1
|
Cao X, Fu YX, Peng H. Promising Cytokine Adjuvants for Enhancing Tuberculosis Vaccine Immunity. Vaccines (Basel) 2024; 12:477. [PMID: 38793728 PMCID: PMC11126114 DOI: 10.3390/vaccines12050477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 04/23/2024] [Accepted: 04/24/2024] [Indexed: 05/26/2024] Open
Abstract
Tuberculosis, caused by Mycobacterium tuberculosis (M. tuberculosis), remains a formidable global health challenge, affecting a substantial portion of the world's population. The current tuberculosis vaccine, bacille Calmette-Guérin (BCG), offers limited protection against pulmonary tuberculosis in adults, underscoring the critical need for innovative vaccination strategies. Cytokines are pivotal in modulating immune responses and have been explored as potential adjuvants to enhance vaccine efficacy. The strategic inclusion of cytokines as adjuvants in tuberculosis vaccines holds significant promise for augmenting vaccine-induced immune responses and strengthening protection against M. tuberculosis. This review delves into promising cytokines, such as Type I interferons (IFNs), Type II IFN, interleukins such as IL-2, IL-7, IL-15, IL-12, and IL-21, alongside the use of a granulocyte-macrophage colony-stimulating factor (GM-CSF) as an adjuvant, which has shown effectiveness in boosting immune responses and enhancing vaccine efficacy in tuberculosis models.
Collapse
Affiliation(s)
- Xuezhi Cao
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510182, China;
- Guangzhou National Laboratory, Bio-Island, Guangzhou 510005, China
| | - Yang-Xin Fu
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Hua Peng
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510182, China;
- Guangzhou National Laboratory, Bio-Island, Guangzhou 510005, China
| |
Collapse
|
2
|
Rahman T, Das A, Abir MH, Nafiz IH, Mahmud AR, Sarker MR, Emran TB, Hassan MM. Cytokines and their role as immunotherapeutics and vaccine Adjuvants: The emerging concepts. Cytokine 2023; 169:156268. [PMID: 37320965 DOI: 10.1016/j.cyto.2023.156268] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 06/02/2023] [Accepted: 06/06/2023] [Indexed: 06/17/2023]
Abstract
Cytokines are a protein family comprising interleukins, lymphokines, chemokines, monokines and interferons. They are significant constituents of the immune system, and they act in accordance with specific cytokine inhibiting compounds and receptors for the regulation of immune responses. Cytokine studies have resulted in the establishment of newer therapies which are being utilized for the treatment of several malignant diseases. The advancement of these therapies has occurred from two distinct strategies. The first strategy involves administrating the recombinant and purified cytokines, and the second strategy involves administrating the therapeutics which inhibits harmful effects of endogenous and overexpressed cytokines. Colony stimulating factors and interferons are two exemplary therapeutics of cytokines. An important effect of cytokine receptor antagonist is that they can serve as anti-inflammatory agents by altering the treatments of inflammation disorder, therefore inhibiting the effects of tumour necrosis factor. In this article, we have highlighted the research behind the establishment of cytokines as therapeutics and vaccine adjuvants, their role of immunotolerance, and their limitations.
Collapse
Affiliation(s)
- Tanjilur Rahman
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, University of Chittagong, Chattogram 4331, Bangladesh
| | - Ayan Das
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, University of Chittagong, Chattogram 4331, Bangladesh
| | - Mehedy Hasan Abir
- Faculty of Food Science and Technology, Chattogram Veterinary and Animal Sciences University, Chattogram 4225, Bangladesh
| | - Iqbal Hossain Nafiz
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, University of Chittagong, Chattogram 4331, Bangladesh
| | - Aar Rafi Mahmud
- Department of Biochemistry and Molecular Biology, Mawlana Bhashani Science and Technology University, Tangail 1902, Bangladesh
| | - Md Rifat Sarker
- Department of Biochemistry and Molecular Biology, Mawlana Bhashani Science and Technology University, Tangail 1902, Bangladesh
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chattogram 4381, Bangladesh; Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh
| | - Mohammad Mahmudul Hassan
- Department of Physiology, Biochemistry and Pharmacology, Faculty of Veterinary Medicine, Chattogram Veterinary and Animal Sciences University, Chattogram 4225, Bangladesh; Queensland Alliance for One Health Sciences, School of Veterinary Science, The University of Queensland, Queensland 4343, Australia.
