1
|
Restrepo CM, Llanes A, Herrera L, Ellis E, Quintero I, Fernández PL. Baseline gene expression in BALB/c and C57BL/6 peritoneal macrophages influences but does not dictate their functional phenotypes. Exp Biol Med (Maywood) 2025; 249:10377. [PMID: 39830895 PMCID: PMC11740880 DOI: 10.3389/ebm.2024.10377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 12/09/2024] [Indexed: 01/22/2025] Open
Abstract
Macrophages are effector cells of the immune system and essential modulators of immune responses. Different functional phenotypes of macrophages with specific roles in the response to stimuli have been described. The C57BL/6 and BALB/c mouse strains tend to selectively display distinct macrophage activation states in response to pathogens, namely, the M1 and M2 phenotypes, respectively. Herein we used RNA-Seq and differential expression analysis to characterize the baseline gene expression pattern of unstimulated resident peritoneal macrophages from C57BL/6 and BALB/c mice. Our aim is to determine if there is a possible predisposition of these mouse strains to any activation phenotype and how this may affect the interpretation of results in studies concerning their interaction with pathogens. We found differences in basal gene expression patterns of BALB/c and C57BL/6 mice, which were further confirmed using RT-PCR for a subset of relevant genes. Despite these differences, our data suggest that baseline gene expression patterns of both mouse strains do not appear to determine by itself a specific macrophage phenotype.
Collapse
Affiliation(s)
- Carlos M. Restrepo
- Centro de Biología Celular y Molecular de Enfermedades, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Panama City, Panama
- Sistema Nacional de Investigación (SNI), Secretaría Nacional de Ciencia Tecnología e Innovación (SENACYT), Panama City, Panama
| | - Alejandro Llanes
- Centro de Biología Celular y Molecular de Enfermedades, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Panama City, Panama
- Sistema Nacional de Investigación (SNI), Secretaría Nacional de Ciencia Tecnología e Innovación (SENACYT), Panama City, Panama
| | - Lizzi Herrera
- Bioterio, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Panama City, Panama
| | - Esteban Ellis
- Centro de Biología Celular y Molecular de Enfermedades, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Panama City, Panama
- Departamento de Biotecnología, Facultad de Ciencias de la Salud, Universidad Latina de Panamá, Panama City, Panama
- Facultad de Ciencia y Tecnología, Universidad Tecnológica de Panamá, Panama City, Panama
| | - Iliana Quintero
- Centro de Biología Celular y Molecular de Enfermedades, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Panama City, Panama
| | - Patricia L. Fernández
- Centro de Biología Celular y Molecular de Enfermedades, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Panama City, Panama
- Sistema Nacional de Investigación (SNI), Secretaría Nacional de Ciencia Tecnología e Innovación (SENACYT), Panama City, Panama
| |
Collapse
|
2
|
Xue Y, Zhao G, Pu X, Jiao F. Construction of T cell exhaustion model for predicting survival and immunotherapy effect of bladder cancer based on WGCNA. Front Oncol 2023; 13:1196802. [PMID: 37324016 PMCID: PMC10266200 DOI: 10.3389/fonc.2023.1196802] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 05/17/2023] [Indexed: 06/17/2023] Open
Abstract
Introduction The prognosis of bladder cancer (BLCA) and response to immune checkpoint inhibitors (ICIs) are determined by multiple factors. Existed biomarkers for predicting the effect of immunotherapy cannot accurately predict the response of BLCA patients to ICIs. Methods To further accurately stratify patients' response to ICIs and identify potential novel predictive biomarkers, we used the known T cell exhaustion (TEX)-related specific pathways, including tumor necrosis factor (TNF), interleukin (IL)-2, interferon (IFN)-g, and T- cell cytotoxicpathways, combined with weighted correlation network analysis (WGCNA) to analyze the characteristics of TEX in BLCA in detail, constructed a TEX model. Results This model including 28 genes can robustly predict the survival of BLCA and immunotherapeutic efficacy. This model could divide BLCA into two groups, TEXhigh and TEXlow, with significantly different prognoses, clinical features, and reactivity to ICIs. The critical characteristic genes, such as potential biomarkers Charged Multivesicular Body Protein 4C (CHMP4C), SH2 Domain Containing 2A (SH2D2A), Prickle Planar Cell Polarity Protein 3 (PRICKLE3) and Zinc Finger Protein 165 (ZNF165) were verified in BLCA clinical samples by real-time quantitative chain reaction (qPCR) and immunohistochemistry (IHC). Discussion Our findings show that the TEX model can serve as biological markers for predicting the response to ICIs, and the involving molecules in the TEX model might provide new potential targets for immunotherapy in BLCA.
