibody-dependent complement-mediated killing of nonmucoid bacteria, they were significantly less effective with mucoid isolates. Mice immunized with the OprF(311-341)-OprI-flagellin fusion had a significantly lower bacterial burden three days postchallenge and cleared the infection significantly faster than control mice. In addition, mice immunized with the OprF(311-341)-OprI-flagellin fusion had significantly less inflammation and lung damage throughout the infection than OprF-OprI-immunized mice. Based on our results, OprF(311-341)-OprI-flagellin fusion proteins have substantial potential as components of a vaccine against nonmucoid P. aeruginosa, which appears to be the phenotype of the bacterium that initially colonizes CF patients.
Collapse
|
20
|
Kanagawa N, Niwa M, Hatanaka Y, Tani Y, Nakagawa S, Fujita T, Yamamoto A, Okada N. CC-chemokine ligand 17 gene therapy induces tumor regression through augmentation of tumor-infiltrating immune cells in a murine model of preexisting CT26 colon carcinoma. Int J Cancer 2007; 121:2013-2022. [PMID: 17621629 DOI: 10.1002/ijc.22908] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Chemokines, which regulate leukocyte trafficking and infiltration of local sites, are attractive candidates for improving the efficacy of cancer immunotherapy by enhancing the accumulation of immune cells in tumor tissue. Herein, we evaluated the antitumor effects of intratumoral injection of RGD fiber-mutant adenoviral vectors (AdRGDs) encoding the chemokines CCL17, CCL19, CCL20, CCL21, CCL22, CCL27, XCL1 or CX3CL1 in a murine model of preexisting CT26 colon carcinoma. Among these 8 chemokine-expressing AdRGDs, injection of AdRGD-CCL17 most effectively induced tumor regression and generated specific immunity in rechallenge experiments. Tumor elimination activity by intratumoral injection of AdRGD-CCL17 depended on both the vector dose and the number of injections, and mainly required CD8+ CTLs in an effector phase as confirmed by analysis using BALB/c nude mice and an in vivo depletion assay. In addition, CCL17 gene transduction induced significant increases in the number of infiltrating macrophages and CD8+ T cells in CT26 tumors, and changed the tumor microenvironment to an immunologic activation state in which there was enhanced expression of lymphocyte activation markers and cell adhesion molecules. Thus, our data provide evidence that CCL17 gene transduction of local tumor sites is a promising approach for the development of a cancer immunogene therapy that can recruit activated tumor-infiltrating immune effector cells.
Collapse
Affiliation(s)
- Naoko Kanagawa
- Department of Biotechnology and Therapeutics, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, Japan
| | - Masakazu Niwa
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, 5 Nakauchi-cho, Misasagi, Yamashina-ku, Kyoto, Japan
| | - Yutaka Hatanaka
- Department of Biomedical Science, Dako Japan Co. Ltd., Nishinotouin-higashiiru, Shijo-dori, Shimogyo-ku, Kyoto, Japan
| | - Yoichi Tani
- Department of Biomedical Science, Dako Japan Co. Ltd., Nishinotouin-higashiiru, Shijo-dori, Shimogyo-ku, Kyoto, Japan
| | - Shinsaku Nakagawa
- Department of Biotechnology and Therapeutics, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, Japan
| | - Takuya Fujita
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, 5 Nakauchi-cho, Misasagi, Yamashina-ku, Kyoto, Japan
| | - Akira Yamamoto
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, 5 Nakauchi-cho, Misasagi, Yamashina-ku, Kyoto, Japan
| | - Naoki Okada
- Department of Biotechnology and Therapeutics, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, Japan
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, 5 Nakauchi-cho, Misasagi, Yamashina-ku, Kyoto, Japan
| |
Collapse
|
21
|
Liu G, Ma H, Jiang L, Peng J, Zhao Y. The immunity of splenic and peritoneal F4/80(+) resident macrophages in mouse mixed allogeneic chimeras. J Mol Med (Berl) 2007; 85:1125-35. [PMID: 17541534 DOI: 10.1007/s00109-007-0215-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2007] [Revised: 03/18/2007] [Accepted: 03/27/2007] [Indexed: 01/08/2023]
Abstract