| |
Collapse
|
3
|
Ashwood P. Preliminary Findings of Elevated Inflammatory Plasma Cytokines in Children with Autism Who Have Co-Morbid Gastrointestinal Symptoms. Biomedicines 2023; 11:436. [PMID: 36830973 PMCID: PMC9952966 DOI: 10.3390/biomedicines11020436] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 01/27/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Autism spectrum disorder (AU) is present in approximately 2% of the population and is often associated with co-morbidities that can impact quality of life. One of the most common co-morbidities in autism is the presence of gastrointestinal (GI) symptoms consisting of irregular bowel habits such as constipation, diarrhea, or alternating bowel habit. Evidence of immune infiltration and immune activation has been shown in the ileum and colon of children with AU with GI symptoms. Moreover, immune dysfunction is a contributing factor in many GI diseases, and we hypothesize that it would be more apparent in children with AU that exhibit GI symptoms than those who do not present with GI symptoms. The aim of this preliminary study was to determine whether there are altered cytokine levels in plasma in children with AU with GI symptoms compared with children with AU without GI symptoms, typically developing (TD) children with GI symptoms and TD children without GI symptoms, from the same population-based cohort. Plasma cytokine levels were assessed by multiplex assays. No differences in plasma cytokines were observed in TD controls with or without GI symptoms; however, many innate (IL-1α, TNFα, GM-CSF, IFNα) and adaptive cytokines (IL-4, IL-13, IL-12p70) were increased in AU children with GI symptoms compared with children with AU with no GI symptoms. The mucosal relevant cytokine IL-15 was increased in AU with GI symptoms compared with all groups. In contrast, the regulatory cytokine IL-10, was reduced in AU with GI symptoms and may suggest an imbalance in pro-inflammatory/regulatory signals. These data suggest that children with AU and GI symptoms have an imbalance in their immune response that is evident in their circulating plasma cytokine levels. A finding that could point to potential therapeutic and/or monitoring strategies for GI issues in AU.
Collapse
Affiliation(s)
- Paul Ashwood
- Department of Medical Microbiology and Immunology, School of Medicine, MIND Institute, University of California Davis, Davis, CA 95616, USA
| |
Collapse
|
4
|
Feng H, Zhao X, Xie J, Bai X, Fu W, Chen H, Tang H, Wang X, Dong C. Pathogen-associated T follicular helper cell plasticity is critical in anti-viral immunity. SCIENCE CHINA LIFE SCIENCES 2022; 65:1075-1090. [DOI: 10.1007/s11427-021-2055-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 09/26/2021] [Indexed: 01/12/2023]
|
5
|
Zhang BD, Wu JJ, Li WH, Hu HG, Zhao L, He PY, Zhao YF, Li YM. STING and TLR7/8 agonists-based nanovaccines for synergistic antitumor immune activation. NANO RESEARCH 2022; 15:6328-6339. [PMID: 35464625 PMCID: PMC9014842 DOI: 10.1007/s12274-022-4282-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 02/28/2022] [Accepted: 03/01/2022] [Indexed: 05/07/2023]
Abstract
UNLABELLED Immunostimulatory therapies based on pattern recognition receptors (PRRs) have emerged as an effective approach in the fight against cancer, with the ability to recruit tumor-specific lymphocytes in a low-immunogenicity tumor environment. The agonist cyclic dinucleotides (CDNs) of the stimulator of interferon gene (STING) are a group of very promising anticancer molecules that increase tumor immunogenicity by activating innate immunity. However, the tumor immune efficacy of CDNs is limited by several factors, including relatively narrow cytokine production, inefficient delivery to STING, and rapid clearance. In addition, a single adjuvant molecule is unable to elicit a broad cytokine response and thus cannot further amplify the anticancer effect. To address this problem, two or more agonist molecules are often used together to synergistically enhance immune efficacy. In this work, we found that a combination of the STING agonist CDGSF and the Toll-like receptor 7/8 (TLR7/8) agonist 522 produced a broader cytokine response. Subsequently, we developed multicomponent nanovaccines (MCNVs) consisting of a PC7A polymer as a