Collapse
Affiliation(s)
- Yuwen Xue
- Department of Pulmonary and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Guanghui Zhao
- Peking University People’s Hospital, Qingdao Women and Children’s Hospital, Qingdao, China
- Medical Laboratory Center, Qilu Hospital (Qingdao) of Shandong University, Qingdao, China
| | - Xiaoxin Pu
- Department of Pulmonary and Critical Care Medicine, Qilu Hospital (Qingdao) of Shandong University, Qingdao, China
| | - Fangdong Jiao
- Department of Urology Surgery, Qilu Hospital (Qingdao) of Shandong University, Qingdao, China
| |
Collapse
|
3
|
Zhu X, Liu B, Ruan Z, Chen M, Li C, Shi H, Huang X, Yu H, Zhou Y, Zhu H, Sun J, Wei Y, Xu W, Dong J. TMT-Based Quantitative Proteomic Analysis Reveals Downregulation of ITGAL and Syk by the Effects of Cycloastragenol in OVA-Induced Asthmatic Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:6842530. [PMID: 36329800 PMCID: PMC9626231 DOI: 10.1155/2022/6842530] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 08/28/2022] [Accepted: 09/29/2022] [Indexed: 12/08/2023]
Abstract
BACKGROUND Cycloastragenol (CAG) has been reported to alleviate airway inflammation in ovalbumin- (OVA-) induced asthmatic mice. However, its specific mechanisms remain unclear. OBJECTIVE This study is aimed at investigating the effects of CAG on asthma, comparing its efficacy with dexamethasone (DEX), and elucidating the mechanism of CAG's regulation. METHODS The asthma mouse model was induced by OVA. CAG at the optimal dose of 125 mg/kg was given every day from day 0 for 20-day prevention or from day 14 for a 7-day treatment. We observed the preventive and therapeutic effects of CAG in asthmatic mice by evaluating the airway inflammation, AHR, and mucus secretion. Lung proteins were used for TMT-based quantitative proteomic analysis to enunciate its regulatory mechanisms. RESULTS The early administration of 125 mg/kg CAG before asthma happened prevented asthmatic mice from AHR, airway inflammation, and mucus hypersecretion, returning to nearly the original baseline. Alternatively, the administration of CAG during asthma also had the same therapeutic effects as DEX. The proteomic analysis revealed that the therapeutical effects of CAG were associated with 248 differentially expressed proteins and 3 enriched KEGG pathways. We then focused on 3 differentially expressed proteins (ITGAL, Syk, and Vav1) and demonstrated that CAG treatment downregulated ITGAL, Syk, and Vav1 by quantitative real-time PCR, western blot analysis, and immunohistochemical staining. CONCLUSION These findings suggest that CAG exerts preventive and protective effects on asthma by inhibiting ITGAL, Syk, and the downstream target Vav1.