Mixed allogeneic chimeras are emerging as a prospective approach to induce immune tolerance in clinics. However, the immunological function of macrophages in mixed chimeras has not been evaluated. Using a B6-->BALB/c mixed chimera model, we investigated the phenotype and function of F4/80(+) resident peritoneal exudate macrophage (PEMs) and splenic macrophages (SPMs) in vitro and in vivo. Recipient F4/80(+)PEMs and SPMs in mixed chimeras expressed significantly lower levels of MHC-II, CD54, and CD23 than those in non-chimeric mice before lipopolysaccharide stimulation. Recipient F4/80(+)PEMs and SPMs in mixed chimeras induced normal cell proliferation and delayed-type hypersensitivity of allo-T cells, but they induced more IFN-gamma and IL-2 products and less IL-10 and TGF-beta products of allo-T cells compared with those of non-chimeras. Furthermore, recipient F4/80(+)PEMs and SPMs had significantly higher phagocytotic capacity against chicken red blood cells or allo-T cells than those of controls while they had normal phagocytosis to Escherichia coli. Although some slight but significant alterations of recipient macrophages have been detected, these results provide direct evidences for the efficient immunity of recipient macrophages in mixed allogeneic chimeras. The present study also, for the first time, offered basic information for macrophages maturing in heterogeneous environments.
Collapse
Affiliation(s)
- Guangwei Liu
- Transplantation Biology Research Division, State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beisihuan Xi Road 25, Beijing, 100080, China
| | | | | | | | | |
Collapse
|
22
|
Nukiwa M, Andarini S, Zaini J, Xin H, Kanehira M, Suzuki T, Fukuhara T, Mizuguchi H, Hayakawa T, Saijo Y, Nukiwa T, Kikuchi T. Dendritic cells modified to express fractalkine/CX3CL1 in the treatment of preexisting tumors. Eur J Immunol 2006; 36:1019-27. [PMID: 16525992 DOI: 10.1002/eji.200535549] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Fractalkine (CX3CL1) is a unique membrane-bound CX3C chemokine that serves as a potent chemoattractant for lymphocytes. The hypothesis of this study is that dendritic cells (DC) genetically modified ex vivo to overexpress fractalkine would enhance the T cell-mediated cellular immune response with a consequent induction of anti-tumor immunity to suppress tumor growth. To prove this hypothesis, established tumors of different mouse cancer cells (B16-F10 melanoma, H-2b, and Colon-26 colon adenocarcinoma, H-2d) were treated with intratumoral injection of bone marrow-derived DC that had been modified in vitro with an RGD fiber-mutant adenovirus vector expressing mouse fractalkine (Ad-FKN). In both tumor models tested, treatment of tumor-bearing mice with Ad-FKN-transduced DC gave rise to a significant suppression of tumor growth along with survival advantages in the treated mice. Immunohistochemical analysis of tumors treated with direct injection of Ad-FKN-transduced DC demonstrated that the treatment prompted CD8+ T cells and CD4+ T cells to accumulate in the tumor milieu, leading to activation of immune-relevant processes. Consistent with the finding, the intratumoral administration of Ad-FKN-transduced DC evoked tumor-specific cytotoxic T lymphocytes, which ensued from in vivo priming of Th1 immune responses in the treated host. In addition, the anti-tumor effect provided by intratumoral injection of Ad-FKN-transduced DC was completely abrogated in CD4+ T cell-deficient mice as well as in CD8+ T cell-deficient mice. These results support the concept that genetic modification of DC with a recombinant fractalkine adenovirus vector may be a useful strategy for cancer immunotherapy protocols.
Collapse
Affiliation(s)
- Mio Nukiwa
- Department of Respiratory Oncology and Molecular Medicine, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Abstract
Dendritic cells are professional antigen presenting cells, which show an extraordinary capacity to initiate primary immune responses by stimulating T cells. This established function of dendritic cells has attracted much attention in efforts to develop useful vaccines for the treatment of cancer and infectious diseases. Designing effective strategies to generate clinical dendritic cell-based vaccine protocols remains a challenging field of research. The successful realization of immunotherapy utilizing dendritic cells will depend on modifications of these protocols to optimize the natural stimulatory properties of dendritic cells, such as genetic modification of dendritic cells. This review focuses on dendritic cell gene modifications for enhancing the multiple effector functions of dendritic cells, including viral and non-viral gene transfer into dendritic cells, and a variety of transferred genes, such as those encoding antigens, co-stimulatory molecules, cytokines, and chemokines.