nanocarrier encapsulating the antigen OVA and adjuvant molecules. These MCNVs activate bone marrow-derived dendritic cells (BMDCs) to produce multiple proinflammatory factors that promote antigen cross-presentation to stimulate specific antitumor T-cell responses. In in vivo experiments, we observed that MCNVs triggered a strong T-cell response in tumor-infiltrating lymphocytes, resulting in significant tumor regression and, notably, a 100% survival rate in mice through 25 days without other partnering therapies. These data suggest that our nanovaccines have great potential to advance cancer immunotherapy with increased durability and potency. ELECTRONIC SUPPLEMENTARY MATERIAL Supplementary material (synthesis of CDGSF, 522, PC7A and OVA; preparation of MCNVs; representative gating strategies for flow cytometry) is available in the online version of this article at 10.1007/s12274-022-4282-x.
Collapse
Affiliation(s)
- Bo-Dou Zhang
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, 100084 China
| | - Jun-Jun Wu
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, 100084 China
| | - Wen-Hao Li
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, 100084 China
| | - Hong-Guo Hu
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, 100084 China
| | - Lang Zhao
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, 100084 China
| | - Pei-Yang He
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, 100084 China
| | - Yu-Fen Zhao
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, 100084 China
- Institute of Drug Discovery Technology, Ningbo University, Ningbo, 315201 China
| | - Yan-Mei Li
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, 100084 China
- Beijing Institute for Brain Disorders, Beijing, 100069 China
| |
Collapse
|
6
|
Abstract
Many options now exist for constructing oral vaccines which, in experimental systems, have shown themselves to be able to generate highly effective immunity against infectious diseases. Their suitability for implementation in clinical practice, however, for prevention of outbreaks, particularly in low- and middle-income countries (LMIC), is not always guaranteed, because of factors such as cost, logistics and cultural and environmental conditions. This brief overview provides a summary of the various approaches which can be adopted, and evaluates them from a pharmaceutical point, taking into account potential regulatory issues, expense, manufacturing complexity, etc., all of which can determine whether a vaccine approach will be successful in the late stages of development. Attention is also drawn to problems arising from inadequate diet, which impacts upon success in stimulating effective immunity, and identifies the use of lipid-based carriers as a way to counteract the problem of nutritional deficiencies in vaccination campaigns.
Collapse
Affiliation(s)
- R. R. C. New
- Middlesex UniversityHendon, LondonUK
- Vaxcine (UK) Limited, London Bioscience Innovation CentreLondonUK
| |
Collapse
|
7
|
Smith SP, Wu G, Fooks AR, Ma J, Banyard AC. Trying to treat the untreatable: experimental approaches to clear rabies virus infection from the CNS. J Gen Virol 2019; 100:1171-1186. [PMID: 31237530 DOI: 10.1099/jgv.0.001269] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Rabies virus causes an invariably fatal encephalitis following the onset of clinical disease. Despite the availability of safe and effective vaccines, the clinical stages of rabies encephalitis remain untreatable, with few survivors being documented. A principal obstacle to the treatment of rabies is the neurotropic nature of the virus, with the blood-brain barrier size exclusion limit rendering the delivery of antiviral drugs and molecules to the central nervous system inherently problematic. This review focuses on efforts to try and overcome barriers to molecule delivery to treat clinical rabies and overviews current progress in the development of experimental live rabies virus vaccines that may have future applications in the treatment of clinical rabies, including the attenuation of rabies virus vectors through either the duplication or mutation of existing genes or the incorporation of non-viral elements within the genome. Rabies post-infection treatment (PIT) remains the holy grail of rabies research.