Collapse
Affiliation(s)
- Xueyi Zhu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Baojun Liu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Zhenhui Ruan
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Mengmeng Chen
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Congcong Li
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Hanlin Shi
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Xi Huang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Hang Yu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yaolong Zhou
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Hehua Zhu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Jing Sun
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Ying Wei
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Weifang Xu
- Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
| | - Jingcheng Dong
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institutes of Integrative Medicine, Fudan University, Shanghai, China
| |
Collapse
|
4
|
Ogongo P, Porterfield JZ, Leslie A. Lung Tissue Resident Memory T-Cells in the Immune Response to Mycobacterium tuberculosis. Front Immunol 2019; 10:992. [PMID: 31130965 PMCID: PMC6510113 DOI: 10.3389/fimmu.2019.00992] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 04/17/2019] [Indexed: 12/12/2022] Open
Abstract
Despite widespread BCG vaccination and effective anti-TB drugs, Tuberculosis (TB) remains the leading cause of death from an infectious agent worldwide. Several recent publications give reasons to be optimistic about the possibility of a more effective vaccine, but the only full-scale clinical trial conducted failed to show protection above BCG. The immunogenicity of vaccines in humans is primarily evaluated by the systemic immune responses they generate, despite the fact that a correlation between these responses and protection from TB disease has not been demonstrated. A novel approach to tackling this problem is to study the local immune responses that occur at the site of TB infection in the human lung, rather than those detectable in blood. There is a growing understanding that pathogen specific T-cell immunity can be highly localized at the site of infection, due to the existence of tissue resident memory T-cells (Trm). Notably, these cells do not recirculate in the blood and thus may not be represented in studies of the systemic immune response. Here, we review the potential role of Trms as a component of the TB immune response and discuss how a better understanding of this response could be harnessed to improve TB vaccine efficacy.
Collapse
Affiliation(s)
- Paul Ogongo
- Africa Health Research Institute, Durban, South Africa
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
- Institute of Primate Research, National Museums of Kenya, Nairobi, Kenya
| | - James Zachary Porterfield
- Africa Health Research Institute, Durban, South Africa
- College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
- Yale School of Public Health, Yale University, New Haven, CT, United States
| | - Alasdair Leslie
- Africa Health Research Institute, Durban, South Africa
- Department of Infection and Immunity, University College London, London, United Kingdom
| |
Collapse
|
5
|
Fagerholm SC, Guenther C, Llort Asens M, Savinko T, Uotila LM. Beta2-Integrins and Interacting Proteins in Leukocyte Trafficking, Immune Suppression, and Immunodeficiency Disease. Front Immunol 2019; 10:254. [PMID: 30837997 PMCID: PMC6389632 DOI: 10.3389/fimmu.2019.00254] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 01/29/2019] [Indexed: 12/21/2022] Open
Abstract
Beta2-integrins are complex leukocyte-specific adhesion molecules that are essential for leukocyte (e.g., neutrophil, lymphocyte) trafficking, as well as for other immunological processes such as neutrophil phagocytosis and ROS production, and T cell activation. Intriguingly, however, they have also been found to negatively regulate cytokine responses, maturation, and migratory responses in myeloid cells such as macrophages and dendritic cells, revealing new, and unexpected roles of these molecules in immunity. Because of their essential role in leukocyte function, a lack of expression or function of beta2-integrins causes rare immunodeficiency syndromes, Leukocyte adhesion deficiency type I, and type III (LAD-I and LAD-III). LAD-I is caused by reduced or lost expression of beta2-integrins, whilst in LAD-III, beta2-integrins are expressed but dysfunctional because a major integrin cytoplasmic regulator, kindlin-3, is mutated. Interestingly, some LAD-related phenotypes such as periodontitis have recently been shown to be due to an uncontrolled inflammatory response rather than to an uncontrolled infection, as was previously thought. This review will focus on the recent advances concerning the regulation and functions of beta2-integrins in leukocyte trafficking, immune suppression, and immune deficiency disease.
