1
|
Zhang J, Luo W, Cui Y, Sun B. B-cell epitope peptide immunotherapy alleviates chitin-binding protein-induced type 2 airway inflammation in a Blomia tropicalis-murine model. Respir Res 2025; 26:129. [PMID: 40205365 PMCID: PMC11983821 DOI: 10.1186/s12931-025-03207-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 03/27/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND Peptide immunotherapy (PIT) offers a safe and effective treatment with minimal side effects. This study aims to identify B-cell epitopes of a novel allergen from Blomia tropicalis (B. tropicalis), specifically the Chitin-binding domain type 2 (ChtBD2) protein, and evaluate the therapeutic effects of peptide treatment in a murine model. METHODS Using Alphafold2, the 3D structure of ChtBD2 was constructed. AI-based and traditional computational tools predicted the predominant B-cell epitopes. Twelve synthesized peptides were assessed for allergenicity and immunogenicity. A murine model of B. tropicalis-induced allergic airway inflammation mimicking human atopic asthma was developed and analyzed. RESULTS Predominant B-cell epitopes of ChtBD2 were identified as promising IgE-binding domains. Peptide 1 (PT1: 1-15) showed significant IgE-binding activity and the highest inhibition rate in competitive IgE-binding assays. PT1 upregulated IL-4, IL-13, and CD63 in B. tropicalis-sensitized patients' PBMCs and basophils, respectively. Notably, IT groups showed reduced lung cellular infiltration and type 2 cytokine expression in BALF. Specific IgE levels were reduced, with a decline in the IgG1/IgG2a ratio. CONCLUSIONS This study represents the first AI-facilitated development of a B-cell epitope-based ChtBD2 PIT, showing promise as an immunotherapy for B. tropicalis-allergic patients with reduced allergenicity and high immunogenicity in inducing IgG-blocking antibodies. CLINICAL TRIAL Not applicable.
Collapse
Affiliation(s)
- Jiale Zhang
- Department of Clinical Laboratory, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
- Guangzhou Laboratory, Guangzhou, China
| | - Wenting Luo
- Department of Clinical Laboratory, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
- Guangzhou Laboratory, Guangzhou, China
| | - YuBao Cui
- Clinical Research Center, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, 214023, China.
| | - Baoqing Sun
- Department of Clinical Laboratory, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China.
- Guangzhou Laboratory, Guangzhou, China.
| |
Collapse
|
2
|
Chen YC, Chung NH, Lin YC, Yu SL, Liu CC, Chow YH. Enhanced construction of a bivalent adenovirus-based vaccine for preventing childhood pathogens: Human respiratory syncytial virus and norovirus. SLAS Technol 2025; 31:100255. [PMID: 39952326 DOI: 10.1016/j.slast.2025.100255] [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: 10/03/2024] [Revised: 01/10/2025] [Accepted: 02/11/2025] [Indexed: 02/17/2025]
Abstract
To impede the spread of respiratory syncytial virus (RSV) and norovirus (NoV) in children, we developed novel recombinant adenoviruses expressing RSV fusion (F) and NoV VP1 proteins driven by different promoter elements (EF1α, SV40, and CMV), resulting in Ad-F/E/NoV, Ad-F/S/NoV, and Ad-F/C/NoV, respectively. The expressed F can be recognized by postfusion-specific antibody targeting to site II and prefusion-specific antibodies targeting to sites V and Ø in the Ad-F-infected cells. However, NoV VP1 is only expressed in Ad-F/E/NoV and Ad-F/C/NoV-infected cells. Intraperitoneal two-dose with monovalent Ad-F and all three bicistronic Ads individually in rodents induced anti-F neutralizing antibodies similarly. Ad-F/C/NoV and Ad-F/E/NoV but not Ad-F/S/NoV significantly induced NoV VP1-specific IgG. The serum from Ad-F/C/NoV-immunized subjects elicited superior anti-NoV activity, as evidenced by reduced binding of NoV VLPs to histo-blood group antigens. Ad-F/C/NoV and Ad-F/E/NoV-immunized mice exhibited elevated cellular immune-mediated splenic IFN-γ and IL-4 secretions. In the RSV challenge study, pulmonary virions were significantly decreased during the vaccination with each of the three bicistronic Ads, confirming their efficacy in preventing RSV infection. Finally, the mucosal (intranasal) immunization in mice with Ad-F/C/NoV induced superior anti-RSV and NoV IgA in both serum and vaginal washes specific to RSV and NoV were highly induced. These findings highlight the optimization of an Ad vaccine targeting two pathogens prevalent in childhood. Additionally, the results underscore the utility of mucosal delivery of Ad vaccines as a valuable strategy for public vaccination efforts.
Collapse
Affiliation(s)
- Ying-Chin Chen
- Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Zhunan Town, Miaoli County 350, Taiwan
| | - Nai-Hsiang Chung
- Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Zhunan Town, Miaoli County 350, Taiwan
| | - Yu-Ching Lin
- Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Zhunan Town, Miaoli County 350, Taiwan
| | - Shu-Ling Yu
- Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Zhunan Town, Miaoli County 350, Taiwan
| | - Chia-Chyi Liu
- Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Zhunan Town, Miaoli County 350, Taiwan
| | - Yen-Hung Chow
- Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Zhunan Town, Miaoli County 350, Taiwan; Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.
| |
Collapse
|
3
|
Pandey B, Wang Z, Jimenez A, Bhatia E, Jain R, Beach A, Maniar D, Hosten J, O'Farrell L, Vantucci C, Hur D, Noel R, Ringquist R, Smith C, Ochoa MA, Roy K. A Dual-Adjuvanted Parenteral-Intranasal Subunit Nanovaccine generates Robust Systemic and Mucosal Immunity Against SARS-CoV-2 in Mice. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402792. [PMID: 39352717 PMCID: PMC11615772 DOI: 10.1002/advs.202402792] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 09/09/2024] [Indexed: 12/06/2024]
Abstract
Existing parenteral SARS-CoV-2 vaccines produce only limited mucosal responses, essential for reducing transmission and achieving sterilizing immunity. Appropriately designed mucosal boosters can overcome the shortcomings of parenteral vaccines and enhance pre-existing systemic immunity. Here, a new protein subunit nanovaccine is developed by utilizing dual-adjuvanted (RIG-I: PUUC RNA and TLR-9: CpG DNA) polysaccharide-amino acid-lipid nanoparticles (PAL-NPs) along with SARS-CoV-2 S1 trimer protein, that can be delivered both intramuscularly (IM) and intranasally (IN) to generate balanced mucosal-systemic SARS-CoV-2 immunity. Mice receiving IM-Prime PUUC+CpG PAL subunit nanovaccine, followed by an IN-Boost, developed high levels of IgA, IgG, and cellular immunity in the lungs and showed robust systemic humoral immunity. Interestingly, as a purely intranasal subunit vaccine (IN-Prime/IN-Boost), PUUC+CpG PAL-NPs induced stronger lung-specific T cell immunity than IM-Prime/IN-Boost, and a comparable IgA and neutralizing antibodies, although with a lower systemic antibody response, indicating that a fully mucosal delivery route for SARS-CoV-2 vaccination may also be feasible. The data suggest that PUUC+CpG PAL subunit nanovaccine is a promising candidate for generating SARS-CoV-2 specific mucosal immunity.
Collapse
MESH Headings
- Animals
- Mice
- Immunity, Mucosal/immunology
- Immunity, Mucosal/drug effects
- SARS-CoV-2/immunology
- Administration, Intranasal/methods
- COVID-19 Vaccines/immunology
- COVID-19 Vaccines/administration & dosage
- COVID-19/immunology
- COVID-19/prevention & control
- Nanoparticles/administration & dosage
- Vaccines, Subunit/immunology
- Vaccines, Subunit/administration & dosage
- Antibodies, Viral/immunology
- Female
- Adjuvants, Vaccine/administration & dosage
- Spike Glycoprotein, Coronavirus/immunology
- Adjuvants, Immunologic/administration & dosage
- Antibodies, Neutralizing/immunology
- Mice, Inbred BALB C
- Nanovaccines
Collapse
Affiliation(s)
- Bhawana Pandey
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of TechnologyAtlantaGAUSA
| | - Zhengying Wang
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of TechnologyAtlantaGAUSA
| | - Angela Jimenez
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of TechnologyAtlantaGAUSA
| | - Eshant Bhatia
- Woodruff School of Mechanical EngineeringGeorgia Institute of TechnologyAtlantaGAUSA
| | - Ritika Jain
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of TechnologyAtlantaGAUSA
| | - Alexander Beach
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of TechnologyAtlantaGAUSA
| | - Drishti Maniar
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of TechnologyAtlantaGAUSA
| | - Justin Hosten
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of TechnologyAtlantaGAUSA
| | - Laura O'Farrell
- Physiological Research LaboratoryGeorgia Institute of TechnologyAtlantaGAUSA
| | - Casey Vantucci
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of TechnologyAtlantaGAUSA
| | - David Hur
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of TechnologyAtlantaGAUSA
| | - Richard Noel
- Physiological Research LaboratoryGeorgia Institute of TechnologyAtlantaGAUSA
| | - Rachel Ringquist
- The Parker H. Petit Institute for Bioengineering and BiosciencesSchool of Chemical & Biomolecular EngineeringGeorgia Institute of TechnologyAtlantaGAUSA
| | - Clinton Smith
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of TechnologyAtlantaGAUSA
| | - Miguel A. Ochoa
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of TechnologyAtlantaGAUSA
| | - Krishnendu Roy
- Wallace H. Coulter Department of Biomedical EngineeringThe Parker H. Petit Institute for Bioengineering and BiosciencesMarcus Center for Therapeutic Cell Characterization and ManufacturingGeorgia Institute of TechnologyAtlantaGAUSA
- Department of Biomedical EngineeringDepartment of Chemical and Biomolecular EngineeringSchool of EngineeringDepartment of Pathology, Microbiology and ImmunologySchool of MedicineVanderbilt UniversityNashvilleTNUSA
| |
Collapse
|
4
|
Yang Y, Zou GM, Wei XS, Zhang Z, Zhuo L, Xu QQ, Li WG. Identification and validation of biomarkers in membranous nephropathy and pan-cancer analysis. Front Immunol 2024; 15:1302909. [PMID: 38846934 PMCID: PMC11153720 DOI: 10.3389/fimmu.2024.1302909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 05/10/2024] [Indexed: 06/09/2024] Open
Abstract
Background Membranous nephropathy (MN) is an autoimmune disease and represents the most prevalent type of renal pathology in adult patients afflicted with nephrotic syndrome. Despite substantial evidence suggesting a possible link between MN and cancer, the precise underlying mechanisms remain elusive. Methods In this study, we acquired and integrated two MN datasets (comprising a single-cell dataset and a bulk RNA-seq dataset) from the Gene Expression Omnibus database for differential expression gene (DEG) analysis, hub genes were obtained by LASSO and random forest algorithms, the diagnostic ability of hub genes was assessed using ROC curves, and the degree of immune cell infiltration was evaluated using the ssGSEA function. Concurrently, we gathered pan-cancer-related genes from the TCGA and GTEx databases, to analyze the expression, mutation status, drug sensitivity and prognosis of hub genes in pan-cancer. Results We conducted intersections between the set of 318 senescence-related genes and the 366 DEGs, resulting in the identification of 13 senescence-related DEGs. Afterwards, we meticulously analyzed these genes using the LASSO and random forest algorithms, which ultimately led to the discovery of six hub genes through intersection (PIK3R1, CCND1, TERF2IP, SLC25A4, CAPN2, and TXN). ROC curves suggest that these hub genes have good recognition of MN. After performing correlation analysis, examining immune infiltration, and conducting a comprehensive pan-cancer investigation, we validated these six hub genes through immunohistochemical analysis using human renal biopsy tissues. The pan-cancer analysis notably accentuates the robust association between these hub genes and the prognoses of individuals afflicted by diverse cancer types, further underscoring the importance of mutations within these hub genes across various cancers. Conclusion This evidence indicates that these genes could potentially play a pivotal role as a critical link connecting MN and cancer. As a result, they may hold promise as valuable targets for intervention in cases of both MN and cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Wen-ge Li
- *Correspondence: Qian-qian Xu, ; Wen-ge Li,
| |
Collapse
|
5
|
Yin W, Xu Z, Chang C, Zhao Y, Wang H, Zhang J, Ma F, Zuo X, Tang B, Lu Y. Alginate di-aldehyde-modified metal-organic framework nanocarriers as delivery platform and adjuvant in inactivated pseudorabies vaccination. MATERIALS HORIZONS 2024; 11:2153-2168. [PMID: 38376908 DOI: 10.1039/d3mh02251j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2024]
Abstract
Pseudorabies virus (PRV) is a highly contagious viral disease, which leads to severe financial losses in the breeding industry worldwide. Presently, PRV is mainly controlled using live attenuated and inactivated vaccines. However, these vaccines have an innate tendency to lose their structural conformation upon exposure to environmental and chemical stressors and cannot provide full protection against the emerging prevalent PRV variants. In this work, first, we synthesized aminated ZIF-7/8 nanoparticles (NPs), and then chemical bond-coated alginate dialdehyde (ADA, a type of dioxide alginate saccharide) on their surface via Schiff base reaction to obtain ZIF-7/8-ADA NPs. The as-fabricated ZIF-7/8-ADA NPs exhibited high stability, monodispersity and a high loading ratio of antigen. Furthermore, the ZIF-7/8-ADA NPs showed good biocompatibility in vitro and in vivo. Using ZIF-7/8-ADA NPs as an adjuvant and inactivated PRV as a model antigen, we constructed a PR vaccine through a simple mixture. The immunity studies indicated that ZIF-7/8-ADA induced an enhancement in the Th1/Th2 immune response, which was superior to that of the commercial ISA201, alum adjuvant and ZIF-7/8. Due to the pH-sensitive release of the antigen in lysosomes, the as-prepared PR vaccine subsequently accelerated the antigen presentation and improved the immune responses in vitro and in vivo. The results of PRV challenge using mice as the model demonstrated that ZIF-7/8-ADA achieved the same preventive effect as the commercial ISA201 and was much better than the alum adjuvant, and thus can serve as a promising delivery system and adjuvant to enhance humoral and cellular responses against PRV infection.
Collapse
Affiliation(s)
- Wenzhu Yin
- Institute of Veterinary Immunology & Engineering, National Research Center of Engineering and Technology for Veterinary Biologicals, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China.