Collapse
Affiliation(s)
- Samuel P Smith
- Wildlife Zoonoses and Vector-borne Diseases Research Group, Animal and Plant Health Agency (APHA), Addlestone, Surrey, KT15 3NB, UK.,Institute for Infection and Immunity, St George's Hospital Medical School, University of London, London, UK
| | - Guanghui Wu
- Wildlife Zoonoses and Vector-borne Diseases Research Group, Animal and Plant Health Agency (APHA), Addlestone, Surrey, KT15 3NB, UK
| | - Anthony R Fooks
- Wildlife Zoonoses and Vector-borne Diseases Research Group, Animal and Plant Health Agency (APHA), Addlestone, Surrey, KT15 3NB, UK.,Institute for Infection and Immunity, St George's Hospital Medical School, University of London, London, UK.,Department of Clinical Infection, Microbiology and Immunology, Institute of Infection and Global Health, University of Liverpool, Liverpool, UK
| | - Julian Ma
- Institute for Infection and Immunity, St George's Hospital Medical School, University of London, London, UK
| | - Ashley C Banyard
- Institute for Infection and Immunity, St George's Hospital Medical School, University of London, London, UK.,School of Life Sciences, University of West Sussex, Falmer, West Sussex, UK.,Wildlife Zoonoses and Vector-borne Diseases Research Group, Animal and Plant Health Agency (APHA), Addlestone, Surrey, KT15 3NB, UK
| |
Collapse
|
8
|
Collier MA, Junkins RD, Gallovic MD, Johnson BM, Johnson MM, Macintyre AN, Sempowski GD, Bachelder EM, Ting JPY, Ainslie KM. Acetalated Dextran Microparticles for Codelivery of STING and TLR7/8 Agonists. Mol Pharm 2018; 15:4933-4946. [PMID: 30281314 DOI: 10.1021/acs.molpharmaceut.8b00579] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Vaccines are the most effective tool for preventing infectious diseases; however, subunit vaccines, considered the safest type, suffer from poor immunogenicity and require adjuvants to create a strong and sustained immune response. As adjuvants, pathogen-associated molecular patterns (PAMPs) offer potent immunostimulatory properties and defined mechanisms of action through their cognate pattern recognition receptors (PRRs). Their activity can be further enhanced through combining two or more PAMPs, particularly those that activate multiple immune signaling pathways. However, the cytosolic localization of many PRRs requires intracellular delivery of PAMPs for optimal biological activity, which is particularly true of the stimulator of interferon genes (STING) PRR. Using acetalated dextran (Ace-DEX) microparticles (MPs) encapsulating STING agonist 3'3'-cyclic GMP-AMP (cGAMP) combined with soluble PAMPS, we screened the effect of codelivery of adjuvants using primary mouse bone marrow derived dendritic cells (BMDCs). We identified that codelivery of cGAMP MPs and soluble Toll-like receptor 7/8 (TLR7/8) agonist resiquimod (R848) elicited the broadest cytokine response. cGAMP and R848 were then coencapsulated within Ace-DEX MPs via electrospray. Using the model antigen ovalbumin, we observed that Ace-DEX MPs coencapsulating cGAMP and R848 (cGAMP/R848 Ace-DEX MPs) induced antigen-specific cellular immunity, and a balanced Th1/Th2 humoral response that was greater than cGAMP Ace-DEX MPs alone and PAMPs delivered in separate MPs. These data indicate that polymeric Ace-DEX MPs loaded with STING and TLR7/8 agonists represent a potent cellular and humoral vaccine adjuvant.