Collapse
Affiliation(s)
- Susanna C Fagerholm
- Molecular and Integrative Biosciences Research Program, Faculty of Bio- and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Carla Guenther
- Molecular and Integrative Biosciences Research Program, Faculty of Bio- and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Marc Llort Asens
- Molecular and Integrative Biosciences Research Program, Faculty of Bio- and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | | | - Liisa M Uotila
- Research Services, University of Helsinki, Helsinki, Finland
| |
Collapse
|
6
|
Knight JM, Mandal P, Morlacchi P, Mak G, Li E, Madison M, Landers C, Saxton B, Felix E, Gilbert B, Sederstrom J, Varadhachary A, Singh MM, Chatterjee D, Corry DB, McMurray JS. Small molecule targeting of the STAT5/6 Src homology 2 (SH2) domains to inhibit allergic airway disease. J Biol Chem 2018; 293:10026-10040. [PMID: 29739850 PMCID: PMC6028980 DOI: 10.1074/jbc.ra117.000567] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Revised: 04/02/2018] [Indexed: 11/06/2022] Open
Abstract
Asthma is a chronic inflammatory disease of the lungs and airways and one of the most burdensome of all chronic maladies. Previous studies have established that expression of experimental and human asthma requires the IL-4/IL-13/IL-4 receptor α (IL-4Rα) signaling pathway, which activates the transcription factor STAT6. However, no small molecules targeting this important pathway are currently in clinical development. To this end, using a preclinical asthma model, we sought to develop and test a small-molecule inhibitor of the Src homology 2 domains in mouse and human STAT6. We previously developed multiple peptidomimetic compounds on the basis of blocking the docking site of STAT6 to IL-4Rα and phosphorylation of Tyr641 in STAT6. Here, we expanded the scope of our initial in vitro structure-activity relationship studies to include central and C-terminal analogs of these peptides to develop a lead compound, PM-43I. Conducting initial dose range, toxicity, and pharmacokinetic experiments with PM-43I, we found that it potently inhibits both STAT5- and STAT6-dependent allergic airway disease in mice. Moreover, PM-43I reversed preexisting allergic airway disease in mice with a minimum ED50 of 0.25 μg/kg. Of note, PM-43I was efficiently cleared through the kidneys with no long-term toxicity. We conclude that PM-43I represents the first of a class of small molecules that may be suitable for further clinical development against asthma.
Collapse
Affiliation(s)
- J Morgan Knight
- From the Departments of Medicine,
- Pathology and Immunology, and
| | - Pijus Mandal
- the Department of Experimental Therapeutics, MD Anderson Cancer Center, Houston, Texas
| | - Pietro Morlacchi
- the Department of Experimental Therapeutics, MD Anderson Cancer Center, Houston, Texas
| | | | - Evan Li
- From the Departments of Medicine
- the Biology of Inflammation Center, Baylor College of Medicine, Houston, Texas 77030
| | - Matthew Madison
- the Translational Biology and Molecular Medicine Program, and
| | - Cameron Landers
- the Translational Biology and Molecular Medicine Program, and
| | | | - Ed Felix
- the Department of Experimental Therapeutics, MD Anderson Cancer Center, Houston, Texas
| | | | - Joel Sederstrom
- the Flow Cytometry Core Facility, Baylor College of Medicine, Houston, Texas
| | - Atul Varadhachary
- Fannin Innovation Studio and Atrapos Therapeutics, LLC, Houston, Texas 77027
| | - Melissa M Singh
- Fannin Innovation Studio and Atrapos Therapeutics, LLC, Houston, Texas 77027
| | - Dev Chatterjee
- Fannin Innovation Studio and Atrapos Therapeutics, LLC, Houston, Texas 77027
| | - David B Corry
- From the Departments of Medicine,
- Pathology and Immunology, and
- the Biology of Inflammation Center, Baylor College of Medicine, Houston, Texas 77030
- the Translational Biology and Molecular Medicine Program, and
- the Michael E. Debakey Veterans Affairs Center for Translational Research in Inflammatory Diseases, Houston, Texas 77030, and
| | - John S McMurray
- the Department of Experimental Therapeutics, MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
7
|
Wawrzyniak P, Akdis CA, Finkelman FD, Rothenberg ME. Advances and highlights in mechanisms of allergic disease in 2015. J Allergy Clin Immunol 2016; 137:1681-1696. [PMID: 27090934 DOI: 10.1016/j.jaci.2016.02.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 02/03/2016] [Accepted: 02/17/2016] [Indexed: 01/08/2023]
Abstract