Collapse
Affiliation(s)
- Toshiaki Kikuchi
- Department of Respiratory Oncology and Molecular Medicine, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan.
| |
Collapse
|
24
|
N/A, 徐 菡, 安 小, 王 子. N/A. Shijie Huaren Xiaohua Zazhi 2005; 13:2464-2468. [DOI: 10.11569/wcjd.v13.i20.2464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/26/2023] Open
|
25
|
Kikuchi T, Andarini S, Xin H, Gomi K, Tokue Y, Saijo Y, Honjo T, Watanabe A, Nukiwa T. Involvement of fractalkine/CX3CL1 expression by dendritic cells in the enhancement of host immunity against Legionella pneumophila. Infect Immun 2005; 73:5350-7. [PMID: 16113250 PMCID: PMC1231053 DOI: 10.1128/iai.73.9.5350-5357.2005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Legionnaires' disease is clinically manifested as severe pneumonia caused by Legionella pneumophila. However, the dendritic cell (DC)-centered immunological framework of the host defense against L. pneumophila has not been fully delineated. For this study, we focused on a potent chemoattractant for lymphocytes, fractalkine/CX3CL1, and observed that the fractalkine expression of DCs was somewhat up-regulated when they encountered L. pneumophila. We therefore hypothesized that fractalkine expressed by Legionella-capturing DCs is involved in the induction of T-cell-mediated immune responses against Legionella, which would be enhanced by a genetic modulation of DCs to overexpress fractalkine. In vivo immunization-challenge experiments demonstrated that DCs modified with a recombinant adenovirus vector to overexpress fractalkine (AdFKN) and pulsed with heat-killed Legionella protected immunized mice from a lethal Legionella infection and that the generation of in vivo protective immunity depended on the host lymphocyte subsets, including CD4(+) T cells, CD8(+) T cells, and B cells. Consistent with this, immunization with AdFKN/Legionella/DC induced significantly higher levels of serum anti-Legionella antibodies of several isotypes than those induced by control immunizations. Further analysis of spleen cells from the immunized mice indicated that the AdFKN/Legionella/DC immunization elicited Th1-dominated immune responses to L. pneumophila. These observations suggest that fractalkine may play an important role in the DC-mediated host defense against intracellular pathogens such as L. pneumophila.
Collapse
Affiliation(s)
- Toshiaki Kikuchi
- Department of Respiratory Oncology and Molecular Medicine, Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryomachi, Aobaku, Sendai 980-8575, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Fu G, Wijburg OLC, Cameron PU, Price JD, Strugnell RA. Salmonella enterica Serovar Typhimurium infection of dendritic cells leads to functionally increased expression of the macrophage-derived chemokine. Infect Immun 2005; 73:1714-22. [PMID: 15731072 PMCID: PMC1064904 DOI: 10.1128/iai.73.3.1714-1722.2005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Gene expression in murine dendritic cells (DCs) infected with green fluorescent protein-expressing Salmonella enterica serovar Typhimurium BRD509 was studied by mRNA differential display. Infected DCs were sorted from uninfected cells by flow cytometry. The mRNA expression patterns of infected and uninfected cells revealed a number of differentially expressed transcripts, which included the macrophage-derived chemokine (MDC). Up-regulation of MDC transcription in infected DCs was confirmed by Northern blotting, and the kinetics of MDC expression was examined by real-time reverse transcription-PCR, with which 31- and 150-fold increases were detected at 2 and 6 h postinfection, respectively. The increased release by DCs of MDC into culture media was detected by an enzyme-linked immunosorbent assay. The biological activity of MDC was investigated in in vitro and in vivo assays. In vitro, supernatants from S. enterica serovar Typhimurium-infected DCs were chemoattractive to T cells, and neutralization of MDC in these supernatants inhibited T-cell migration. Passive transfer of anti-MDC antibody to mice infected with BRD509 revealed that neither growth of the bacterium nor resistance of the mice to reinfection was affected and that in vivo inhibition of MDC did not affect T-cell responses, as measured by the gamma interferon ELISPOT method 3 days after challenge infection.