Collapse
Affiliation(s)
| | | | | | | | | | - Andrew N Macintyre
- Duke Human Vaccine Institute , Duke University Medical Center , Durham , North Carolina 27710 , United States
| | - Gregory D Sempowski
- Duke Human Vaccine Institute , Duke University Medical Center , Durham , North Carolina 27710 , United States
| | | | | | | |
Collapse
|
9
|
Rose DR, Yang H, Serena G, Sturgeon C, Ma B, Careaga M, Hughes HK, Angkustsiri K, Rose M, Hertz-Picciotto I, Van de Water J, Hansen RL, Ravel J, Fasano A, Ashwood P. Differential immune responses and microbiota profiles in children with autism spectrum disorders and co-morbid gastrointestinal symptoms. Brain Behav Immun 2018; 70:354-368. [PMID: 29571898 PMCID: PMC5953830 DOI: 10.1016/j.bbi.2018.03.025] [Citation(s) in RCA: 139] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 03/08/2018] [Accepted: 03/19/2018] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVES Many studies have reported the increased presence of gastrointestinal (GI) symptoms in children with autism spectrum disorders (ASD). Altered microbiome profiles, pro-inflammatory responses and impaired intestinal permeability have been observed in children with ASD and co-morbid GI symptoms, yet few studies have compared these findings to ASD children without GI issues or similarly aged typical developing children. The aim of this study was to determine whether there are biological signatures in terms of immune dysfunction and microbiota composition in children with ASD with GI symptoms. METHODS Children were enrolled in one of four groups: ASD and GI symptoms of irregular bowel habits (ASDGI), children with ASD but without current or previous GI symptoms (ASDNoGI), typically developing children with GI symptoms (TDGI) and typically developing children without current or previous GI symptoms (TDNoGI). Peripheral blood mononuclear cells (PBMC) were isolated from the blood, stimulated and assessed for cytokine production, while stool samples were analyzed for microbial composition. RESULTS Following Toll-Like receptor (TLR)-4 stimulation, the ASDGI group produced increased levels of mucosa-relevant cytokines including IL-5, IL-15 and IL-17 compared to ASDNoGI. The production of the regulatory cytokine TGFβ1 was decreased in the ASDGI group compared with both the ASDNoGI and TDNoGI groups. Analysis of the microbiome at the family level revealed differences in microbiome composition between ASD and TD children with GI symptoms; furthermore, a predictive metagenome functional content analysis revealed that pathways were differentially represented between ASD and TD subjects, independently of the presence of GI symptoms. The ASDGI also showed an over-representation of the gene encoding zonulin, a molecule regulating gut permeability, compared to the other groups. CONCLUSIONS Overall our findings suggest that children with ASD who experience GI symptoms have an imbalance in their immune response, possibly influenced by or influencing metagenomic changes, and may have a propensity to impaired gut barrier function which may contribute to their symptoms and clinical outcome.
Collapse
Affiliation(s)
- Destanie R Rose
- Department of Medical Microbiology and Immunology, University of California Davis, USA; MIND Institute, University of California Davis, USA
| | - Houa Yang
- Department of Medical Microbiology and Immunology, University of California Davis, USA; MIND Institute, University of California Davis, USA
| | - Gloria Serena
- Division of Pediatric Gastroenterology and Nutrition, Center for Celiac Research, MassGeneral Hospital for Children, Boston, MA, USA; Graduate Program in Life Sciences University of Maryland School of Medicine, Baltimore, MD, USA
| | - Craig Sturgeon
- Division of Pediatric Gastroenterology and Nutrition, Center for Celiac Research, MassGeneral Hospital for Children, Boston, MA, USA; Graduate Program in Life Sciences University of Maryland School of Medicine, Baltimore, MD, USA
| | - Bing Ma
- Institute of Genomic Science, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Milo Careaga
- Department of Medical Microbiology and Immunology, University of California Davis, USA; MIND Institute, University of California Davis, USA
| | - Heather K Hughes