This review highlights some of the advances in mechanisms of allergic disease, particularly anaphylaxis, including food allergy, drug hypersensitivity, atopic dermatitis (AD), allergic conjunctivitis, and airway diseases. During the last year, a mechanistic advance in food allergy was achieved by focusing on mechanisms of allergen sensitization. Novel biomarkers and treatment for mastocytosis were presented in several studies. Novel therapeutic approaches in the treatment of atopic dermatitis and psoriasis showed that promising supplementation of the infant's diet in the first year of life with immunoactive prebiotics might have a preventive role against early development of AD and that therapeutic approaches to treat AD in children might be best directed to the correction of a TH2/TH1 imbalance. Several studies were published emphasizing the role of the epithelial barrier in patients with allergic diseases. An impaired skin barrier as a cause for sensitization to food allergens in children and its relationship to filaggrin mutations has been an important development. Numerous studies presented new approaches for improvement of epithelial barrier function and novel biologicals used in the treatment of inflammatory skin and eosinophilic diseases. In addition, novel transcription factors and signaling molecules that can develop as new possible therapeutic targets have been reported.
Collapse
Affiliation(s)
- Paulina Wawrzyniak
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zürich, Davos, Switzerland, Christine Kühne-Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zürich, Davos, Switzerland, Christine Kühne-Center for Allergy Research and Education (CK-CARE), Davos, Switzerland.
| | - Fred D Finkelman
- Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, University of Cincinnati College of Medicine, and the Department of Medicine, Cincinnati Veterans Affairs Medical Center, Cincinnati, Ohio
| | - Marc E Rothenberg
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio
| |
Collapse
|
8
|
Abstract
PURPOSE OF REVIEW Research on atopic dermatitis is actively growing and continuously completing our knowledge on the pathophysiology of this complex disease. RECENT FINDINGS Genome-wide association studies revealed new susceptibility loci for atopic dermatitis. In addition, different tissue-specific patterns of DNA methylation have been identified as first evidence for the relevance of epigenetic modifications in atopic dermatitis. Moreover, interest is emerging on the role of the skin and gut microbiome in atopic dermatitis. Signals mediated via pattern recognition receptors of the innate immune system have been analyzed in more detail, and the role of cytokines, such as IL-22, IL-25, IL-31 and IL-33 as well as innate lymphoid cells, has been studied. SUMMARY Taken together, better knowledge of atopic dermatitis pathways will form the basis for the development of rationale-based therapeutic approaches in the future.
Collapse
|
9
|
Gupta A, Espinosa V, Galusha LE, Rahimian V, Miro KL, Rivera-Medina A, Kasinathan C, Capitle E, Aguila HA, Kachlany SC. Expression and targeting of lymphocyte function-associated antigen 1 (LFA-1) on white blood cells for treatment of allergic asthma. J Leukoc Biol 2014; 97:439-46. [PMID: 25341726 DOI: 10.1189/jlb.5hi0414-196r] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Allergic asthma is a chronic respiratory disease that results from an exaggerated inflammatory response in the airways. Environment stimuli, such as pollen and HDM, cause activation and migration of inflammatory WBCs into the respiratory tract, where they cause lung damage. Migration of these WBCs is dependent on the active configuration of the β2 integrin LFA-1. The experimental therapeutic agent LtxA specifically targets active LFA-1 and causes cell death. We investigated the association between LFA-1 and allergic asthma and hypothesized that targeting LFA-1 with LtxA could be an attractive strategy for treatment of the condition. We examined LFA-1 (CD11a) levels on PBMCs from patients with allergic asthma compared with healthy controls. Patients exhibited a significantly higher percentage of PBMCs expressing LFA-1 than healthy controls. Furthermore, the level of LFA-1 expression on patient PBMCs was greater than on healthy PBMCs. We identified a unique cellular population in patients that consisted of CD4(-) CD11a(hi) cells. We also evaluated LtxA in a HDM extract-induced mouse model for allergic asthma. LtxA caused resolution of disease in mice, as demonstrated by a decrease in BALF WBCs, a reduction in pulmonary inflammation and tissue remodeling, and a decrease in proinflammatory cytokines IL-4, IL-5, IL-9, IL-17F, and IL-23α in lung tissue. LFA-1 may serve as an important marker in allergic asthma, and the elimination of activated WBCs by use of LtxA could be a viable therapeutic strategy for treating patients with this condition.