Collapse
Affiliation(s)
- Guo Fu
- CRC for Vaccine Technology, Parkville, Victoria 3010, Australia
| | | | | | | | | |
Collapse
|
27
|
Shao C, Qu J, He L, Zhang Y, Wang J, Zhou H, Wang Y, Liu X. Dendritic cells transduced with an adenovirus vector encoding interleukin-12 are a potent vaccine for invasive pulmonary aspergillosis. Genes Immun 2005; 6:103-14. [PMID: 15674391 DOI: 10.1038/sj.gene.6364167] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Invasive pulmonary aspergillosis (IPA) is a common and devastating pneumonia. We developed a novel antiinfective vaccine that couples the potent Ag-presenting capacity of dendritic cells (DCs) with paracrine delivery of interleukin-12 (IL-12) to local immune response sites. Our results showed that DCs engulfed Aspergillus conidia through coiling phagocytosis. Transfection of DCs with adenovirus encoding the cDNA of IL-12 did not affect their morphology and capacity to engulf conidia. The transduced DCs secreted IL-12, which was biologically active, to induce the production of gamma interferon (IFN-gamma) from spleen cells. Adoptive transfer of DCs pulsed with heat-inactivated Aspergillus fumigatus (HAF) to naive mice induced the Ag-specific production of IFN-gamma; the transduced HAF-pulsed DCs augmented this immune response further. Animals receiving HAF-pulsed DCs had lower fungal burdens, a more than three-fold higher survival rate at day 3. This protection was associated with a pronounced enhancement in the Aspergillus-specific IFN-gamma response. IL-12-engineered DCs augmented this protection strikingly as judged by a higher survival, and almost no Aspergillus could be detected in the lung of mice that had received IL-12-transduced HAF-pulsed DCs. These results suggest that antigen-pulsed DCs and IL-12 gene therapy could be used as adjunct therapy for aspergillosis.
Collapse
Affiliation(s)
- C Shao
- Department of Pulmonary Medicine, Zhongshan Hospital, University of Fudan, Shanghai, China
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Abstract
The chemokine/chemokine receptor network is an essential part of an intricate system of immunosurveillance and homeostasis, it promotes or suppresses neovascularization, affects and regulates directly or indirectly growth and metastasis of malignant cells. Numerous studies have been conducted to harness this network as therapeutic agents for cancer to redress the chemokine balance and control angiogenesis and tumour growth and metastasis. Second generation of immunotherapeutics and chemoattractant-based vaccines use chemokines and chemoattractant peptides to elicit antitumor immunity by a specific targeting and modulating subsets of effector leukocytes, including professional antigen presenting cells.
Collapse
Affiliation(s)
- Marta Coscia
- Laboratorio di Ematologia Oncologica, Divisione di Ematologia dell'Universita' di Torino, CeRMS, Azienda Ospedaliera San Giovanni Battista, Torino, Italy
| | | |
Collapse
|
29
|
Kikuchi T, Kobayashi T, Gomi K, Suzuki T, Tokue Y, Watanabe A, Nukiwa T. Dendritic cells pulsed with live and dead Legionella pneumophila elicit distinct immune responses. THE JOURNAL OF IMMUNOLOGY 2004; 172:1727-34. [PMID: 14734755 DOI: 10.4049/jimmunol.172.3.1727] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Legionella pneumophila is the causative pathogen of Legionnaires' disease, which is characterized by severe pneumonia. In regard to the pathophysiology of Legionella infection, the role of inflammatory phagocytes such as macrophages has been well documented, but the involvement of dendritic cells (DCs) has not been clarified. In this study, we have investigated the immune responses that DCs generate in vitro and in vivo after contact with L. pneumophila. Heat- and formalin-killed L. pneumophila, but not live L. pneumophila, induced immature DCs to undergo similar phenotypic maturation, but the secreted proinflammatory cytokines showed different patterns. The mechanisms of the DC maturation by heat- or formalin-killed L. pneumophila depended, at least in part, on Toll-like receptor 4 signaling or on Legionella LPS, respectively. After transfer to naive mice, DCs pulsed with dead Legionella produced serum Ig isotype responses specific for Legionella, leading to protective immunity against an otherwise lethal respiratory challenge with L. pneumophila. The in vivo immune responses required the Ag presentation of DCs, especially that on MHC class II molecules, and the immunity yielded cross-protection between clinical and environmental strains of L. pneumophila. Although the DC maturation was impaired by live Legionella, macrophages were activated by live as well as dead L. pneumophila, as evidenced by the up-regulation of MHC class II. Finally, DCs, but not macrophages, exhibited a proliferative response to live L. pneumophila that was consistent with their cell cycle progression. These findings provide a better understanding of the role of DCs in adaptive immunity to Legionella infection.