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- GuoTai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou 225300, China
| | - Zeyu Xu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Chen Chang
- Institute of Veterinary Immunology & Engineering, National Research Center of Engineering and Technology for Veterinary Biologicals, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China.
| | - Yanhong Zhao
- Institute of Veterinary Immunology & Engineering, National Research Center of Engineering and Technology for Veterinary Biologicals, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China.
| | - Haiyan Wang
- Institute of Veterinary Immunology & Engineering, National Research Center of Engineering and Technology for Veterinary Biologicals, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China.
| | - Jinqiu Zhang
- Institute of Veterinary Immunology & Engineering, National Research Center of Engineering and Technology for Veterinary Biologicals, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China.
| | - Fang Ma
- Institute of Veterinary Immunology & Engineering, National Research Center of Engineering and Technology for Veterinary Biologicals, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China.
| | - Xiaoxin Zuo
- Institute of Veterinary Immunology & Engineering, National Research Center of Engineering and Technology for Veterinary Biologicals, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China.
| | - Bo Tang
- Institute of Veterinary Immunology & Engineering, National Research Center of Engineering and Technology for Veterinary Biologicals, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China.
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Yu Lu
- Institute of Veterinary Immunology & Engineering, National Research Center of Engineering and Technology for Veterinary Biologicals, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China.
- GuoTai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou 225300, China
| |
Collapse
|
6
|
Pandey B, Wang Z, Jimenez A, Bhatia E, Jain R, Beach A, Maniar D, Hosten J, O'Farrell L, Vantucci C, Hur D, Noel R, Ringuist R, Smith C, Ochoa MA, Roy K. A multiadjuvant polysaccharide-amino acid-lipid (PAL) subunit nanovaccine generates robust systemic and lung-specific mucosal immune responses against SARS-CoV-2 in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.05.539395. [PMID: 37215018 PMCID: PMC10197586 DOI: 10.1101/2023.05.05.539395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Existing parenteral SARS-CoV-2 vaccines produce only limited mucosal responses, which are essential for reducing transmission and achieving sterilizing immunity. Appropriately designed mucosal boosters could overcome the shortcomings of parenteral vaccines and enhance pre- existing systemic immunity. Here we present a new protein subunit nanovaccine using multiadjuvanted (e.g. RIG-I: PUUC, TLR9: CpG) polysaccharide-amino acid-lipid nanoparticles (PAL-NPs) that can be delivered both intramuscularly (IM) and intranasally (IN) to generate balanced mucosal-systemic SARS-CoV-2 immunity. Mice receiving IM-Prime PUUC+CpG PAL- NPs, followed by an IN-Boost, developed high levels of IgA, IgG, and cellular immunity in the lung, and showed robust systemic humoral immunity. Interestingly, as a purely intranasal vaccine (IN-Prime/IN-Boost), PUUC+CpG PAL-NPs induced stronger lung-specific T cell immunity than IM-Prime/IN-Boost, and a comparable IgA and neutralizing antibodies, although with a lower systemic antibody response, indicating that a fully mucosal delivery route for SARS-CoV-2 vaccination may also be feasible. Our data suggest that PUUC+CpG PAL-NP subunit vaccine is a promising candidate for generating SARS-CoV-2 specific mucosal immunity.
Collapse
|
7
|
Sadeghi Z, Fasihi-Ramandi M, Davoudi Z, Bouzari S. Multi-Epitope Vaccine Candidates Associated with Mannosylated Chitosan and LPS Conjugated Chitosan Nanoparticles Against Brucella Infection. J Pharm Sci 2023; 112:991-999. [PMID: 36623693 DOI: 10.1016/j.xphs.2022.12.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 12/24/2022] [Accepted: 12/25/2022] [Indexed: 01/09/2023]
Abstract
One promising approach to increase protection against infectious diseases is to use adjuvants that can selectively stimulate the immune responses. In this study, multi-epitope antigens associated with LPS loaded chitosan (LLC) as toll-like receptor agonist or mannosylated chitosan nanoparticle (MCN) as vaccine delivery system were evaluated for their ability to stimulate immune responses to Brucella infection in mice model. Our results indicated that the addition of MCN to our vaccine formulations significantly elicited IFN-γ and IL-2 cytokines and antibody titers, in comparison with the non-adjuvanted vaccine candidates. The present results indicated that multi-epitopes and their administration with LLC or MCN induced Th1 immune response. In addition, vaccine candidates containing MCN provided high percentage of protection against B. melitensis and B. abortus infection. Our results provided support to previous reports indicating that MCNs are attractive adjuvants and addition of this adjuvant to multi-epitopes antigens play an important role in the development of vaccine against Brucella.
Collapse
Affiliation(s)
- Zohre Sadeghi
- Department of Molecular Biology, Pasteur Institute of Iran, Tehran, Iran
| | - Mahdi Fasihi-Ramandi
- Molecular Biology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Zahra Davoudi
- Department of Medical Biotechnology, Zanjan University of Medical Science, Zanjan, Iran
| | - Saeid Bouzari
- Department of Molecular Biology, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
8
|
Toll-like Receptor 9 Induced Dendritic Cell Activation Promotes Anti-Myeloperoxidase Autoimmunity and Glomerulonephritis. Int J Mol Sci 2023; 24:ijms24021339. [PMID: 36674855 PMCID: PMC9864438 DOI: 10.3390/ijms24021339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/23/2022] [Accepted: 12/25/2022] [Indexed: 01/13/2023] Open
Abstract
ANCA-associated vasculitis (AAV) is intricately linked with infections. Toll-like receptors (TLR) provide a potential link between infection and anti-myeloperoxidase (MPO) autoimmunity. TLR9 ligation has been shown to promote anti-MPO autoimmunity and glomerular vasculitis in murine MPO-AAV. This study investigates dendritic cell TLR9 ligation in murine experimental anti-MPO glomerulonephritis. We analyzed autoimmune responses to MPO following transfer of TLR9 stimulated, MPO pulsed dendritic cells and kidney injury following a sub-nephritogenic dose of sheep anti-mouse glomerular basement membrane globulin. TLR9 ligation enhanced dendritic cell activation upregulating CD40 and CD80 expression, promoting systemic anti-MPO autoimmunity and T cell recall responses and exacerbating kidney injury. CD40 upregulation by TLR9 was critical for the induction of nephritogenic autoimmunity. The presence of DEC205, which transports the TLR9 ligand to TLR9 located in the endosome, also promoted kidney injury. This confirms TLR9 mediated dendritic cell activation as a mechanism of anti-MPO autoimmunity in AAV and further defines the link between infection and the generation of MPO specific autoimmune inflammation.
Collapse
|
9
|
Melo ARDS, de Macêdo LS, Invenção MDCV, de Moura IA, da Gama MATM, de Melo CML, Silva AJD, Batista MVDA, de Freitas AC. Third-Generation Vaccines: Features of Nucleic Acid Vaccines and Strategies to Improve Their Efficiency. Genes (Basel) 2022; 13:genes13122287. [PMID: 36553554 PMCID: PMC9777941 DOI: 10.3390/genes13122287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/23/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
Gene immunization comprises mRNA and DNA vaccines, which stand out due to their simple design, maintenance, and high efficacy. Several studies indicate promising results in preclinical and clinical trials regarding immunization against ebola, human immunodeficiency virus (HIV), influenza, and human papillomavirus (HPV). The efficiency of nucleic acid vaccines has been highlighted in the fight against COVID-19 with unprecedented approval of their use in humans. However, their low intrinsic immunogenicity points to the need to use strategies capable of overcoming this characteristic and increasing the efficiency of vaccine campaigns. These strategies include the improvement of the epitopes' presentation to the system via MHC, the evaluation of immunodominant epitopes with high coverage against emerging viral subtypes, the use of adjuvants that enhance immunogenicity, and the increase in the efficiency of vaccine transfection. In this review, we provide updates regarding some characteristics, construction, and improvement of such vaccines, especially about the production of synthetic multi-epitope genes, widely employed in the current gene-based vaccines.
Collapse
Affiliation(s)
- Alanne Rayssa da Silva Melo
- Laboratory of Molecular Studies and Experimental Therapy—LEMTE, Department of Genetics, Federal University of Pernambuco, Recife 50670-901, Brazil
| | - Larissa Silva de Macêdo
- Laboratory of Molecular Studies and Experimental Therapy—LEMTE, Department of Genetics, Federal University of Pernambuco, Recife 50670-901, Brazil
| | - Maria da Conceição Viana Invenção
- Laboratory of Molecular Studies and Experimental Therapy—LEMTE, Department of Genetics, Federal University of Pernambuco, Recife 50670-901, Brazil
| | - Ingrid Andrêssa de Moura
- Laboratory of Molecular Studies and Experimental Therapy—LEMTE, Department of Genetics, Federal University of Pernambuco, Recife 50670-901, Brazil
| | - Marco Antonio Turiah Machado da Gama
- Laboratory of Molecular Studies and Experimental Therapy—LEMTE, Department of Genetics, Federal University of Pernambuco, Recife 50670-901, Brazil
| | - Cristiane Moutinho Lagos de Melo
- Laboratory of Immunological and Antitumor Analysis, Department of Antibiotics, Bioscience Center, and Keizo Asami Imunophatology Laboratory, Federal University of Pernambuco, Recife 50670-901, Brazil
| | - Anna Jéssica Duarte Silva
- Laboratory of Molecular Studies and Experimental Therapy—LEMTE, Department of Genetics, Federal University of Pernambuco, Recife 50670-901, Brazil
| | - Marcus Vinicius de Aragão Batista
- Laboratory of Molecular Genetics and Biotechnology (GMBio), Department of Biology, Center for Biological and Health Sciences, Federal University of Sergipe, São Cristóvão 49100-000, Brazil
| | - Antonio Carlos de Freitas
- Laboratory of Molecular Studies and Experimental Therapy—LEMTE, Department of Genetics, Federal University of Pernambuco, Recife 50670-901, Brazil
- Correspondence: ; Tel.: +55-8199-6067-671
| |
Collapse
|
10
|
Ardalan M, Ahmadian E, Hosseiniyan Khatibi SM, Rahbar Saadat Y, Bastami M, Bagheri Y, Zununi Vahed F, Shoja MM, Zununi Vahed S. Microbiota and glomerulonephritis: An immunological point of view. Am J Med Sci 2022; 364:695-705. [PMID: 35870511 DOI: 10.1016/j.amjms.2022.05.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 03/26/2022] [Accepted: 05/05/2022] [Indexed: 01/25/2023]
Abstract
Glomerular injury is the major cause of chronic kidney diseases (CKD) worldwide and is characterized by proteinuria. Glomerulonephritis (GN) has a wide spectrum of etiologies, the intensity of glomerular damage, histopathology, and clinical outcomes that can be associated with the landscape of the nephritogenic immune response. Beyond impaired immune responses and genetic factors, recent evidence indicates that microbiota can be contributed to the pathogenesis of GN and patients' outcomes by impacting many aspects of the innate and adaptive immune systems. It is still unknown whether dysbiosis induces GN or it is a secondary effect of the disease. Several factors such as drugs and nutritional problems can lead to dysbiosis in GN patients. It has been postulated that gut dysbiosis activates immune responses, promotes a state of systemic inflammation, and produces uremic toxins contributing to kidney tissue inflammation, apoptosis, and subsequent proteinuric nephropathy. In this review, the impact of gastrointestinal tract (GI) microbiota on the pathogenesis of the primary GN will be highlighted. The application of therapeutic interventions based on the manipulation of gut microbiota with special diets and probiotic supplementation can be effective in GN.
Collapse
Affiliation(s)
| | - Elham Ahmadian
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | - Milad Bastami
- Non-communicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Yasin Bagheri
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mohammadali M Shoja
- Clinical Academy of Teaching and Learning, Ross University School of Medicine, Miramar, FL, USA
| | | |
Collapse
|
11
|
Seroprevalence of Toxoplasma gondii among Hemodialysis Patients: A Possible Link to Main T-lymphocyte Subsets Levels and Dialysis Adequacy. Acta Trop 2022; 237:106703. [DOI: 10.1016/j.actatropica.2022.106703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 09/27/2022] [Accepted: 09/27/2022] [Indexed: 11/23/2022]
|
12
|
Glycosphingolipids form characteristic-sized liposomes that correlate with their antibody-inducing activities in mice. Biochem Biophys Res Commun 2022; 634:48-54. [DOI: 10.1016/j.bbrc.2022.09.094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 09/22/2022] [Accepted: 09/24/2022] [Indexed: 11/18/2022]
|
13
|
Chan WH, Hsu YJ, Cheng CP, Chou KN, Chen CL, Huang SM, Kan WC, Chiu YL. Assessing the Global Impact on the Mouse Kidney After Traumatic Brain Injury: A Transcriptomic Study. J Inflamm Res 2022; 15:4833-4851. [PMID: 36042866 PMCID: PMC9420446 DOI: 10.2147/jir.s375088] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 08/19/2022] [Indexed: 11/23/2022] Open
Abstract
Purpose In this study, we use animal models combined with bioinformatics strategies to investigate the potential changes in overall renal transcriptional expression after traumatic brain injury. Methods Microarray analysis was performed after kidney acquisition using unilateral controlled cortical impact as the primary mouse TBI model. Multi-oriented gene set enrichment analysis was performed for differentially expressed genes. Results The results showed that TBI affected the gene set associated with mitochondria function in kidney cells, and a negative enrichment of gene sets associated with immune cell migration and epidermal development was also observed. Analysis of the disease phenotype gene set revealed that differential expression of mitochondria-related genes was associated with lactate metabolism. Alternatively, activation and adhesion of immune cells associated with the complement system may promote autoinflammation in kidney tissue. The simulated immune cell infiltration analysis showed an increase in the proportion of activated memory CD4 T cells and a decrease in the proportion of resting memory CD4 T cells, suggesting that activated memory CD4 T cell infiltration may be involved in the inflammation of renal tissue and cause damage to renal cells, such as principal cells, mesangial cells and loops of Henle cells. Conclusion This study is the first to reveal the effects of brain trauma on the kidney. TBI may affect the expression of mitochondria function-related gene sets in renal cells by increasing lactate. It may also affect renal mesangial cells by inducing increased infiltration of immune cells through mechanisms related to complement system activation or autoimmune antibodies.