Collapse
Affiliation(s)
- Anukriti Gupta
- *Department of Oral Biology, Rutgers School of Dental Medicine, Newark, New Jersey, USA; Departments of Medicine, Center for Immunity and Inflammation and Division of Allergy and Immunology, and Pediatrics, Rutgers New Jersey Medical School, Newark, New Jersey, USA; and Actinobac Biomed, New Brunswick, New Jersey, USA
| | - Vanessa Espinosa
- *Department of Oral Biology, Rutgers School of Dental Medicine, Newark, New Jersey, USA; Departments of Medicine, Center for Immunity and Inflammation and Division of Allergy and Immunology, and Pediatrics, Rutgers New Jersey Medical School, Newark, New Jersey, USA; and Actinobac Biomed, New Brunswick, New Jersey, USA
| | - Lindsey E Galusha
- *Department of Oral Biology, Rutgers School of Dental Medicine, Newark, New Jersey, USA; Departments of Medicine, Center for Immunity and Inflammation and Division of Allergy and Immunology, and Pediatrics, Rutgers New Jersey Medical School, Newark, New Jersey, USA; and Actinobac Biomed, New Brunswick, New Jersey, USA
| | - Vahid Rahimian
- *Department of Oral Biology, Rutgers School of Dental Medicine, Newark, New Jersey, USA; Departments of Medicine, Center for Immunity and Inflammation and Division of Allergy and Immunology, and Pediatrics, Rutgers New Jersey Medical School, Newark, New Jersey, USA; and Actinobac Biomed, New Brunswick, New Jersey, USA
| | - Katie L Miro
- *Department of Oral Biology, Rutgers School of Dental Medicine, Newark, New Jersey, USA; Departments of Medicine, Center for Immunity and Inflammation and Division of Allergy and Immunology, and Pediatrics, Rutgers New Jersey Medical School, Newark, New Jersey, USA; and Actinobac Biomed, New Brunswick, New Jersey, USA
| | - Amariliz Rivera-Medina
- *Department of Oral Biology, Rutgers School of Dental Medicine, Newark, New Jersey, USA; Departments of Medicine, Center for Immunity and Inflammation and Division of Allergy and Immunology, and Pediatrics, Rutgers New Jersey Medical School, Newark, New Jersey, USA; and Actinobac Biomed, New Brunswick, New Jersey, USA
| | - Chinnaswamy Kasinathan
- *Department of Oral Biology, Rutgers School of Dental Medicine, Newark, New Jersey, USA; Departments of Medicine, Center for Immunity and Inflammation and Division of Allergy and Immunology, and Pediatrics, Rutgers New Jersey Medical School, Newark, New Jersey, USA; and Actinobac Biomed, New Brunswick, New Jersey, USA
| | - Eugenio Capitle
- *Department of Oral Biology, Rutgers School of Dental Medicine, Newark, New Jersey, USA; Departments of Medicine, Center for Immunity and Inflammation and Division of Allergy and Immunology, and Pediatrics, Rutgers New Jersey Medical School, Newark, New Jersey, USA; and Actinobac Biomed, New Brunswick, New Jersey, USA
| | - Helen A Aguila
- *Department of Oral Biology, Rutgers School of Dental Medicine, Newark, New Jersey, USA; Departments of Medicine, Center for Immunity and Inflammation and Division of Allergy and Immunology, and Pediatrics, Rutgers New Jersey Medical School, Newark, New Jersey, USA; and Actinobac Biomed, New Brunswick, New Jersey, USA
| | - Scott C Kachlany