Collapse
MESH Headings
- Animals
- Antibodies, Bacterial/biosynthesis
- Antibodies, Bacterial/blood
- Cell Division/immunology
- Cell Membrane/immunology
- Cell Membrane/metabolism
- Cytokines/metabolism
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Dendritic Cells/microbiology
- Dendritic Cells/transplantation
- Escherichia coli Infections/immunology
- Escherichia coli Infections/microbiology
- Escherichia coli Infections/prevention & control
- Female
- Formaldehyde
- Histocompatibility Antigens Class I/biosynthesis
- Histocompatibility Antigens Class I/genetics
- Histocompatibility Antigens Class II/biosynthesis
- Histocompatibility Antigens Class II/genetics
- Hot Temperature
- Immunophenotyping
- Legionella pneumophila/growth & development
- Legionella pneumophila/immunology
- Legionnaires' Disease/immunology
- Legionnaires' Disease/microbiology
- Legionnaires' Disease/prevention & control
- Lipopolysaccharides/pharmacology
- Macrophages, Peritoneal/cytology
- Macrophages, Peritoneal/immunology
- Macrophages, Peritoneal/metabolism
- Macrophages, Peritoneal/microbiology
- Membrane Glycoproteins/physiology
- Mice
- Mice, Inbred A
- Mice, Inbred BALB C
- Mice, Inbred C3H
- Mice, Inbred C57BL
- Mice, Mutant Strains
- Receptors, Cell Surface/physiology
- Signal Transduction/immunology
- Toll-Like Receptors
Collapse
Affiliation(s)
- Toshiaki Kikuchi
- Department of Respiratory Oncology and Molecular Medicine, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan.
| | | | | | | | | | | | | |
Collapse
|
30
|
Andarini S, Kikuchi T, Nukiwa M, Pradono P, Suzuki T, Ohkouchi S, Inoue A, Maemondo M, Ishii N, Saijo Y, Sugamura K, Nukiwa T. Adenovirus Vector-Mediated in Vivo Gene Transfer of OX40 Ligand to Tumor Cells Enhances Antitumor Immunity of Tumor-Bearing Hosts. Cancer Res 2004; 64:3281-7. [PMID: 15126371 DOI: 10.1158/0008-5472.can-03-3911] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OX40 ligand (OX40L), the ligand for OX40 on activated CD4+ T cells, has adjuvant properties for establishing effective T-cell immunity, a potent effector arm of the immune system against cancer. The hypothesis of this study is that in vivo genetic engineering of tumor cells to express OX40L will stimulate tumor-specific T cells by the OX40L-OX40 engagement, leading to an induction of systemic antitumor immunity. To investigate this hypothesis, s.c. established tumors of three different mouse cancer cells (B16 melanoma, H-2b; Lewis lung carcinoma, H-2b; and Colon-26 colon adenocarcinoma, H-2d) were treated with intratumoral injection of a recombinant adenovirus vector expressing mouse OX40L (AdOX40L). In all tumor models tested, treatment of tumor-bearing mice with AdOX40L induced a significant suppression of tumor growth along with survival advantages in the treated mice. The in vivo AdOX40L modification of tumors evoked tumor-specific cytotoxic T lymphocytes in the treated host correlated with in vivo priming of T helper 1 immune responses in a tumor-specific manner. Consistent with the finding, the antitumor effect provided by intratumoral injection of AdOX40L was completely abrogated in a CD4+ T cell-deficient or CD8+ T cell-deficient condition. In addition, ex vivo AdOX40L-transduced B16 cells also elicited B16-specific cytotoxic T lymphocyte responses, and significantly suppressed the B16 tumor growth in the immunization-challenge experiment. All of these results support the concept that genetic modification of tumor cells with a recombinant OX40L adenovirus vector may be of benefit in cancer immunotherapy protocols.