Collapse
Affiliation(s)
- Wei-Hung Chan
- Department of Anesthesiology, Tri-Service General Hospital, National Defense Medical Center, Taipei City, Taiwan, Republic of China.,Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei City, Taiwan, Republic of China
| | - Yu-Juei Hsu
- Division of Nephrology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei City, Taiwan, Republic of China
| | - Chiao-Pei Cheng
- Department of Anesthesiology, Tri-Service General Hospital, National Defense Medical Center, Taipei City, Taiwan, Republic of China
| | - Kuan-Nien Chou
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei City, Taiwan, Republic of China.,Department of Neurosurgery, Tri-Service General Hospital, National Defense Medical Center, Taipei City, Taiwan, Republic of China
| | - Chin-Li Chen
- Division of Urology, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei City, Taiwan, Republic of China
| | - Shih-Ming Huang
- Department of Biochemistry, National Defense Medical Center, Taipei City, Taiwan, Republic of China
| | - Wei-Chih Kan
- Department of Nephrology, Department of Internal Medicine, Chi-Mei Medical Center, Tainan City, Taiwan, Republic of China.,Department of Biological Science and Technology, Chung Hwa University of Medical Technology, Tainan City, Taiwan, Republic of China
| | - Yi-Lin Chiu
- Department of Biochemistry, National Defense Medical Center, Taipei City, Taiwan, Republic of China
| |
Collapse
|
14
|
Atalis A, Keenum MC, Pandey B, Beach A, Pradhan P, Vantucci C, O'Farrell L, Noel R, Jain R, Hosten J, Smith C, Kramer L, Jimenez A, Ochoa MA, Frey D, Roy K. Nanoparticle-delivered TLR4 and RIG-I agonists enhance immune response to SARS-CoV-2 subunit vaccine. J Control Release 2022; 347:476-488. [PMID: 35577151 PMCID: PMC9121740 DOI: 10.1016/j.jconrel.2022.05.023] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 04/04/2022] [Accepted: 05/10/2022] [Indexed: 01/25/2023]
Abstract
Despite success in vaccinating populations against SARS-CoV-2, concerns about immunity duration, continued efficacy against emerging variants, protection from infection and transmission, and worldwide vaccine availability remain. Molecular adjuvants targeting pattern recognition receptors (PRRs) on antigen-presenting cells (APCs) could improve and broaden the efficacy and durability of vaccine responses. Native SARS-CoV-2 infection stimulates various PRRs, including toll-like receptors (TLRs) and retinoic acid-inducible gene I (RIG-I)-like receptors. We hypothesized that targeting PRRs using molecular adjuvants on nanoparticles (NPs) along with a stabilized spike protein antigen could stimulate broad and efficient immune responses. Adjuvants targeting TLR4 (MPLA), TLR7/8 (R848), TLR9 (CpG), and RIG-I (PUUC) delivered on degradable polymer NPs were combined with the S1 subunit of spike protein and assessed in vitro with isogeneic mixed lymphocyte reactions (isoMLRs). For in vivo studies, the adjuvant-NPs were combined with stabilized spike protein or spike-conjugated NPs and assessed using a two-dose intranasal or intramuscular vaccination model in mice. Combination adjuvant-NPs simultaneously targeting TLR and RIG-I receptors (MPLA+PUUC, CpG+PUUC, and R848+PUUC) differentially induced T cell proliferation and increased proinflammatory cytokine secretion by APCs in vitro. When delivered intranasally, MPLA+PUUC NPs enhanced CD4+CD44+ activated memory T cell responses against spike protein in the lungs while MPLA NPs increased anti-spike IgA in the bronchoalveolar (BAL) fluid and IgG in the blood. Following intramuscular delivery, PUUC NPs induced strong humoral immune responses, characterized by increases in anti-spike IgG in the blood and germinal center B cell populations (GL7+ and BCL6+ B cells) in the draining lymph nodes (dLNs). MPLA+PUUC NPs further boosted spike protein-neutralizing antibody titers and T follicular helper cell populations in the dLNs. These results suggest that protein subunit vaccines with particle-delivered molecular adjuvants targeting TLR4 and RIG-I could lead to robust and unique route-specific adaptive immune responses against SARS-CoV-2.
Collapse
Affiliation(s)
- Alexandra Atalis
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Mark C Keenum
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Bhawana Pandey
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Alexander Beach
- School of Materials Science and Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Pallab Pradhan
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA; Marcus Center for Therapeutic Cell Characterization and Manufacturing, Georgia Institute of Technology, Atlanta, GA, USA; The Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
| | - Casey Vantucci
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Laura O'Farrell
- Physiological Research Laboratory, Georgia Institute of Technology, Atlanta, GA, USA
| | - Richard Noel
- Physiological Research Laboratory, Georgia Institute of Technology, Atlanta, GA, USA
| | - Ritika Jain
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Justin Hosten
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Clinton Smith
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Liana Kramer
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Angela Jimenez
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Miguel Armenta Ochoa
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - David Frey
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Krishnendu Roy
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA; Marcus Center for Therapeutic Cell Characterization and Manufacturing, Georgia Institute of Technology, Atlanta, GA, USA; The Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
15
|
Potential contribution of the immune system to the emergence of renal diseases. Immunol Lett 2022; 248:1-6. [DOI: 10.1016/j.imlet.2022.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 06/04/2022] [Indexed: 11/21/2022]
|
16
|
Volpedo G, Pacheco-Fernandez T, Holcomb EA, Zhang WW, Lypaczewski P, Cox B, Fultz R, Mishan C, Verma C, Huston RH, Wharton AR, Dey R, Karmakar S, Oghumu S, Hamano S, Gannavaram S, Nakhasi HL, Matlashewski G, Satoskar AR. Centrin-deficient Leishmania mexicana confers protection against New World cutaneous leishmaniasis. NPJ Vaccines 2022; 7:32. [PMID: 35236861 PMCID: PMC8891280 DOI: 10.1038/s41541-022-00449-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 01/27/2022] [Indexed: 01/01/2023] Open
Abstract
Leishmaniasis is a neglected protozoan disease affecting over 12 million people globally with no approved vaccines for human use. New World cutaneous leishmaniasis (CL) caused by L. mexicana is characterized by the development of chronic non-healing skin lesions. Using the CRISPR/Cas9 technique, we have generated live attenuated centrin knockout L. mexicana (LmexCen-/-) parasites. Centrin is a cytoskeletal protein important for cellular division in eukaryotes and, in Leishmania, is required only for intracellular amastigote replication. We have investigated the safety and immunogenicity characteristics of LmexCen-/- parasites by evaluating their survival and the cytokine production in bone-marrow-derived macrophages (BMDMs) and dendritic cells (BMDCs) in vitro. Our data shows that LmexCen-/- amastigotes present a growth defect, which results in significantly lower parasitic burdens and increased protective cytokine production in infected BMDMs and BMDCs, compared to the wild type (WT) parasites. We have also determined the safety and efficacy of LmexCen-/- in vivo using experimental murine models of L. mexicana. We demonstrate that LmexCen-/- parasites are safe and do not cause lesions in susceptible mouse models. Immunization with LmexCen-/- is also efficacious against challenge with WT L. mexicana parasites in genetically different BALB/c and C57BL/6 mouse models. Vaccinated mice did not develop cutaneous lesions, displayed protective immunity, and showed significantly lower parasitic burdens at the infection site and draining lymph nodes compared to the control group. Overall, we demonstrate that LmexCen-/- parasites are safe and efficacious against New World cutaneous leishmaniasis in pre-clinical models.
Collapse
Affiliation(s)
- Greta Volpedo
- Department of Microbiology, The Ohio State University, Columbus, OH, 43210, USA.,Department of Pathology, Wexner Medical Center, The Ohio State University, Columbus, OH, 43210, USA
| | - Thalia Pacheco-Fernandez
- Department of Pathology, Wexner Medical Center, The Ohio State University, Columbus, OH, 43210, USA
| | - Erin A Holcomb
- Department of Pathology, Wexner Medical Center, The Ohio State University, Columbus, OH, 43210, USA
| | - Wen-Wei Zhang
- Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
| | - Patrick Lypaczewski
- Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
| | - Blake Cox
- Department of Pathology, Wexner Medical Center, The Ohio State University, Columbus, OH, 43210, USA
| | - Rebecca Fultz
- Department of Pathology, Wexner Medical Center, The Ohio State University, Columbus, OH, 43210, USA
| | - Chelsea Mishan
- Department of Pathology, Wexner Medical Center, The Ohio State University, Columbus, OH, 43210, USA
| | - Chaitenya Verma
- Department of Pathology, Wexner Medical Center, The Ohio State University, Columbus, OH, 43210, USA
| | - Ryan H Huston
- Department of Pathology, Wexner Medical Center, The Ohio State University, Columbus, OH, 43210, USA
| | - Abigail R Wharton
- Department of Pathology, Wexner Medical Center, The Ohio State University, Columbus, OH, 43210, USA
| | - Ranadhir Dey
- Division of Emerging and Transfusion Transmitted Diseases, CBER, FDA, Silver Spring, MD, USA
| | - Subir Karmakar
- Division of Emerging and Transfusion Transmitted Diseases, CBER, FDA, Silver Spring, MD, USA
| | - Steve Oghumu
- Department of Pathology, Wexner Medical Center, The Ohio State University, Columbus, OH, 43210, USA
| | - Shinjiro Hamano
- Department of Parasitology, Institute of Tropical Medicine (NEKKEN), The Joint Usage/Research Center on Tropical Disease, Nagasaki University, Nagasaki University Graduate School of Biomedical Sciences Doctoral Leadership Program, Nagasaki, Japan
| | - Sreenivas Gannavaram
- Division of Emerging and Transfusion Transmitted Diseases, CBER, FDA, Silver Spring, MD, USA
| | - Hira L Nakhasi
- Division of Emerging and Transfusion Transmitted Diseases, CBER, FDA, Silver Spring, MD, USA.
| | - Greg Matlashewski
- Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada.
| | - Abhay R Satoskar
- Department of Microbiology, The Ohio State University, Columbus, OH, 43210, USA. .,Department of Pathology, Wexner Medical Center, The Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
17
|
Atalis A, Keenum MC, Pandey B, Beach A, Pradhan P, Vantucci C, Jain R, Hosten J, Smith C, Kramer L, Jimenez A, Ochoa MA, Frey D, Roy K. Nanoparticle-delivered TLR4 and RIG-I agonists enhance immune response to SARS-CoV-2 subunit vaccine. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.01.31.478507. [PMID: 35132413 PMCID: PMC8820660 DOI: 10.1101/2022.01.31.478507] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Despite recent success in vaccinating populations against SARS-CoV-2, concerns about immunity duration, continued efficacy against emerging variants, protection from infection and transmission, and worldwide vaccine availability, remain. Although mRNA, pDNA, and viral-vector based vaccines are being administered, no protein subunit-based SARS-CoV-2 vaccine is approved. Molecular adjuvants targeting pathogen-recognition receptors (PRRs) on antigen-presenting cells (APCs) could improve and broaden the efficacy and durability of vaccine responses. Native SARS-CoV-2 infection stimulate various PRRs, including toll-like receptors (TLRs) and retinoic-acid-inducible gene I-like receptors (RIG-I). We hypothesized that targeting the same PRRs using adjuvants on nanoparticles along with a stabilized spike (S) protein antigen could provide broad and efficient immune responses. Formulations targeting TLR4 (MPLA), TLR7/8 (R848), TLR9 (CpG), and RIG-I (PUUC) delivered on degradable polymer-nanoparticles (NPs) were combined with the S1 subunit of S protein and assessed in vitro with isogeneic mixed lymphocyte reactions (iso-MLRs). For in vivo studies, the adjuvanted nanoparticles were combined with stabilized S protein and assessed using intranasal and intramuscular prime-boost vaccination models in mice. Combination NP-adjuvants targeting both TLR and RIG-I (MPLA+PUUC, CpG+PUUC, or R848+PUUC) differentially increased proinflammatory cytokine secretion (IL-1β, IL-12p70, IL-27, IFN-β) by APCs cultured in vitro, and induced differential T cell proliferation. When delivered intranasally, MPLA+PUUC NPs enhanced local CD4+CD44+ activated memory T cell responses while MPLA NPs increased anti-S-protein-specific IgG and IgA in the lung. Following intramuscular delivery, PUUC-carrying NPs induced strong humoral immune responses, characterized by increases in anti-S-protein IgG and neutralizing antibody titers and germinal center B cell populations (GL7+ and BCL6+ B cells). MPLA+PUUC NPs further boosted S-protein-neutralizing antibody titers and T follicular helper cell populations in draining lymph nodes. These results suggest that SARS-CoV-2-mimicking adjuvants and subunit vaccines could lead to robust and unique route-specific adaptive immune responses and may provide additional tools against the pandemic.
Collapse
|
18
|
Chung NH, Chen YC, Yang SJ, Lin YC, Dou HY, Hui-Ching Wang L, Liao CL, Chow YH. Induction of Th1 and Th2 in the protection against SARS-CoV-2 through mucosal delivery of an adenovirus vaccine expressing an engineered spike protein. Vaccine 2021; 40:574-586. [PMID: 34952759 PMCID: PMC8677488 DOI: 10.1016/j.vaccine.2021.12.024] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 11/01/2021] [Accepted: 12/12/2021] [Indexed: 12/21/2022]
Abstract
A series of recombinant human type 5 adenoviruses that express the full-length or membrane-truncated spike protein (S) of SARS-CoV-2 (AdCoV2-S or AdCoV2-SdTM, respectively) was tested the efficacy against SARS-CoV-2 via intranasal (i.n.) or subcutaneous (s.c.) immunization in a rodent model. Mucosal delivery of adenovirus (Ad) vaccines could induce anti-SARS-CoV-2 IgG and IgA in the serum and in the mucosal, respectively as indicated by vaginal wash (vw) and bronchoalveolar lavage fluid (BALF). Serum anti-SARS-CoV-2 IgG but not IgA in the vw and BALF was induced by AdCoV2-S s.c.. Administration of AdCoV2-S i.n. was able to induce higher anti-SARS-CoV-2 binding and neutralizing antibody levels than s.c. injection. AdCoV2-SdTM i.n. induced a lower antibody responses than AdCoV2-S i.n.. Induced anti-S antibody responses by AdCoV2-S via i.n. or s.c. were not influenced by the pre-existing serum anti-Ad antibody. Novelty, S-specific IgG1 which represented Th2-mediated humoral response was dominantly induced in Ad i.n.-immunized serum in contrast to more IgG2a which represented Th1-mediated cellular response found in Ad s.c.-immunized serum. The activation of S-specific IFN-ɣ and IL-4 in splenic Th1 and Th2 cells, respectively, was observed in the AdCoV2-S i.n. and s.c. groups, indicating the Th1 and Th2 immunity were activated. AdCoV2-S and AdCoV2-SdTM significantly prevented body weight loss and reduced pulmonary viral loads in hamsters. A reduction in inflammation in the lungs was observed in AdCoV-S via i.n. or s.c.-immunized hamsters following a SARS-CoV-2 challenge. It correlated to Th1 cytokine but no inflammatory cytokines secretions found in AdCoV-S i.n. -immunized BALF. These results indicate that intranasal delivery of AdCoV2-S vaccines is safe and potent at preventing SARS-CoV-2 infections.