- *Department of Oral Biology, Rutgers School of Dental Medicine, Newark, New Jersey, USA; Departments of Medicine, Center for Immunity and Inflammation and Division of Allergy and Immunology, and Pediatrics, Rutgers New Jersey Medical School, Newark, New Jersey, USA; and Actinobac Biomed, New Brunswick, New Jersey, USA
| |
Collapse
|
10
|
Wang Q, Tan MT, Keegan BP, Barry MA, Heffernan MJ. Time course study of the antigen-specific immune response to a PLGA microparticle vaccine formulation. Biomaterials 2014; 35:8385-93. [PMID: 24986256 DOI: 10.1016/j.biomaterials.2014.05.067] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 05/22/2014] [Indexed: 11/28/2022]
Abstract
Microparticle-based vaccine delivery systems are known to promote enhanced immune responses to protein antigens and can elicit TH1-biased responses when used in combination with Toll-like receptor (TLR) agonists. It is important to understand the kinetics of the immune responses to microparticle-based protein vaccines in order to predict the duration of protective immunity and to optimize prime-boost vaccination regimens. We carried out a 10-week time course study to investigate the magnitude and kinetics of the antibody and cellular immune responses to poly(lactic-co-glycolic acid) (PLGA) microparticles containing 40 μg ovalbumin (OVA) protein and 16 μg CpG-ODN adjuvant (MP/OVA/CpG) in comparison to OVA-containing microparticles, soluble OVA plus CpG, or OVA formulated with Alhydrogel(®) aluminum adjuvant. Mice vaccinated with MP/OVA/CpG developed the highest TH1-associated IgG2b and IgG2c antibody titers, while also eliciting TH2-associated IgG1 antibody titers on par with Alhydrogel(®)-formulated OVA, with all IgG subtype titers peaking at day 56. The MP/OVA/CpG vaccine also induced the highest antigen-specific splenocyte IFN-γ responses, with high levels of IFN-γ responses persisting until day 42. Thus the MP/OVA/CpG formulation produced a sustained and heightened humoral and cellular immune response, with an overall TH1 bias, while maintaining high levels of IgG1 antibody equivalent to that seen with Alhydrogel(®) adjuvant. The time course kinetics study provides a useful baseline for designing vaccination regimens for microparticle-based protein vaccines.
Collapse
Affiliation(s)
- Qian Wang
- Sabin Vaccine Institute and Texas Children's Hospital Center for Vaccine Development, Department of Pediatrics (Section of Pediatric Tropical Medicine), Baylor College of Medicine, 1102 Bates Street, Houston, TX 77030, USA
| | - Melody T Tan
- Department of Bioengineering, Rice University, 6100 Main Street, Houston, TX 77005, USA
| | - Brian P Keegan
- Sabin Vaccine Institute and Texas Children's Hospital Center for Vaccine Development, Department of Pediatrics (Section of Pediatric Tropical Medicine), Baylor College of Medicine, 1102 Bates Street, Houston, TX 77030, USA
| | - Meagan A Barry
- Medical Scientist Training Program and Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Michael J Heffernan
- Sabin Vaccine Institute and Texas Children's Hospital Center for Vaccine Development, Department of Pediatrics (Section of Pediatric Tropical Medicine), Baylor College of Medicine, 1102 Bates Street, Houston, TX 77030, USA; Department of Molecular Virology & Microbiology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; National School of Tropical Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA.
| |
Collapse
|