- Department of Medical Microbiology and Immunology, University of California Davis, USA; MIND Institute, University of California Davis, USA
| | - Kathy Angkustsiri
- MIND Institute, University of California Davis, USA; Department of Pediatrics, University of California Davis, USA; Children's Center for Environmental Health, University of California Davis, CA, USA
| | - Melissa Rose
- Children's Center for Environmental Health, University of California Davis, CA, USA; Public Health Sciences, University of California Davis, CA, USA
| | - Irva Hertz-Picciotto
- MIND Institute, University of California Davis, USA; Children's Center for Environmental Health, University of California Davis, CA, USA; Public Health Sciences, University of California Davis, CA, USA
| | - Judy Van de Water
- MIND Institute, University of California Davis, USA; Children's Center for Environmental Health, University of California Davis, CA, USA; Division of Rheumatology, Allergy and Clinical Immunology, University of California Davis, CA, USA
| | - Robin L Hansen
- MIND Institute, University of California Davis, USA; Department of Pediatrics, University of California Davis, USA; Children's Center for Environmental Health, University of California Davis, CA, USA
| | - Jacques Ravel
- Institute of Genomic Science, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Alessio Fasano
- Division of Pediatric Gastroenterology and Nutrition, Center for Celiac Research, MassGeneral Hospital for Children, Boston, MA, USA
| | - Paul Ashwood
- Department of Medical Microbiology and Immunology, University of California Davis, USA; MIND Institute, University of California Davis, USA; Children's Center for Environmental Health, University of California Davis, CA, USA.
| |
Collapse
|
10
|
Ganshina IV. Serous cavities of coelomic origin as possible organs of the immune system. Part 1. ACTA ACUST UNITED AC 2016. [DOI: 10.1134/s2079086416060025] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
11
|
Kraszewska-Głomba B, Matkowska-Kocjan A, Miśkiewicz K, Szymańska-Toczek Z, Wójcik M, Banyś D, Szenborn L. Mumps, measles and rubella vaccination in children with PFAPA syndrome. Vaccine 2016; 34:5903-5906. [PMID: 27997341 DOI: 10.1016/j.vaccine.2016.10.035] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 10/12/2016] [Accepted: 10/13/2016] [Indexed: 11/26/2022]
Abstract
There is no published data regarding immunologic response to vaccinations in children with PFAPA syndrome (periodic fever, aphthous stomatitis, pharyngitis and cervical adenitis). The aim of this study was to evaluate mumps, measles and rubella immunity in children with PFAPA. 31 children with PFAPA syndrome and 22 healthy children (control group - CG) were recruited to the study. All children were previously vaccinated with one dose of MMR vaccine according to the Polish obligatory vaccination schedule. The patients from both groups were evaluated for anti-measles, anti-mumps and anti-rubella IgG antibodies concentrations (ELISA tests; the reference values for protective antibody levels were 150IU/L, 16RU/L and 11IU/ml respectively). The percentage of patients with protective antibodies levels was as follows: measles - 93.55% of PFAPA and 95.45% of CG patients (p=0.77); mumps - 74.19% of PFAPA and 95.45% of CG patients (p=0.02); rubella - 80.65% of PFAPA and 90.9% of CG patients (p=0.30). CONCLUSIONS Children with PFAPA syndrome present a good response to the measles and rubella component of the MMR vaccine, however immunity against mumps after one dose of MMR may not be sufficient. Further investigation concerning immunity against vaccine-preventable diseases and the safety of vaccinations in children with periodic fever syndromes is required.
Collapse
Affiliation(s)
- Barbara Kraszewska-Głomba
- Department and Clinic of Paediatric Infectious Diseases, Wrocław Medical University, 2-2A Chałubińskiego, 50-368 Wrocław, Poland.
| | - Agnieszka Matkowska-Kocjan
- Department and Clinic of Paediatric Infectious Diseases, Wrocław Medical University, 2-2A Chałubińskiego, 50-368 Wrocław, Poland.
| | - Katarzyna Miśkiewicz
- Department and Clinic of Paediatric Infectious Diseases, Wrocław Medical University, 2-2A Chałubińskiego, 50-368 Wrocław, Poland.