Collapse
Affiliation(s)
- Nai-Hsiang Chung
- National Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Zhunan, Taiwan; Graduate Program of Biotechnology in Medicine, National Tsing Hua University, Hsinchu, Taiwan; Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Ying-Chin Chen
- National Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Zhunan, Taiwan
| | - Shiu-Ju Yang
- National Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Zhunan, Taiwan
| | - Yu-Ching Lin
- National Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Zhunan, Taiwan
| | - Horng-Yunn Dou
- National Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Zhunan, Taiwan
| | - Lily Hui-Ching Wang
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Ching-Len Liao
- National Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Zhunan, Taiwan
| | - Yen-Hung Chow
- National Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Zhunan, Taiwan; Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.
| |
Collapse
|
19
|
Deb C, Salinas AN, Zheng T, Middleton A, Kern K, Penoyer D, Borsadia R, Hunley C, Abomoelak B, Mehta V, Irastorza L, Mehta DI, Huo Q. A 1-minute blood test detects decreased immune function and increased clinical risk in COVID-19 patients. Sci Rep 2021; 11:23491. [PMID: 34873223 PMCID: PMC8648859 DOI: 10.1038/s41598-021-02863-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 11/11/2021] [Indexed: 01/08/2023] Open
Abstract
Upon infection with SARS-CoV-2, the virus that causes COVID-19, most people will develop no or mild symptoms. However, a small percentage of the population will become severely ill, and some will succumb to death. The clinical severity of COVID-19 has a close connection to the dysregulation of the patient's immune functions. We previously developed a simple, nanoparticle-enabled blood test that can determine the humoral immune status in animals. In this study, we applied this new test to analyze the immune function in relation to disease severity in COVID-19 patients. From the testing of 153 COVID-19 patient samples and 142 negative controls, we detected a drastic decrease of humoral immunity in COVID-19 patients who developed moderate to severe symptoms, but not in patients with no or mild symptoms. The new test may be potentially used to monitor the immunity change and predict the clinical risk of patients with COVID-19.
Collapse
Affiliation(s)
- Chirajyoti Deb
- Translational Research and Specialty Diagnostic Laboratory, Arnold Palmer Hospital for Children, Orlando Health, 110 Bonnie Loch Court, Orlando, FL, 32806, USA.
| | - Allan N Salinas
- Translational Research and Specialty Diagnostic Laboratory, Arnold Palmer Hospital for Children, Orlando Health, 110 Bonnie Loch Court, Orlando, FL, 32806, USA
| | - Tianyu Zheng
- Nano Discovery Inc., 1060 Woodcock Road Suite 131, Orlando, FL, 32803, USA
| | - Aurea Middleton
- Center for Nursing Research, Orlando Health, 1414 Kuhl Ave, Orlando, FL, 32806, USA
| | - Katelyn Kern
- Center for Digestive Health and Nutrition, Arnold Palmer Hospital for Children, Orlando Health, 60 Gore St., Orlando, FL, 32806, USA
| | - Daleen Penoyer
- Center for Nursing Research, Orlando Health, 1414 Kuhl Ave, Orlando, FL, 32806, USA
| | - Rahul Borsadia
- Internal Medicine Group, Orlando Health, 1414 Kuhl Ave, Orlando, FL, 32806, USA
| | - Charles Hunley
- Critical Care Medicine, Orlando Health, 1414 Kuhl Ave, Orlando, FL, 32806, USA
| | - Bassam Abomoelak
- Translational Research and Specialty Diagnostic Laboratory, Arnold Palmer Hospital for Children, Orlando Health, 110 Bonnie Loch Court, Orlando, FL, 32806, USA
| | - Vijay Mehta
- Center for Digestive Health and Nutrition, Arnold Palmer Hospital for Children, Orlando Health, 60 Gore St., Orlando, FL, 32806, USA
| | - Laura Irastorza
- Center for Digestive Health and Nutrition, Arnold Palmer Hospital for Children, Orlando Health, 60 Gore St., Orlando, FL, 32806, USA
| | - Devendra I Mehta
- Translational Research and Specialty Diagnostic Laboratory, Arnold Palmer Hospital for Children, Orlando Health, 110 Bonnie Loch Court, Orlando, FL, 32806, USA.
- Center for Digestive Health and Nutrition, Arnold Palmer Hospital for Children, Orlando Health, 60 Gore St., Orlando, FL, 32806, USA.
| | - Qun Huo
- Department of Chemistry and NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL, 32826, USA.
| |
Collapse
|
20
|
Valera-Vera EA, Concepción JL, Cáceres AJ, Acevedo GR, Fernández M, Hernández Y, Digirolamo FA, Duschak VG, Soprano LL, Pereira CA, Miranda MR, Gómez KA. IgE antibodies against Trypanosoma cruzi arginine kinase in patients with chronic Chagas disease. Mol Immunol 2021; 138:68-75. [PMID: 34364074 DOI: 10.1016/j.molimm.2021.06.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 06/15/2021] [Accepted: 06/29/2021] [Indexed: 11/17/2022]
Abstract
Arginine kinase (AK) is an enzyme present in various invertebrates, as well as in some trypanosomatids such as T. cruzi, the etiological agent that causes Chagas disease. In invertebrates, this protein acts as an allergen inducing an IgE-type humoral immune response. Since AK is a highly conserved protein, we decided to study whether patients with chronic Chagas disease (CCD) produce specific antibodies against T. cruzi AK (TcAK). Plasma from patients with CCD, with and without cardiac alterations and non-infected individuals were evaluated for the presence of anti-TcAK IgG and IgE antibodies by ELISA, including detection of specific IgG subclasses. Our results showed that the levels of specific anti-TcAK IgG and IgE were different between infected and non-infected individuals, but comparable between those with different clinical manifestations. Interestingly, anti-TcAK IgG4 antibodies associated with IgE-mediated allergenic processes were also increased in CCD patients. Finally, we found that several of the predicted B cell epitopes in TcAK matched allergenic peptides previously described for its homologues in other organisms. Our results revealed for the first time a parasite's specific IgE antibody target and suggest that TcAK could contribute to delineate an inefficient B cell response by prompting a bias towards a Th2 profile. These findings also shed light on a potential allergenic response in the context of T. cruzi infection.
Collapse
Affiliation(s)
- Edward Augusto Valera-Vera
- Universidad de Buenos Aires, Facultad de Medicina, Instituto de Investigaciones Médicas "Alfredo Lanari", Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Instituto de Investigaciones Médicas (IDIM), Laboratorio de Parasitología Molecular, Buenos Aires, Argentina
| | - Juan Luis Concepción
- Laboratorio de Enzimología de Parásitos, Departamento de Biología, Facultad de Ciencias, Universidad de Los Andes, Mérida, 5101, Venezuela
| | - Ana Judith Cáceres
- Laboratorio de Enzimología de Parásitos, Departamento de Biología, Facultad de Ciencias, Universidad de Los Andes, Mérida, 5101, Venezuela
| | - Gonzalo Raúl Acevedo
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI-CONICET), Buenos Aires, Argentina
| | - Marisa Fernández
- Instituto Nacional de Parasitología "Dr. Mario Fatala Chabén", ANLIS-Malbrán, Ministerio de Salud, Buenos Aires, Argentina
| | - Yolanda Hernández
- Instituto Nacional de Parasitología "Dr. Mario Fatala Chabén", ANLIS-Malbrán, Ministerio de Salud, Buenos Aires, Argentina
| | - Fabio Augusto Digirolamo
- Universidad de Buenos Aires, Facultad de Medicina, Instituto de Investigaciones Médicas "Alfredo Lanari", Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Instituto de Investigaciones Médicas (IDIM), Laboratorio de Parasitología Molecular, Buenos Aires, Argentina
| | - Vilma Gladys Duschak
- Área de Bioquímica de Proteínas y Glicobiología de Parásitos, Departamento de Investigación, Instituto Nacional de Parasitología "Dr. Mario Fatala Chaben", ANLIS-Malbrán, Ministerio de Salud de la Nación, Buenos Aires, Argentina
| | - Luciana Lía Soprano
- Área de Bioquímica de Proteínas y Glicobiología de Parásitos, Departamento de Investigación, Instituto Nacional de Parasitología "Dr. Mario Fatala Chaben", ANLIS-Malbrán, Ministerio de Salud de la Nación, Buenos Aires, Argentina
| | - Claudio Alejandro Pereira
- Universidad de Buenos Aires, Facultad de Medicina, Instituto de Investigaciones Médicas "Alfredo Lanari", Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Instituto de Investigaciones Médicas (IDIM), Laboratorio de Parasitología Molecular, Buenos Aires, Argentina
| | - Mariana Reneé Miranda
- Universidad de Buenos Aires, Facultad de Medicina, Instituto de Investigaciones Médicas "Alfredo Lanari", Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Instituto de Investigaciones Médicas (IDIM), Laboratorio de Parasitología Molecular, Buenos Aires, Argentina
| | - Karina Andrea Gómez
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI-CONICET), Buenos Aires, Argentina.
| |
Collapse
|
21
|
Rio-Aige K, Azagra-Boronat I, Castell M, Selma-Royo M, Collado MC, Rodríguez-Lagunas MJ, Pérez-Cano FJ. The Breast Milk Immunoglobulinome. Nutrients 2021; 13:nu13061810. [PMID: 34073540 PMCID: PMC8230140 DOI: 10.3390/nu13061810] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 05/19/2021] [Accepted: 05/23/2021] [Indexed: 12/24/2022] Open
Abstract
Breast milk components contribute to the infant’s immune development and protection, and among other immune factors, immunoglobulins (Igs) are the most studied. The presence of IgA in milk has been known for a long time; however, less information is available about the presence of other Igs such as IgM, IgG, and their subtypes (IgG1, IgG2, IgG3, and IgG4) or even IgE or IgD. The total Ig concentration and profile will change during the course of lactation; however, there is a great variability among studies due to several variables that limit establishing a clear pattern. In this context, the aim of this review was firstly to shed light on the Ig concentration in breast milk based on scientific evidence and secondly to study the main factors contributing to such variability. A search strategy provided only 75 studies with the prespecified eligibility criteria. The concentrations and proportions found have been established based on the intrinsic factors of the study—such as the sampling time and quantification technique—as well as participant-dependent factors, such as lifestyle and environment. All these factors contribute to the variability of the immunoglobulinome described in the literature and should be carefully addressed for further well-designed studies and data interpretation.
Collapse
Affiliation(s)
- Karla Rio-Aige
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (K.R.-A.); (I.A.-B.); (M.C.); (M.J.R.-L.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Ignasi Azagra-Boronat
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (K.R.-A.); (I.A.-B.); (M.C.); (M.J.R.-L.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Margarida Castell
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (K.R.-A.); (I.A.-B.); (M.C.); (M.J.R.-L.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Marta Selma-Royo
- Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), 46890 Paterna, Valencia, Spain; (M.S.-R.); (M.C.C.)
| | - María Carmen Collado
- Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), 46890 Paterna, Valencia, Spain; (M.S.-R.); (M.C.C.)
| | - María J. Rodríguez-Lagunas
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (K.R.-A.); (I.A.-B.); (M.C.); (M.J.R.-L.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Francisco J. Pérez-Cano
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (K.R.-A.); (I.A.-B.); (M.C.); (M.J.R.-L.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
- Correspondence: ; Tel.: +34-934-024-505
| |
Collapse
|
22
|
Waag DM, Chance TB, Trevino SR, Rossi FD, Fetterer DP, Amemiya K, Dankmeyer JL, Ingavale SS, Tobery SA, Zeng X, Kern SJ, Worsham PL, Cote CK, Welkos SL. Comparison of three non-human primate aerosol models for glanders, caused by Burkholderia mallei. Microb Pathog 2021; 155:104919. [PMID: 33915206 DOI: 10.1016/j.micpath.2021.104919] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 03/30/2021] [Accepted: 04/01/2021] [Indexed: 01/15/2023]
Abstract
Burkholderia mallei is a gram-negative obligate animal pathogen that causes glanders, a highly contagious and potentially fatal disease of solipeds including horses, mules, and donkeys. Humans are also susceptible, and exposure can result in a wide range of clinical forms, i.e., subclinical infection, chronic forms with remission and exacerbation, or acute and potentially lethal septicemia and/or pneumonia. Due to intrinsic antibiotic resistance and the ability of the organisms to survive intracellularly, current treatment regimens are protracted and complicated; and no vaccine is available. As a consequence of these issues, and since B. mallei is infectious by the aerosol route, B. mallei is regarded as a major potential biothreat agent. To develop optimal medical countermeasures and diagnostic tests, well characterized animal models of human glanders are needed. The goal of this study was to perform a head-to-head comparison of models employing three commonly used nonhuman primate (NHP) species, the African green monkey (AGM), Rhesus macaque, and the Cynomolgus macaque. The natural history of infection and in vitro clinical, histopathological, immunochemical, and bacteriological parameters were examined. The AGMs were the most susceptible NHP to B. mallei; five of six expired within 14 days. Although none of the Rhesus or Cynomolgus macaques succumbed, the Rhesus monkeys exhibited abnormal signs and clinical findings associated with B. mallei infection; and the latter may be useful for modeling chronic B. mallei infection. Based on the disease progression observations, gross and histochemical pathology, and humoral and cellular immune response findings, the AGM appears to be the optimal model of acute, lethal glanders infection. AGM models of infection by B. pseudomallei, the etiologic agent of melioidosis, have been characterized recently. Thus, the selection of the AGM species provides the research community with a single NHP model for investigations on acute, severe, inhalational melioidosis and glanders.