Collapse
Affiliation(s)
- Sita Andarini
- Department of Respiratory Oncology and Molecular Medicine, Division of Cancer Control, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Lee JM, Merritt RE, Mahtabifard A, Yamada R, Kikuchi T, Crystal RG, Korst RJ. Intratumoral expression of macrophage-derived chemokine induces CD4+ T cell-independent antitumor immunity in mice. J Immunother 2003; 26:117-29. [PMID: 12616103 DOI: 10.1097/00002371-200303000-00004] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Macrophage-derived chemokine is chemotactic for a variety of leukocytes, and has been shown to be involved in T 2-mediated cellular immunity. To evaluate the role of this chemokine in tumor immunity in vivo, an adenovirus vector encoding the human macrophage-derived chemokine cDNA (AdMDC) was administered to established murine tumors. Gene transfer with AdMDC significantly inhibited tumor growth and prolonged animal survival. AdMDC was not directly cytotoxic to tumor cells, but splenocytes from animals that received intratumoral AdMDC were able to lyse syngeneic tumor cells, and purified splenic CD8 cells secreted interferon-gamma in a tumor-specific manner. The antitumor activity of AdMDC was lost in mice lacking CD8 T lymphocytes, but surprisingly, it was preserved in animals lacking CD4 cells, as was the systemic cytotoxic T lymphocyte response. Systemic NK cells did not play a role in the antitumor immune response induced by AdMDC. Experiments using knockout mice demonstrated that host expression of MHC Class I, but not Class II, IL-4, or IL-12, was necessary for AdMDC to exert its antitumor effect, and immunohistochemistry demonstrated infiltrates of CD8 and CD86 cells, but not CD4 cells in treated tumors. These studies highlight a new function for macrophage-derived chemokine by demonstrating that it possesses in vivo antitumor activity with CD8 T cells as the effector cells, and interestingly, that the CD4 cell/MHC II pathway of CD8 cell activation is not required for the antitumor effects of this chemokine.(H)
Collapse
Affiliation(s)
- Jay M Lee
- Deparment of Cardiothoracic Surgery, Division of Pulmonary and Critical Care Medicine, Sloan-Kettering Cancer Center, New York, NY, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Azenabor AA, Chaudhry AU. Chlamydia pneumoniae survival in macrophages is regulated by free Ca2+ dependent reactive nitrogen and oxygen species. J Infect 2003; 46:120-8. [PMID: 12634074 DOI: 10.1053/jinf.2002.1098] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
OBJECTIVES Despite an efficient macrophage immune capability, Chlamydia pneumoniae infects host cells and causes chronic diseases. To gain better insights into C. pneumoniae survival mechanisms in macrophages, its growth in regular RAW-264.7 cells (nitric oxide sufficient NO (+)) and RAW-264.7 cells (nitric oxide insufficient NO (-)) were studied. METHODS Role of Ca(2+), NO and reactive oxygen species (ROS) during C. pneumoniae infection in macrophages were determined. RESULTS RAW-264.7 NO (-) cells supported significantly Chlamydia growth, showing an upregulation of ROS, superoxide dismutase (SOD) and catalase activities as compared with RAW-264.7 NO (+) cell. Ascorbic acid, inducible nitric oxide synthase inhibitor and glutathione significantly prompted Chlamydia inclusion formation. Cytosolic Ca(2+) had regulatory effect on organism growth, NO generation, SOD and catalase activities in both cell types. CONCLUSIONS These findings suggest that minimal Ca(2+) signaling in macrophages at early stages of infection, NO and ROS release have modulatory effects onC. pneumoniae survival, onset of persistence and chronicity, processes which are needed for the initiation of diseases in which C. pneumoniae has been implicated as a possible etiologic agent.