Collapse
Affiliation(s)
- David M Waag
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD, USA
| | - Taylor B Chance
- Pathology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD, USA
| | - Sylvia R Trevino
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD, USA
| | - Franco D Rossi
- Applied and Advanced Technology-Aerobiology, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD, USA
| | - David P Fetterer
- Biostatistics Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD, USA
| | - Kei Amemiya
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD, USA
| | - Jennifer L Dankmeyer
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD, USA
| | - Susham S Ingavale
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD, USA
| | - Steven A Tobery
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD, USA
| | - Xiankun Zeng
- Pathology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD, USA
| | - Steven J Kern
- Biostatistics Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD, USA
| | - Patricia L Worsham
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD, USA
| | - Christopher K Cote
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD, USA.
| | - Susan L Welkos
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD, USA.
| |
Collapse
|
23
|
Peddapalli A, Gehani M, Kalle AM, Peddapalli SR, Peter AE, Sharad S. Demystifying Excess Immune Response in COVID-19 to Reposition an Orphan Drug for Down-Regulation of NF-κB: A Systematic Review. Viruses 2021; 13:378. [PMID: 33673529 PMCID: PMC7997247 DOI: 10.3390/v13030378] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 02/12/2021] [Accepted: 02/23/2021] [Indexed: 12/18/2022] Open
Abstract
The immunological findings from autopsies, biopsies, and various studies in COVID-19 patients show that the major cause of morbidity and mortality in COVID-19 is excess immune response resulting in hyper-inflammation. With the objective to review various mechanisms of excess immune response in adult COVID-19 patients, Pubmed was searched for free full articles not related to therapeutics or co-morbid sub-groups, published in English until 27.10.2020, irrespective of type of article, country, or region. Joanna Briggs Institute's design-specific checklists were used to assess the risk of bias. Out of 122 records screened for eligibility, 42 articles were included in the final review. The review found that eventually, most mechanisms result in cytokine excess and up-regulation of Nuclear Factor-κB (NF-κB) signaling as a common pathway of excess immune response. Molecules blocking NF-κB or targeting downstream effectors like Tumour Necrosis Factor α (TNFα) are either undergoing clinical trials or lack specificity and cause unwanted side effects. Neutralization of upstream histamine by histamine-conjugated normal human immunoglobulin has been demonstrated to inhibit the nuclear translocation of NF-κB, thereby preventing the release of pro-inflammatory cytokines Interleukin (IL) 1β, TNF-α, and IL-6 and IL-10 in a safer manner. The authors recommend repositioning it in COVID-19.
Collapse
Affiliation(s)
- Apparao Peddapalli
- Department of Microbiology, King George Hospital, Visakhapatnam 531011, Andhra Pradesh, India;
| | - Manish Gehani
- Department of Biological Sciences, Birla Institute of Technology and Science, Pilani-Hyderabad Campus, Hyderabad 500078, Telangana, India;
| | - Arunasree M. Kalle
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad 500046, Telangana, India;
| | - Siva R. Peddapalli
- Department of Biological Sciences-Biotechnology, Florida Institute of Technology, Melbourne, FL 32901, USA;
| | - Angela E. Peter
- Department of Biotechnology, College of Science & Technology, Andhra University, Visakhapatnam 530003, Andhra Pradesh, India;
| | - Shashwat Sharad
- Center for Prostate Disease Research, John P. Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, Bethesda, MD 20817, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817, USA
| |
Collapse
|
24
|
Rio-Aige K, Azagra-Boronat I, Massot-Cladera M, Selma-Royo M, Parra-Llorca A, González S, García-Mantrana I, Castell M, Rodríguez-Lagunas MJ, Collado MC, Pérez Cano FJ. Association of Maternal Microbiota and Diet in Cord Blood Cytokine and Immunoglobulin Profiles. Int J Mol Sci 2021; 22:ijms22041778. [PMID: 33579027 PMCID: PMC7916816 DOI: 10.3390/ijms22041778] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/04/2021] [Accepted: 02/06/2021] [Indexed: 12/16/2022] Open
Abstract
Mothers confer natural passive immunization to their infants through the transplacental pathway during the gestation period. The objective of the present study was to establish at birth the maternal and cord plasma concentration and relationship of immunoglobulins (Igs), cytokines (CKs), and adipokines. In addition, the impact of the maternal microbiota and diet was explored. The plasma profile of these components was different between mothers and babies, with the levels of many CKs, IgM, IgG2a, IgE, IgA, and leptin significantly higher in mothers than in the cord sample. Moreover, the total Igs, all IgG subtypes, IgE, and the Th1/Th2 ratio positively correlated in the mother–infant pair. Maternal dietary components such as monounsaturated fatty acids-polyunsaturated fatty acids and fiber were positively associated with some immune factors such as IgA in cord samples. The microbiota composition clustering also influenced the plasma profile of some factors (i.e., many CKs, some Ig, and adiponectin). In conclusion, we have established the concentration of these immunomodulatory factors in the maternal–neonatal pair at birth, some positive associations, and the influence of maternal diet and the microbiota composition, suggesting that the immune status during pregnancy, in terms of CKs and Igs levels, can influence the immune status of the infant at birth.
Collapse
Affiliation(s)
- Karla Rio-Aige
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (K.R.-A.); (I.A.-B.); (M.M.-C.); (M.C.); (F.J.P.C.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Ignasi Azagra-Boronat
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (K.R.-A.); (I.A.-B.); (M.M.-C.); (M.C.); (F.J.P.C.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Malén Massot-Cladera
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (K.R.-A.); (I.A.-B.); (M.M.-C.); (M.C.); (F.J.P.C.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Marta Selma-Royo
- Institute of Agrochemistry and Food Technology (IATA-CSIC), National Research Council, 46980 Valencia, Spain; (M.S.-R.); (I.G.-M.); (M.C.C.)
| | - Anna Parra-Llorca
- Neonatal Research Group, Health Research Institute La Fe, 46026 Valencia, Spain;
| | - Sonia González
- Department of Functional Biology, Faculty of Medicine, University of Oviedo, 33006 Oviedo, Spain;
- Diet, Microbiota and Health Group, Instituto de Investigación Sanitaria del Principado de Asturias (DIMISA, ISPA), 33011 Oviedo, Spain
| | - Izaskun García-Mantrana
- Institute of Agrochemistry and Food Technology (IATA-CSIC), National Research Council, 46980 Valencia, Spain; (M.S.-R.); (I.G.-M.); (M.C.C.)
| | - Margarida Castell
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (K.R.-A.); (I.A.-B.); (M.M.-C.); (M.C.); (F.J.P.C.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - María J. Rodríguez-Lagunas
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (K.R.-A.); (I.A.-B.); (M.M.-C.); (M.C.); (F.J.P.C.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
- Correspondence:
| | - María Carmen Collado
- Institute of Agrochemistry and Food Technology (IATA-CSIC), National Research Council, 46980 Valencia, Spain; (M.S.-R.); (I.G.-M.); (M.C.C.)
| | - Francisco José Pérez Cano
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (K.R.-A.); (I.A.-B.); (M.M.-C.); (M.C.); (F.J.P.C.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| |
Collapse
|
25
|
Steinbuck MP, Seenappa LM, Jakubowski A, McNeil LK, Haqq CM, DeMuth PC. A lymph node-targeted Amphiphile vaccine induces potent cellular and humoral immunity to SARS-CoV-2. SCIENCE ADVANCES 2021; 7:7/6/eabe5819. [PMID: 33547083 PMCID: PMC7864572 DOI: 10.1126/sciadv.abe5819] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 12/17/2020] [Indexed: 05/17/2023]
Abstract
The profound consequences of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) mandate urgent development of effective vaccines. Here, we evaluated an Amphiphile (AMP) vaccine adjuvant, AMP-CpG, composed of diacyl lipid-modified CpG, admixed with the SARS-CoV-2 Spike-2 receptor binding domain protein as a candidate vaccine (ELI-005) in mice. AMP modification efficiently delivers CpG to lymph nodes, where innate and adaptive immune responses are generated. Compared to alum, immunization with AMP-CpG induced >25-fold higher antigen-specific T cells that produced multiple T helper 1 (TH1) cytokines and trafficked into lung parenchyma. Antibody responses favored TH1 isotypes (IgG2c and IgG3) and potently neutralized Spike-2-ACE2 receptor binding, with titers 265-fold higher than natural convalescent patient COVID-19 responses; T cell and antibody responses were maintained despite 10-fold dose reduction in Spike antigen. Both cellular and humoral immune responses were preserved in aged mice. These advantages merit clinical translation to SARS-CoV-2 and other protein subunit vaccines.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Animals
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/immunology
- COVID-19/prevention & control
- COVID-19/virology
- COVID-19 Vaccines/administration & dosage
- COVID-19 Vaccines/immunology
- Female
- HEK293 Cells
- Humans
- Immunity, Cellular
- Immunity, Humoral
- Immunogenicity, Vaccine
- Lymph Nodes/immunology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Neutralization Tests
- Oligodeoxyribonucleotides/administration & dosage
- Oligodeoxyribonucleotides/immunology
- Protein Interaction Domains and Motifs/immunology
- SARS-CoV-2/immunology
- Spike Glycoprotein, Coronavirus/chemistry
- Spike Glycoprotein, Coronavirus/immunology
- Surface-Active Agents/administration & dosage
- Treatment Outcome
- Vaccination/methods
- Vaccines, Subunit/immunology
Collapse
Affiliation(s)
- Martin P Steinbuck
- Elicio Therapeutics, One Kendall Square, Suite 14303, Cambridge, MA 02139, USA
| | - Lochana M Seenappa
- Elicio Therapeutics, One Kendall Square, Suite 14303, Cambridge, MA 02139, USA
| | - Aniela Jakubowski
- Elicio Therapeutics, One Kendall Square, Suite 14303, Cambridge, MA 02139, USA
| | - Lisa K McNeil
- Elicio Therapeutics, One Kendall Square, Suite 14303, Cambridge, MA 02139, USA
| | - Christopher M Haqq
- Elicio Therapeutics, One Kendall Square, Suite 14303, Cambridge, MA 02139, USA.
| | - Peter C DeMuth
- Elicio Therapeutics, One Kendall Square, Suite 14303, Cambridge, MA 02139, USA
| |
Collapse
|
26
|
Malignancy-associated membranous nephropathy with PLA2R double-positive for glomeruli and carcinoma. CEN Case Rep 2021; 10:281-286. [PMID: 33393071 DOI: 10.1007/s13730-020-00556-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 11/13/2020] [Indexed: 10/22/2022] Open
Abstract
Phospholipase A2 receptor (PLA2R) is the most common primary target antigen of idiopathic membranous nephropathy (MN) although PLA2R antibodies are also reported to be present in malignancy-associated MN. However, a case of PLA2R-positive MN secondary to PLA2R-positive carcinoma has not been reported. A 26-year-old Japanese woman presented with general fatigue, fever, and nonproductive cough. Computed tomography demonstrated a left kidney mass with pathologic diagnosis of Xp11.2 translocation renal cell carcinoma (RCC). After the second time of administration with Sunitinib, the patients exhibited significant proteinuria and hypoalbuminemia. Renal biopsy revealed a diagnosis of diffuse MN secondary to RCC. Immunofluorescence staining showed granular patterns positive for immunoglobulin (Ig) G, IgA, and C3c. PLA2R and IgG1-3 were positive, while IgG4 was negative. For the treatment of severe nephrotic syndrome, we attempted steroid therapy without any clinical improvement. Open nephrectomy was performed and surprisingly, RCC was stained for PLA2R with polarity for the basal side. At outpatient follow-up, 4 months after the operation, urinary protein had still persisted, although serum albumin was slightly increased. We report a case of PLA2R-positive MN secondary to PLA2R-positive RCC.
Collapse
|
27
|
Myrcene Attenuates Renal Inflammation and Oxidative Stress in the Adrenalectomized Rat Model. Molecules 2020; 25:molecules25194492. [PMID: 33007969 PMCID: PMC7582976 DOI: 10.3390/molecules25194492] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 09/21/2020] [Accepted: 09/25/2020] [Indexed: 11/16/2022] Open
Abstract
Physiological Glucocorticoids are important regulators of the immune system. Pharmacological GCs are in widespread use to treat inflammatory diseases. Adrenalectomy (ADX) has been shown to exacerbate renal injury through inflammation and oxidative stress that results in renal impairment due to depletion of GCs. In this study, the effect of myrcene to attenuate renal inflammation and oxidative stress was evaluated in the adrenalectomized rat model. Rats were adrenalectomized bilaterally or the adrenals were not removed after surgery (sham). Myrcene (50 mg/kg body weight, orally) was administered post ADX. Myrcene treatment resulted in significant downregulation of pro-inflammatory cytokines (IL-1β, IL-6, and TNF-α) compared to untreated ADX rats. In addition, myrcene resulted in significant downregulation of immunomodulatory factors (IFNγ and NF-κB) and anti-inflammatory markers (IL-4 and IL-10) in treated ADX compared to untreated ADX. Myrcene significantly increased the antioxidant molecules (CAT, GSH, and SOD) and decreased MDA levels in treated ADX compared to untreated. Moreover, myrcene treatment reduced the expression of COX-2, iNOS, KIM-1, and kidney functional molecules (UREA, LDH, total protein, and creatinine) in ADX treated compared to ADX untreated. These results suggest that myrcene could be further developed as a therapeutic drug for treatment of kidney inflammation and injury.
Collapse
|
28
|
Itoh I, Kasuno K, Yamamoto C, Takahashi N, Shimizu H, Ojima T, Hayashi S, Kimura H, Iwano M. IgA Vasculitis Developed as an Adverse Effect of Tofacitinib Taken for Rheumatoid Arthritis. Intern Med 2020; 59:817-821. [PMID: 31813912 PMCID: PMC7118374 DOI: 10.2169/internalmedicine.3668-19] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Tofacitinib is a new small-molecule inhibitor of the JAK/STAT signaling pathway used to treat rheumatoid arthritis. We herein report a case of IgA vasculitis apparently caused by tofacitinib. A 67-year-old woman with rheumatoid arthritis developed IgA vasculitis after taking tofacitinib for 6 months. She presented with proteinuria and purpura of the lower extremities. Biopsy specimens from her skin and kidney were compatible with IgA vasculitis. Following termination of tofacitinib, the patient completely recovered from the IgA vasculitis. Drug-induced IgA vasculitis has been previously described for anti-tumor necrosis factor-(TNF)α therapies, but this is the first report of this adverse effect with anti-JAK therapy.
Collapse
Affiliation(s)
- Izumi Itoh
- Department of Internal Medicine, Fukui General Hospital, Japan
| | - Kenji Kasuno
- Department of Nephrology, Faculty of Medical Sciences, University of Fukui, Japan
| | - Chie Yamamoto
- Department of Internal Medicine, Fukui General Hospital, Japan
| | - Naoki Takahashi
- Department of Nephrology, Faculty of Medical Sciences, University of Fukui, Japan
| | | | - Tomohiro Ojima
- Department of Rheumatology, Fukui General Hospital, Japan
| | | | - Hideki Kimura
- Department of Nephrology, Faculty of Medical Sciences, University of Fukui, Japan
| | - Masayuki Iwano
- Department of Nephrology, Faculty of Medical Sciences, University of Fukui, Japan
| |
Collapse
|
29
|
Zhao Q, Yan T, Chopp M, Venkat P, Chen J. Brain-kidney interaction: Renal dysfunction following ischemic stroke. J Cereb Blood Flow Metab 2020; 40:246-262. [PMID: 31766979 PMCID: PMC7370616 DOI: 10.1177/0271678x19890931] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Stroke is a leading cause of mortality and morbidity, with long-term debilitating effects. Accumulating evidence from experimental studies as well as observational studies in patients suggests a cross talk between the brain and kidney after stroke. Stroke may lead to kidney dysfunction which can adversely impact patient outcome. In this review article, we discuss the epidemiology and mechanisms of brain–kidney interaction following ischemic stroke. Specifically, we discuss the role of the central autonomic network, autoregulation, inflammatory and immune responses, the role of extracellular vesicles and their cargo microRNA, in mediating brain–kidney interaction following stroke. Understanding the bidirectional nature of interaction between the brain and kidney after cerebral injury would have clinical implications for the treatment of stroke and overall patient outcome.
Collapse
Affiliation(s)
- Qiang Zhao
- Tianjin Neurological Institute, Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Tao Yan
- Tianjin Neurological Institute, Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA.,Department of Physics, Oakland University, Rochester, MI, USA
| | - Poornima Venkat
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| | - Jieli Chen
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| |
Collapse
|
30
|
Sadeghi Z, Fasihi-Ramandi M, Azizi M, Bouzari S. Mannosylated chitosan nanoparticles loaded with FliC antigen as a novel vaccine candidate against Brucella melitensis and Brucella abortus infection. J Biotechnol 2020; 310:89-96. [PMID: 32017955 DOI: 10.1016/j.jbiotec.2020.01.016] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 01/12/2020] [Accepted: 01/30/2020] [Indexed: 12/27/2022]
Abstract
Brucellosis is a worldwide bacterial zoonosis disease. Live attenuated Brucella vaccines have several drawbacks. Thus development of a safe and effective vaccine for brucellosis is a concern of many scientists. FliC protein contributes in virulence of Brucella; hence, it is a promising target for brucellosis vaccine. In this study, Mannosylated Chitosan Nanoparticles (MCN) loaded with FliC protein were synthesized as a targeted vaccine delivery system. The immunogenicity and protective efficacy of FliC and FliC-MCN against Brucella infection were evaluated in BALB/c mice. After cloning, expression and purification, FliC protein was loaded on MCN. The particle size, loading efficiency and in vitro release of the NPs were determined. Our investigation revealed that FliC and FliC-MCN could significantly increase specific IgG response (higher IgG2a titers). Besides, spleen cells from immunized mice produced high level of IFN-γ and IL-2 and low level IL-10 cytokines. Immunization with FliC and FliC-MCN conferred significant degree of protection against B. melitensis 16 M and B. abortus 544 infections. Overall these results indicate that FliC protein would be a novel potential antigen candidate for the development of a subunit vaccine against B. melitensis and B. abortus. Moreover, MCN could be used as an adjuvant and targeted vaccine delivery system.
Collapse
Affiliation(s)
- Zohre Sadeghi
- Department of Molecular Biology, Pasteur Institute of Iran, Tehran, Iran
| | - Mahdi Fasihi-Ramandi
- Molecular Biology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mohammad Azizi
- Department of Medical Biotechnology, Pasteur Institute of Iran, Tehran, Iran
| | - Saeid Bouzari
- Department of Molecular Biology, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
31
|
Associations of Breast Milk Microbiota, Immune Factors, and Fatty Acids in the Rat Mother-Offspring Pair. Nutrients 2020; 12:nu12020319. [PMID: 31991792 PMCID: PMC7071194 DOI: 10.3390/nu12020319] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Revised: 01/17/2020] [Accepted: 01/21/2020] [Indexed: 12/21/2022] Open
Abstract
The present study aimed to analyze the rat breast milk profile of fatty acids (FA), immunoglobulins (Ig), microbiota, and their relationship, and to further assess their associations in the mother–offspring pair. Dams were monitored during the three weeks of gestation, allowed to deliver at term, and followed during two weeks of lactation. At the end of the study, milk was obtained from the dams for the analysis of fatty acids, microbiota composition, immunoglobulins, and cytokines. Moreover, the cecal content and plasma were obtained from both the dams and pups to study the cecal microbiota composition and the plasmatic levels of fatty acids, immunoglobulins, and cytokines. Rat breast milk lipid composition was ~65% saturated FA, ~15% monounsaturated FA, and ~20% polyunsaturated FA. Moreover, the proportions of IgM, IgG, and IgA were ~2%, ~88%, and ~10%, respectively. Breast milk was dominated by members of Proteobacteria, Firmicutes, and Bacteroidetes phyla. In addition, forty genera were shared between the milk and cecal content of dams and pups. The correlations performed between variables showed, for example, that all IgGs subtypes correlated between the three compartments, evidencing their association in the mother-milk-pup line. We established the profile of FA, Ig, and the microbiota composition of rat breast milk. Several correlations in these variables evidenced their association through the mother-milk-pup line. Therefore, it would be interesting to perform dietary interventions during pregnancy and/or lactation that influence the quality of breast milk and have an impact on the offspring.
Collapse
|
32
|
Kang HJ, Chu KB, Lee SH, Kim MJ, Park H, Jin H, Quan FS. Virus-like Particle Vaccine Containing Toxoplasma gondii Rhoptry Protein 13 Induces Protection against T. gondii ME49 Infection in Mice. THE KOREAN JOURNAL OF PARASITOLOGY 2019; 57:543-547. [PMID: 31715698 PMCID: PMC6851246 DOI: 10.3347/kjp.2019.57.5.543] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Accepted: 09/23/2019] [Indexed: 01/17/2023]
Abstract
Toxoplasma gondii can infect humans worldwide, causing serious diseases in pregnant women and immunocompromised individuals. T. gondii rhoptry protein 13 (ROP13) is known as one of the key proteins involved in host cell invasion. In this study, we generated virus-like particles (VLPs) vaccine expressing T. gondii rhoptry ROP13 and investigated VLPs vaccine efficacy in mice. Mice immunized with ROP13 VLPs vaccine elicited significantly higher levels of T. gondii-specific IgG, IgG1, IgG2a, and IgA antibody responses following boost immunization and challenge infection, whereas antibody inductions were insignificant upon prime immunization. Differing immunization routes resulted in differing antibody induction, as intranasal immunization (IN) induced greater antibody responses than intramuscular immunization (IM) after boost and challenge infection. IN immunization induced significantly higher levels of IgG and IgA antibody responses from feces, antibody-secreting cells (ASCs), CD4+ T, CD8+ T cells and germinal center B cell responses in the spleen compared to IM immunization. Compared to IM immunization, IN immunization resulted in significantly reduced cyst counts in the brain as well as lesser body weight loss, which contributed to better protection. All of the mice immunized through either route survived, whereas all naïve control mice perished. These results indicate that the ROP13 VLPs vaccine could be a potential vaccine candidate against T. gondii infection.
Collapse
Affiliation(s)
- Hae-Ji Kang
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Korea
| | - Ki-Back Chu
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Korea
| | - Su-Hwa Lee
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Korea
| | - Min-Ju Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Korea
| | | | - Hui Jin
- Health Park Co., Ltd., Seoul 06627, Korea
| | - Fu-Shi Quan
- Department of Medical Zoology, Kyung Hee University School of Medicine, Seoul 02447, Korea.,Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, School of Medicine, Graduate school, Kyung Hee University, Seoul 02447, Korea
| |
Collapse
|
33
|
Huang CH, Mendez N, Echeagaray OH, Weeks J, Wang J, Vallez CN, Gude N, Trogler WC, Carson DA, Hayashi T, Kummel AC. Conjugation of a Small-Molecule TLR7 Agonist to Silica Nanoshells Enhances Adjuvant Activity. ACS APPLIED MATERIALS & INTERFACES 2019; 11:26637-26647. [PMID: 31276378 DOI: 10.1021/acsami.9b08295] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Stimulation of Toll-like receptors (TLRs) and/or NOD-like receptors on immune cells initiates and directs immune responses that are essential for vaccine adjuvants. The small-molecule TLR7 agonist, imiquimod, has been approved by the FDA as an immune response modifier but is limited to topical application due to its poor pharmacokinetics that causes undesired adverse effects. Nanoparticles are increasingly used with innate immune stimulators to mitigate side effects and enhance adjuvant efficacy. In this study, a potent small-molecule TLR7 agonist, 2-methoxyethoxy-8-oxo-9-(4-carboxybenzyl)adenine (1V209), was conjugated to hollow silica nanoshells (NS). Proinflammatory cytokine (IL-6, IL-12) release by mouse bone-marrow-derived dendritic cells and human peripheral blood mononuclear cells revealed that the potency of silica nanoshells-TLR7 conjugates (NS-TLR) depends on nanoshell size and ligand coating density. Silica nanoshells of 100 nm diameter coated with a minimum of ∼6000 1V209 ligands/particle displayed 3-fold higher potency with no observed cytotoxicity when compared to an unconjugated TLR7 agonist. NS-TLR activated the TLR7-signaling pathway, triggered caspase activity, and stimulated IL-1β release, while neither unconjugated TLR7 ligands nor silica shells alone produced IL-1β. An in vivo murine immunization study, using the model antigen ovalbumin, demonstrated that NS-TLR increased antigen-specific IgG antibody induction by 1000× with a Th1-biased immune response, compared to unconjugated TLR7 agonists. The results show that the TLR7 ligand conjugated to silica nanoshells is capable of activating an inflammasome pathway to enhance both innate immune-stimulatory and adjuvant potencies of the TLR7 agonist, thereby broadening applications of innate immune stimulators.
Collapse
Affiliation(s)
- Ching-Hsin Huang
- Department of Chemistry & Department of Medicine , University of California, San Diego , 9500 Gilman Drive , La Jolla , California 92093-0358 , United States
| | - Natalie Mendez
- Department of Chemistry & Department of Medicine , University of California, San Diego , 9500 Gilman Drive , La Jolla , California 92093-0358 , United States
| | - Oscar Hernandez Echeagaray
- Molecular Biology Institute , San Diego State University , 5500 Campanile Drive , San Diego , California 92182 , United States
| | - Joi Weeks
- Molecular Biology Institute , San Diego State University , 5500 Campanile Drive , San Diego , California 92182 , United States
| | - James Wang
- Department of Chemistry & Department of Medicine , University of California, San Diego , 9500 Gilman Drive , La Jolla , California 92093-0358 , United States
| | - Charles N Vallez
- Molecular Biology Institute , San Diego State University , 5500 Campanile Drive , San Diego , California 92182 , United States
| | - Natalie Gude
- Molecular Biology Institute , San Diego State University , 5500 Campanile Drive , San Diego , California 92182 , United States
| | - William C Trogler
- Department of Chemistry & Department of Medicine , University of California, San Diego , 9500 Gilman Drive , La Jolla , California 92093-0358 , United States
| | - Dennis A Carson
- Moores Cancer Center , University of California , 9500 Gilman Drive , La Jolla , California 92093-0695 , United States
| | - Tomoko Hayashi
- Moores Cancer Center , University of California , 9500 Gilman Drive , La Jolla , California 92093-0695 , United States
| | - Andrew C Kummel
- Department of Chemistry & Department of Medicine , University of California, San Diego , 9500 Gilman Drive , La Jolla , California 92093-0358 , United States
| |
Collapse
|
34
|
Qin AB, Su T, Wang SX, Zhang F, Zhou FD, Zhao MH. Mercury-associated glomerulonephritis: a retrospective study of 35 cases in a single Chinese center. BMC Nephrol 2019; 20:228. [PMID: 31221121 PMCID: PMC6587289 DOI: 10.1186/s12882-019-1413-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 06/06/2019] [Indexed: 01/23/2023] Open
Abstract
Background Long-term exposure of mercury may induce glomerulonephritis. Clinical and pathological features of mercury-associated glomerulonephritis are not fully clear. This study retrospectively analyzed 35 cases of mercury-associated glomerulonephritis in a single Chinese center. Methods Thirty-five patients of mercury-associated glomerulonephritis were enrolled. Clinical data on diagnosis and during follow-up were collected. Plasma anti-phospholipase A2 receptor (PLA2R) antibody, glomerular PLA2R and glomerular IgG subclasses deposition were detected in the cases with membranous nephropathy (MN). Results Mercury exposure was caused by skin lighting cream (20 patients), mercury-containing pills (9 patients), hair-dyeing agents (4 patients), and unidentified reasons (2 patients). All patients presented with proteinuria and normal renal function. The median of urinary protein was 4.6 (range 1.6~19.7) g/24 h. Twenty-two patients (62.9%) had nephrotic syndrome. Renal histopathology showed minimal change disease (MCD) in 21 patients (60.0%), MN in 13 (37.1%) and focal segmental glomerular sclerosis (FSGS) in 1 patient (2.9%). The proportion of MCD increased along with urinary mercury concentration (P = 0.024). In 13 cases of MN, all patients were negative for plasma anti-PLA2R antibody and glomerular PLA2R antigen. IgG1 (61.5%) and IgG4 (46.2%) deposits were noted along the glomerular capillary loops. Among the 16 patients received mercury detoxification monotherapy, 14 patients received 4.5 ± 2.8 (range 1~12) rounds of regimen and achieved complete remission in 4.5 (range 0.3~23.0) months, 2 patients stayed no remission. Conclusions MCD was the most common pathological type of mercury-associated glomerulonephritis, followed by MN. The proportion of MCD increased along with the increase of urinary mercury concentration. Most patients could achieve complete remission after mercury detoxification.