Collapse
Affiliation(s)
- A A Azenabor
- Department of Health Sciences, University of Wisconsin, Milwaukee, WI 53211, USA.
| | | |
Collapse
|
33
|
Biragyn A, Belyakov IM, Chow YH, Dimitrov DS, Berzofsky JA, Kwak LW. DNA vaccines encoding human immunodeficiency virus-1 glycoprotein 120 fusions with proinflammatory chemoattractants induce systemic and mucosal immune responses. Blood 2002; 100:1153-9. [PMID: 12149191 DOI: 10.1182/blood-2002-01-0086] [Citation(s) in RCA: 103] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
DNA immunizations with glycoprotein 120 (gp120) of human immunodeficiency virus-1 (HIV-1) usually require boosting with protein or viral vaccines to achieve optimal efficacy. Here, we demonstrate for the first time that mice immunized with DNA encoding gp120 fused with proinflammatory chemoattractants of immature dendritic cells, such as beta-defensin 2, monocyte chemoattractant protein-3 (MCP-3/CCL7) or macrophage-derived chemokine (MDC/CCL22), elicited anti-gp120 antibodies with high titers of virus-neutralizing activity. The immunogenicity was further augmented with the use of chemokine fusion constructs with gp140, gp120 linked to the extracellular domain of gp41 via a 14-amino acid spacer peptide sequence. This construct elicited antibodies with more effective neutralizing activity than corresponding constructs expressing gp120. Responses were dependent on physical linkage with chemokine moiety, as no immunity was detected following immunization of mice with DNA encoding a free mixture of chemokine and gp120. Although the route of immunization was inoculation into skin, both systemic and mucosal CD8(+) cytolytic immune responses were elicited in mice immunized with DNA expressing MCP-3 or beta-defensin 2 fusion constructs. In contrast, no cytotoxic T lymphocyte activity (CTL) was detected in mice immunized with DNA encoding gp120 either alone or as fusion with MDC. Therefore, the potential for broad application of this approach lies in the induction of mucosal CTL and neutralizing antibodies to HIV-1 envelope, both key requirements for prevention of viral transmission and clearance of pathogenic HIV from mucosal reservoirs.
Collapse
Affiliation(s)
- Arya Biragyn
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | | | | | | | | | | |
Collapse
|
34
|
Abstract
Organ transplant rejection is mediated largely by circulating peripheral leukocytes induced to infiltrate the graft by various inflammatory stimuli. Of these, chemotactic cytokines called chemokines, expressed by inflamed graft tissues, as well as by early innate-responding leukocytes that infiltrate the graft, are responsible for the recruitment of alloreactive leukocytes. This report discusses the impact of these leukocyte-directing proteins on transplant outcome and novel therapeutic approaches for antirejection therapy based on targeting of chemokines and/or their receptors.
Collapse
Affiliation(s)
- Bridget L Colvin
- Thomas E. Starzl Transplantation Institute and Departments of Surgery, Molecular Genetics and Biochemistry, and Inmunology, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | | |
Collapse
|
35
|
Abstract
Prevention and treatment of respiratory infections remain an important health care challenge as the US population ages, contains more susceptible or high-risk people, and encounters new pathogens or antibiotic resistant bacteria. Reasonably protective vaccines against very common microbes are available for childhood and adult immunization, but, generally, these are underutilized. A broader definition of higher risk individuals is evolving, which will include more for immunization. Different approaches to vaccine development through design of new component vaccines are necessary. This review has updated host defense mechanisms at three levels in the human respiratory tract: naso-oropharynx (upper airways), conducting airways, and alveolar space. Examples of representative pathogenic microbes have been inserted at the respective airway segment where they may colonize or create infection (influenza, measles virus, Porphyromonas gingivalis causing periodontitis, Bordetella pertussis, Chlamydia pneumoniae, Streptococcus pneumoniae, and Bacillus anthracis ). Hopefully, microbe-host interactions will suggest new approaches for preventing these kinds of infections.
Collapse
Affiliation(s)
- Herbert Y Reynolds
- J. Lloyd Huck Professor of Medicine, Chair, Milton S. Hershey Medical Center, The Pennsylvania State University, Hershey, Pennsylvania 17033-0850, USA.
| |
Collapse
|