Collapse
Affiliation(s)
- Ai-Bo Qin
- Renal Division, Department of Medicine, Peking University First Hospital, Institute of Nephrology, Peking University, Key Laboratory of Renal Disease, Ministry of Health of China, Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, 100034, China
| | - Tao Su
- Renal Division, Department of Medicine, Peking University First Hospital, Institute of Nephrology, Peking University, Key Laboratory of Renal Disease, Ministry of Health of China, Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, 100034, China
| | - Su-Xia Wang
- Electron Microscopy Laboratory, Peking University First Hospital, Beijing, China
| | - Fan Zhang
- Renal Division, Department of Medicine, Peking University First Hospital, Institute of Nephrology, Peking University, Key Laboratory of Renal Disease, Ministry of Health of China, Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, 100034, China
| | - Fu-de Zhou
- Renal Division, Department of Medicine, Peking University First Hospital, Institute of Nephrology, Peking University, Key Laboratory of Renal Disease, Ministry of Health of China, Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, 100034, China.
| | - Ming-Hui Zhao
- Renal Division, Department of Medicine, Peking University First Hospital, Institute of Nephrology, Peking University, Key Laboratory of Renal Disease, Ministry of Health of China, Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, 100034, China.,Peking-Tsinghua Center for Life Sciences, Beijing, China
| |
Collapse
|
35
|
Bai J, Wu L, Chen X, Wang L, Li Q, Zhang Y, Wu J, Cai G, Chen X. Suppressor of Cytokine Signaling-1/STAT1 Regulates Renal Inflammation in Mesangial Proliferative Glomerulonephritis Models. Front Immunol 2018; 9:1982. [PMID: 30214448 PMCID: PMC6125399 DOI: 10.3389/fimmu.2018.01982] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 08/13/2018] [Indexed: 12/18/2022] Open
Abstract
Mesangial proliferative glomerulonephritis (MsGN) is a significant global threat to public health. Inflammation plays a crucial role in MsGN; however, the underlying mechanism remains unknown. Herein, we demonstrate that suppression of the cytokine signaling-1 (SOCS1)/signal transducer and activator of transcription 1 (STAT1) signaling pathway is associated with renal inflammation and renal injury in MsGN. Using MsGN rat (Thy1.1 GN) and mouse (Habu GN) models, renal SOCS1/STAT1 was determined to be associated with CD4+ T cell infiltration and related cytokines. In vitro, SOCS1 overexpression repressed IFN-γ-induced MHC class II and cytokine levels and STAT1 phosphorylation in mesangial cells. SOCS1 and STAT1 inhibitors significantly inhibited IFN-γ-induced CIITA promoter activity and MHC class II expression. In conclusion, our study emphasizes the pivotal role of the SOCS1/STAT1 axis in the regulation of inflammation in MsGN.
Collapse
Affiliation(s)
- Jiuxu Bai
- State Key Laboratory of Kidney Diseases, Department of Nephrology, National Clinical Research Center for Kidney Diseases, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, Beijing, China
| | - Lingling Wu
- State Key Laboratory of Kidney Diseases, Department of Nephrology, National Clinical Research Center for Kidney Diseases, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, Beijing, China
| | - Xiaoniao Chen
- Department of Ophthalmology, Ophthalmology and Visual Science Key Lab of PLA, Chinese PLA General Hospital, Beijing, China
| | - Liqiang Wang
- Department of Ophthalmology, Ophthalmology and Visual Science Key Lab of PLA, Chinese PLA General Hospital, Beijing, China
| | - Qinggang Li
- State Key Laboratory of Kidney Diseases, Department of Nephrology, National Clinical Research Center for Kidney Diseases, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, Beijing, China
| | - Yingjie Zhang
- State Key Laboratory of Kidney Diseases, Department of Nephrology, National Clinical Research Center for Kidney Diseases, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, Beijing, China
| | - Jie Wu
- State Key Laboratory of Kidney Diseases, Department of Nephrology, National Clinical Research Center for Kidney Diseases, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, Beijing, China
| | - Guangyan Cai
- State Key Laboratory of Kidney Diseases, Department of Nephrology, National Clinical Research Center for Kidney Diseases, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, Beijing, China
| | - Xiangmei Chen
- State Key Laboratory of Kidney Diseases, Department of Nephrology, National Clinical Research Center for Kidney Diseases, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, Beijing, China
| |
Collapse
|
36
|
Yan S, Gu W, Zhang B, Rolfe BE, Xu ZP. High adjuvant activity of layered double hydroxide nanoparticles and nanosheets in anti-tumour vaccine formulations. Dalton Trans 2018; 47:2956-2964. [PMID: 29168855 DOI: 10.1039/c7dt03725b] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Effective adjuvants in anti-tumour vaccine formulations are very important in the development of new-generation vaccines. In this study, two layered double hydroxide (LDH) nanomaterial forms, i.e. nanoparticles (NPs) and nanosheets (NSs), were synthesised and examined as adjuvants to provoke the immune responses for anti-tumour purpose. Immunogen ovalbumin (OVA) delivered by both nanomaterials induced much stronger humoral and cell-medicated immune responses, together with an immune stimulant (TLR9 ligand CpG), as evidenced by higher levels of IgG1, IgG2a and interferon-γ. By comparison, LDH NSs showed higher activity to promote specific antibody responses than LDH NPs but with a similar cell-mediated immune response. The mice immunised with OVA-CpG vaccines formulated with both nanomaterials showed stronger inhibition of the inoculated tumour growth and had a longer survival. Altogether, these data indicate that LDH NPs and NSs can be used as potential nanoadjuvants for efficient protein-based anti-tumour vaccines.
Collapse
Affiliation(s)
- Shiyu Yan
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia.
| | | | | | | | | |
Collapse
|
37
|
Kamezaki M, Kusaba T, Adachi T, Yamashita N, Nakata M, Ota N, Shiotsu Y, Ishida M, Usui T, Tamagaki K. Unusual Proliferative Glomerulonephritis in a Patient Diagnosed to Have Hypoparathyroidism, Sensorineural Deafness, and Renal Dysplasia (HDR) Syndrome with a Novel Mutation in the GATA3 Gene. Intern Med 2017; 56:1393-1397. [PMID: 28566604 PMCID: PMC5498205 DOI: 10.2169/internalmedicine.56.7930] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 10/03/2016] [Indexed: 11/06/2022] Open
Abstract
Hypoparathyroidism, sensorineural deafness, and renal dysplasia (HDR) syndrome is a rare autosomal dominant disease caused by GATA3 mutations. Although several cases with variable renal features have been reported, the presence of histological changes within the glomeruli in adult patients is unclear. We herein report an adult case of HDR syndrome with a novel p.C288W (TGC>TGG) missense mutation in GATA3. His renal histology showed a membranoproliferative glomerulonephritis-like glomerular lesion. Additional renal histological analyses of HDR syndrome patients will be needed to clarify the role of GATA3 in both the developing and adult kidney.
Collapse
Affiliation(s)
- Michitsugu Kamezaki
- Division of Nephrology, Department of Medicine, Kyoto Prefectural University of Medicine, Japan
| | - Tetsuro Kusaba
- Division of Nephrology, Department of Medicine, Kyoto Prefectural University of Medicine, Japan
| | - Takaomi Adachi
- Division of Nephrology, Department of Medicine, Kyoto Prefectural University of Medicine, Japan
| | - Noriyuki Yamashita
- Division of Nephrology, Department of Medicine, Kyoto Prefectural University of Medicine, Japan
| | - Mayumi Nakata
- Division of Nephrology, Department of Medicine, Kyoto Prefectural University of Medicine, Japan
| | - Noriyoshi Ota
- Division of Nephrology, Department of Medicine, Kyoto Prefectural University of Medicine, Japan
| | - Yayoi Shiotsu
- Division of Nephrology, Department of Medicine, Kyoto Prefectural University of Medicine, Japan
| | - Mami Ishida
- Division of Nephrology, Department of Medicine, Kyoto Prefectural University of Medicine, Japan
| | - Takeshi Usui
- Department of Medical Genetics, Shizuoka General Hospital, Japan
| | - Keiichi Tamagaki
- Division of Nephrology, Department of Medicine, Kyoto Prefectural University of Medicine, Japan
| |
Collapse
|
38
|
Luque Y, Cathelin D, Vandermeersch S, Xu X, Sohier J, Placier S, Xu-Dubois YC, Louis K, Hertig A, Bories JC, Vasseur F, Campagne F, Di Santo JP, Vosshenrich C, Rondeau E, Mesnard L. Glomerular common gamma chain confers B- and T-cell–independent protection against glomerulonephritis. Kidney Int 2017; 91:1146-1158. [DOI: 10.1016/j.kint.2016.10.037] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 10/19/2016] [Accepted: 10/27/2016] [Indexed: 12/22/2022]
|
39
|
Ge YC, Jin B, Zeng CH, Zhang MC, Chen DC, Yin R, Le WB. PLA2R antibodies and PLA2R glomerular deposits in psoriasis patients with membranous nephropathy. BMC Nephrol 2016; 17:185. [PMID: 27876003 PMCID: PMC5118880 DOI: 10.1186/s12882-016-0407-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 11/15/2016] [Indexed: 11/26/2022] Open
Abstract
Background The association between psoriasis and membranous nephropathy (MN) remains largely unclear. We examined the prevalence of serum PLA2R antibody and characterized the expression of PLA2R and THSD7A in glomeruli in patients with MN and psoriasis. Methods A total of 24 patients with MN without evidence of a secondary cause except psoriasis were enrolled. The clinical and pathological features were retrospectively analyzed. Serum anti-PLA2R antibody was measured using IFA Mosaic. Renal tissue samples stored in the laboratory bio-bank were used for PLA2R staining under immunofluorescence microscopy and THSD7A immunohistochemical analysis. Results Twenty-four patients (21 male and 3 female) with a mean age of 43.6 ± 15.7 years old were enrolled. Serum anti-PLA2R antibody was positive in 7 patients, which was significantly lower than the positivity observed in idiopathic MN (29.2% vs. 81.7%, P < 0.001). Glomerular PLA2R staining was positive in 7 patients with positive serum anti-PLA2R antibody. THSD7A staining was negative in all 24 patients. During the follow-up visits, 13 patients with negative serum PLA2R antibody achieved CR. In contrast, CR was only achieved in 1 patient with positive serum PLA2R antibody, PR was achieved in 2 patients. Conclusions The prevalence of serum anti-PLA2R antibody and glomerular expression of PLA2R was significantly lower in patients with psoriasis and MN than in those with idiopathic MN, and THSD7A staining was negative, suggesting that MN is associated with psoriasis in the majority of patients. However, idiopathic MN might also accompany psoriasis in a minority of psoriatic patients with positive serum anti-PLA2R antibody.
Collapse
Affiliation(s)
- Yong-Chun Ge
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, 210016, People's Republic of China.
| | - Bo Jin
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, 210016, People's Republic of China
| | - Cai-Hong Zeng
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, 210016, People's Republic of China
| | - Ming-Chao Zhang
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, 210016, People's Republic of China
| | - Da-Cheng Chen
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, 210016, People's Republic of China
| | - Ru Yin
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, 210016, People's Republic of China
| | - Wei-Bo Le
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, 210016, People's Republic of China
| |
Collapse
|
40
|
Abstract
Glomerular diseases are common and important. They can arise from systemic inflammatory or metabolic diseases that affect the kidney. Alternately, they are caused primarily by local glomerular abnormalities, including genetic diseases. Both intrinsic glomerular cells and leukocytes are critical to the healthy glomerulus and to glomerular dysregulation in disease. Mesangial cells, endothelial cells, podocytes, and parietal epithelial cells within the glomerulus all play unique and specialized roles. Although a specific disease often primarily affects a particular cell type, the close proximity, and interdependent functions and interactions between cells mean that even diseases affecting one cell type usually indirectly influence others. In addition to those cells intrinsic to the glomerulus, leukocytes patrol the glomerulus in health and mediate injury in disease. Distinct leukocyte types and subsets are present, with some being involved in different ways in an individual glomerular disease. Cells of the innate and adaptive immune systems are important, directing systemic immune and inflammatory responses, locally mediating injury, and potentially dampening inflammation and facilitating repair. The advent of new genetic and molecular techniques, and new disease models means that we better understand both the basic biology of the glomerulus and the pathogenesis of glomerular disease. This understanding should lead to better diagnostic techniques, biomarkers, and predictors of prognosis, disease severity, and relapse. With this knowledge comes the promise of better therapies in the future, directed toward halting pathways of injury and fibrosis, or interrupting the underlying pathophysiology of the individual diseases that lead to significant and progressive glomerular disease.
Collapse
Affiliation(s)
- A. Richard Kitching
- Centre for Inflammatory Diseases, Department of Medicine, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia
- Department of Nephrology, and
- Department of Pediatric Nephrology, Monash Medical Centre, Clayton, Victoria, Australia
| | - Holly L. Hutton
- Centre for Inflammatory Diseases, Department of Medicine, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia
- Department of Nephrology, and
| |
Collapse
|
41
|
Release from Th1-type immune tolerance in spleen and enhanced production of IL-5 in Peyer’s patch by cholera toxin B induce the glomerular deposition of IgA. Immunobiology 2016; 221:577-85. [DOI: 10.1016/j.imbio.2015.12.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 12/01/2015] [Accepted: 12/01/2015] [Indexed: 12/28/2022]
|
42
|
Abstract
Biological therapeutics (biologics) that target autoimmune responses and inflammatory injury pathways have a marked beneficial impact on the management of many chronic diseases, including rheumatoid arthritis, psoriasis, inflammatory bowel disease, and ankylosing spondylitis. Accumulating data suggest that a growing number of renal diseases result from autoimmune injury - including lupus nephritis, IgA nephropathy, anti-neutrophil cytoplasmic antibody-associated glomerulonephritis, autoimmune (formerly idiopathic) membranous nephropathy, anti-glomerular basement membrane glomerulonephritis, and C3 nephropathy - and one can speculate that biologics might also be applicable to these diseases. As many autoimmune renal diseases are relatively uncommon, with long natural histories and diverse outcomes, clinical trials that aim to validate potentially useful biologics are difficult to design and/or perform. Some excellent consortia are undertaking cohort studies and clinical trials, but more multicentre international collaborations are needed to advance the introduction of new biologics to patients with autoimmune renal disorders. This Review discusses the key molecules that direct injurious inflammation and the biologics that are available to modulate them. The opportunities and challenges for the introduction of relevant biologics into treatment protocols for autoimmune renal diseases are also discussed.
Collapse
|
43
|
Sui D, Ma L, Li M, Shao W, Du H, Li K, Li Z, Li Z. Mucin 1 and poly I:C activates dendritic cells and effectively eradicates pituitary tumors as a prophylactic vaccine. Mol Med Rep 2016; 13:3675-83. [PMID: 26935338 DOI: 10.3892/mmr.2016.4964] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 12/02/2015] [Indexed: 11/05/2022] Open
Abstract
Pituitary tumors are the most common type of cancer within the central nervous system. In the present study, the expression levels of mucin 1 (Muc1) were examined in invasive and non‑invasive pituitary tumor samples, and the results of immunohistochemical staining and Western blot analysis demonstrated marked positive expression of Muc1. In addition, Muc1 + polyinosinic:polycytidylic acid (poly I:C) was found to stimulate the expression levels of the surface molecules cluster of differentiation (CD)40, CD83 and CD80, and HLA‑DRm and decreased the expression of CD14 in the dendritic cells, determined using fluorescence‑activated cell sorting. The secretions of interleukin (IL)‑6, tumor necrosis factor‑α and IL‑1β cytokines were also significantly induced, in a dose‑dependent manner. In in vivo experiments, a higher percentage of CD3+CD4+ T lymphocytes was detected, and the levels of interferon‑γ and IL‑2 in the splenocytes were also upregulated. Furthermore, the combination treatment of Muc1 with poly I:C increased anti‑Muc1 IgM and anti‑Muc1 IgG titers, and altered the balance of IgG2a and IgG1, all of which increased the T helper (Th)1 polarized immune response. Thus, the tumor antigen, Muc1, with poly I:C may produce potent protective effects, which polarize immune responses towards Th1, and elicit antitumor immunity to inhibit the progression of pituitary tumors.
Collapse
Affiliation(s)
- Dehua Sui
- Department of Neurosurgery, Affiliated Hospital of Binzhou Medical College, Binzhou, Shandong 256603, P.R. China
| | - Lixin Ma
- Department of Neurosurgery, Affiliated Hospital of Binzhou Medical College, Binzhou, Shandong 256603, P.R. China
| | - Meng Li
- Department of Neurosurgery, Affiliated Hospital of Binzhou Medical College, Binzhou, Shandong 256603, P.R. China
| | - Wei Shao
- Department of Neurosurgery, Affiliated Hospital of Binzhou Medical College, Binzhou, Shandong 256603, P.R. China
| | - Hongpeng Du
- Department of Neurosurgery, Affiliated Hospital of Binzhou Medical College, Binzhou, Shandong 256603, P.R. China
| | - Ke Li
- Department of Neurosurgery, Affiliated Hospital of Binzhou Medical College, Binzhou, Shandong 256603, P.R. China
| | - Zhenzhu Li
- Department of Neurosurgery, Affiliated Hospital of Binzhou Medical College, Binzhou, Shandong 256603, P.R. China
| | - Zefu Li
- Department of Neurosurgery, Affiliated Hospital of Binzhou Medical College, Binzhou, Shandong 256603, P.R. China
| |
Collapse
|
44
|
Changrob S, Wang B, Han JH, Lee SK, Nyunt MH, Lim CS, Tsuboi T, Chootong P, Han ET. Naturally-Acquired Immune Response against Plasmodium vivax Rhoptry-Associated Membrane Antigen. PLoS One 2016; 11:e0148723. [PMID: 26886867 PMCID: PMC4757550 DOI: 10.1371/journal.pone.0148723] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 01/22/2016] [Indexed: 11/18/2022] Open
Abstract
Rhoptry-associated membrane antigen (RAMA) is an abundant glycophosphatidylinositol (GPI)-anchored protein that is embedded within the lipid bilayer and is implicated in parasite invasion. Antibody responses against rhoptry proteins are produced by individuals living in a malaria-endemic area, suggesting the immunogenicity of Plasmodium vivax RAMA (PvRAMA) for induction of immune responses during P. vivax infection. To determine whether PvRAMA contributes to the acquisition of immunity to malaria and could be a rational candidate for a vaccine, the presence of memory T cells and the stability of the antibody response against PvRAMA were evaluated in P. vivax-exposed individuals. The immunogenicity of PvRAMA for the induction of T cell responses was evaluated by in vitro stimulation of peripheral blood mononuclear cells (PBMCs). High levels of interferon (IFN)-γ and interleukin (IL)-10 cytokines were detected in the culture supernatant of PBMCs, and the CD4+ T cells predominantly produced IL-10 cytokine. The levels of total anti-PvRAMA immunoglobulin G (IgG) antibody were significantly elevated, and these antibodies persisted over the 12 months of the study. Interestingly, IgG1, IgG2 and IgG3 were the major antibody subtypes in the response to PvRAMA. The frequency of IgG3 in specific to PvRAMA antigen maintained over 12 months. These data could explain the immunogenicity of PvRAMA antigen in induction of both cell-mediated and antibody-mediated immunity in natural P. vivax infection, in which IFN-γ helps antibody class switching toward the IgG1, IgG2 and IgG3 isotypes and IL-10 supports PvRAMA-specific antibody production.
Collapse
Affiliation(s)
- Siriruk Changrob
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand
| | - Bo Wang
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, Republic of Korea
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People’s Republic of China
| | - Jin-Hee Han
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, Republic of Korea
| | - Seong-Kyun Lee
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, Republic of Korea
| | - Myat Htut Nyunt
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, Republic of Korea
- Department of Medical Research, Yangon, Myanmar
| | - Chae Seung Lim
- Department of Laboratory Medicine, College of Medicine, Korea University Guro Hospital, Guro Gu, Seoul, Republic of Korea
| | - Takafumi Tsuboi
- Division of Malaria Research Proteo-Science Center, Ehime University, Matsuyama, Ehime, Japan
| | - Patchanee Chootong
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand
- * E-mail: (PC); (E-TH)
| | - Eun-Taek Han
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People’s Republic of China
- * E-mail: (PC); (E-TH)
| |
Collapse
|
45
|
Aberrantly methylated DNA regions lead to low activation of CD4+ T-cells in IgA nephropathy. Clin Sci (Lond) 2016; 130:733-46. [PMID: 26846681 DOI: 10.1042/cs20150711] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 02/04/2016] [Indexed: 12/12/2022]
Abstract
IgAN (IgA nephropathy) is the most common form of primary glomerulonephritis worldwide and has a strong genetic component. In this setting, DNA methylation could also be an important factor influencing this disease. We performed a genome-wide screening for DNA methylation in CD4(+) T-cells from IgAN patients and found three regions aberrantly methylated influencing genes involved in the response and proliferation of CD4(+) T-cells. Two hypomethylated regions codified genes involved in TCR (T-cell receptor) signalling, TRIM27 (tripartite motif-containing 27) and DUSP3 (dual-specificity phosphatase 3), and an hypermethylated region included the VTRNA2-1 (vault RNA 2-1) non-coding RNA, also known as miR-886 precursor. We showed that the aberrant methylation influences the expression of these genes in IgAN patients. Moreover, we demonstrated that the hypermethylation of the miR-886 precursor led to a decreased CD4(+) T-cell proliferation following TCR stimulation and to the overexpression of TGFβ (transforming growth factor β). Finally, we found a Th1/Th2 imbalance in IgAN patients. The IL (interleukin)-2/IL-5 ratio was notably higher in IgAN patients and clearly indicated a Th1 shift. In conclusion, we identified for the first time some specific DNA regions abnormally methylated in IgAN patients that led to the reduced TCR signal strength of the CD4(+) T-cells and to their anomalous response and activation that could explain the T-helper cell imbalance. The present study reveals new molecular mechanisms underlying the abnormal CD4(+) T-cell response in IgAN patients.
Collapse
|
46
|
Barbour TD, Ling GS, Ruseva MM, Fossati-Jimack L, Cook HT, Botto M, Pickering MC. Complement receptor 3 mediates renal protection in experimental C3 glomerulopathy. Kidney Int 2016; 89:823-32. [PMID: 26924054 PMCID: PMC4869622 DOI: 10.1016/j.kint.2015.11.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 11/16/2015] [Accepted: 11/19/2015] [Indexed: 12/14/2022]
Abstract
C3 glomerulopathy is a complement-mediated renal disease that is frequently associated with abnormalities in regulation of the complement alternative pathway. Mice with deficiency of factor H (Cfh–/–), a negative alternative pathway regulator, are an established experimental model of C3 glomerulopathy in which complement C3 fragments including iC3b accumulate along the glomerular basement membrane. Here we show that deficiency of complement receptor 3 (CR3), the main receptor for iC3b, enhances the severity of spontaneous renal disease in Cfh–/– mice. This effect was found to be dependent on CR3 expression on bone marrow–derived cells. CR3 also mediated renal protection outside the setting of factor H deficiency, as shown by the development of enhanced renal injury in CR3-deficient mice during accelerated nephrotoxic nephritis. The iC3b–CR3 interaction downregulated the proinflammatory cytokine response of both murine and human macrophages to lipopolysaccharide stimulation in vitro, suggesting that the protective effect of CR3 on glomerular injury was mediated via modulation of macrophage-derived proinflammatory cytokines. Thus, CR3 has a protective role in glomerulonephritis and suggests that pharmacologic potentiation of the macrophage CR3 interaction with iC3b could be therapeutically beneficial.
Collapse
Affiliation(s)
- Thomas D Barbour
- Centre for Complement and Inflammation Research, Imperial College, London, UK
| | - Guang Sheng Ling
- Centre for Complement and Inflammation Research, Imperial College, London, UK
| | - Marieta M Ruseva
- Centre for Complement and Inflammation Research, Imperial College, London, UK
| | - Liliane Fossati-Jimack
- Centre for Complement and Inflammation Research, Imperial College, London, UK; Centre for Experimental Medicine and Rheumatology, Queen Mary University of London, London, UK
| | - H Terence Cook
- Centre for Complement and Inflammation Research, Imperial College, London, UK
| | - Marina Botto
- Centre for Complement and Inflammation Research, Imperial College, London, UK
| | - Matthew C Pickering
- Centre for Complement and Inflammation Research, Imperial College, London, UK.
| |
Collapse
|
47
|
Ramani K, Biswas PS. Emerging roles of the Th17/IL-17-axis in glomerulonephritis. Cytokine 2016; 77:238-44. [DOI: 10.1016/j.cyto.2015.07.029] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 07/30/2015] [Accepted: 07/31/2015] [Indexed: 12/25/2022]
|
48
|
Kalavrizioti D, Gerolymos M, Rodi M, Kalliakmani P, Provatopoulou S, Eleftheriadis T, Mouzaki A, Goumenos DS. T helper (Th)-cytokines in the urine of patients with primary glomerulonephritis treated with immunosuppressive drugs: Can they predict outcome? Cytokine 2015; 76:260-269. [PMID: 26307557 DOI: 10.1016/j.cyto.2015.08.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 07/15/2015] [Accepted: 08/05/2015] [Indexed: 12/29/2022]
Abstract
BACKGROUND Glomerulonephritides (GNs) represent common causes of chronic kidney disease associated with a wide spectrum of clinical and histological features. Various factors that activate the inflammatory cascade are involved in the development of kidney injury. The aim of this study was to estimate the urinary excretion of pro-inflammatory (IL-2, INF-γ, TNF-α, IL-6, IL-17) and anti-inflammatory (IL-4, IL-10, TGF-β1) cytokines, as well as the chemokine MCP-1 in patients with various types of GN treated by immunosuppressive drugs and to identify any prognostic value of excreted cytokines for future renal function. PATIENTS AND METHODS Ninety-seven patients (62 M/35 F, age 53.1 ± 15.6 years) with primary glomerulonephritis and 32 healthy controls were studied. The original diagnoses were membranous nephropathy (MN, n=36), IgA nephropathy (IgAN, n=31) and minimal changes disease or focal segmental glomerulosclerosis (MCD/FSGS, n=30). All patients had been treated with immunosuppressive drugs and, at the time of measurement of urinary cytokine excretion, were either in clinical remission or still had active disease with persistent proteinuria. RESULTS GN patients had significantly higher levels of all cytokines and MCP-1 compared to healthy controls. A strong positive correlation between TGF-β1 and MCP-1 concentrations was observed in all GN patients. Increased urinary excretion of all tested cytokines apart from TNF-α and TGF-β1 was observed even in patients with clinical remission. The main difference between patients with proteinuria and those in clinical remission was the level of MCP-1 urinary excretion. The urinary excretion of MCP-1 and TGF-β1 was significantly higher in patients with MN who showed deterioration of renal function over a follow-up period of five years. CONCLUSIONS Increased levels of cytokines are observed in the urine of patients with different types of glomerulonephritis, even after the achievement of clinical remission with the administration of immunosuppressive drugs. Urinary excretion of MCP-1 and TGF-β1 indicates the ongoing inflammatory and fibrotic processes in the kidney and is probably related to unfavourable outcomes.
Collapse
Affiliation(s)
- Dimitra Kalavrizioti
- Department of Nephrology and Renal Transplantation, University Hospital of Patras, Patras, Greece.
| | - Miltiadis Gerolymos
- Department of Nephrology and Renal Transplantation, University Hospital of Patras, Patras, Greece.
| | - Maria Rodi
- Division of Hematology, Department of Internal Medicine, Medical School, University of Patras, Patras, Greece.
| | - Pantelitsa Kalliakmani
- Department of Nephrology and Renal Transplantation, University Hospital of Patras, Patras, Greece.
| | - Simela Provatopoulou
- Department of Nephrology and Renal Transplantation, University Hospital of Patras, Patras, Greece.
| | | | - Athanasia Mouzaki
- Division of Hematology, Department of Internal Medicine, Medical School, University of Patras, Patras, Greece.
| | - Dimitrios S Goumenos
- Department of Nephrology and Renal Transplantation, University Hospital of Patras, Patras, Greece.
| |
Collapse
|
49
|
Ooi JD, Li M, Kourkoutzelos K, Yagita H, Azuma M, Holdsworth SR, Kitching AR. Programmed death 1 and its ligands do not limit experimental foreign antigen-induced immune complex glomerulonephritis. Nephrology (Carlton) 2015; 20:892-8. [DOI: 10.1111/nep.12532] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/05/2015] [Indexed: 01/22/2023]
Affiliation(s)
- Joshua D Ooi
- Centre for Inflammatory Diseases; Monash University Department of Medicine; Melbourne Victoria Australia
| | - Ming Li
- Centre for Inflammatory Diseases; Monash University Department of Medicine; Melbourne Victoria Australia
| | - Katerina Kourkoutzelos
- Centre for Inflammatory Diseases; Monash University Department of Medicine; Melbourne Victoria Australia
| | - Hideo Yagita
- Department of Immunology; Juntendo University School of Medicine
| | - Miyuki Azuma
- Department of Molecular Immunology; Graduate School of Medical and Dental Sciences; Tokyo Medical and Dental University; Tokyo Japan
| | - Stephen R Holdsworth
- Centre for Inflammatory Diseases; Monash University Department of Medicine; Melbourne Victoria Australia
- Department of Nephrology; Monash Health; Melbourne Victoria Australia
| | - A Richard Kitching
- Centre for Inflammatory Diseases; Monash University Department of Medicine; Melbourne Victoria Australia
- Department of Nephrology; Monash Health; Melbourne Victoria Australia
- Department of Paediatric Nephrology; Monash Health; Melbourne Victoria Australia
| |
Collapse
|
50
|
IgG anti-tTG responses in different autoimmune conditions differ in their epitope targets and subclass usage. Mol Immunol 2015; 67:369-76. [DOI: 10.1016/j.molimm.2015.06.026] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 06/18/2015] [Accepted: 06/24/2015] [Indexed: 02/07/2023]
|