| | - Zofia Szymańska-Toczek
- Department and Clinic of Paediatric Infectious Diseases, Wrocław Medical University, 2-2A Chałubińskiego, 50-368 Wrocław, Poland.
| | - Marta Wójcik
- Department and Clinic of Paediatric Infectious Diseases, Wrocław Medical University, 2-2A Chałubińskiego, 50-368 Wrocław, Poland.
| | - Dorota Banyś
- Department and Clinic of Paediatric Infectious Diseases, Wrocław Medical University, 2-2A Chałubińskiego, 50-368 Wrocław, Poland.
| | - Leszek Szenborn
- Department and Clinic of Paediatric Infectious Diseases, Wrocław Medical University, 2-2A Chałubińskiego, 50-368 Wrocław, Poland.
| |
Collapse
|
12
|
Patel S, Akalkotkar A, Bivona JJ, Lee JY, Park YK, Yu M, Colpitts SL, Vajdy M. Vitamin A or E and a catechin synergize as vaccine adjuvant to enhance immune responses in mice by induction of early interleukin-15 but not interleukin-1β responses. Immunology 2016; 148:352-62. [PMID: 27135790 DOI: 10.1111/imm.12614] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 04/16/2016] [Accepted: 04/22/2016] [Indexed: 12/31/2022] Open
Abstract
Vitamins A and E and select flavonoids in the family of catechins are well-defined small molecules that, if proven to possess immunomodulatory properties, hold promise as vaccine adjuvants and various therapies. In an effort to determine the in vivo immunomodulatory properties of these molecules, we found that although mucosal and systemic vaccinations with a recombinant HIV-1BaL gp120 with either a catechin, epigallo catechin gallate (EGCG) or pro-vitamin A (retinyl palmitate) alone in a vegetable-oil-in-water emulsion (OWE) suppressed antigen-specific responses, the combination of EGCG and vitamin A or E in OWE (Nutritive Immune-enhancing Delivery System, NIDS) synergistically enhanced adaptive B-cell, and CD4(+) and CD8(+) T-cell responses, following induction of relatively low local and systemic innate tumour necrosis factor-α (TNF-α), interleukin-6 (IL-6) and IL-17, but relatively high levels of early systemic IL-15 responses. For induction of adaptive interferon-γ and TNF-α responses by CD4(+) and CD8(+) T cells, the adjuvant effect of NIDS was dependent on both IL-15 and its receptor. In addition, the anti-oxidant activity of NIDS correlated positively with higher expression of the superoxide dismutase 1, an enzyme involved in reactive oxygen species elimination but negatively with secretion of IL-1β. This suggests that the mechanism of action of NIDS is dependent on anti-oxidant activity and IL-15, but independent of IL-1β and inflammasome formation. These data show that this approach in nutritive vaccine adjuvant design holds promise for the development of potentially safer effective vaccines.
Collapse
Affiliation(s)
| | | | | | - Ji-Young Lee
- Department of Nutritional Sciences, College of Agriculture Health and Natural Resources, University of Connecticut, Storrs, CT, USA
| | - Young-Ki Park
- Department of Nutritional Sciences, College of Agriculture Health and Natural Resources, University of Connecticut, Storrs, CT, USA
| | | | - Sara L Colpitts
- Department of Immunology, Health Center for Integrated Immunology and Vaccine Research, University of Connecticut, Farmington, CT, USA
| | | |
Collapse
|
13
|
Treatment with Interleukin-7 Restores Host Defense against Pneumocystis in CD4+ T-Lymphocyte-Depleted Mice. Infect Immun 2015; 84:108-19. [PMID: 26483405 DOI: 10.1128/iai.01189-15] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 10/10/2015] [Indexed: 12/16/2022] Open
Abstract
Pneumocystis pneumonia (PCP) is a major cause of morbidity and mortality in patients with HIV infection. CD4(+) T lymphocytes are critical for host defense against this infection, but in the absence of CD4(+) T lymphocytes, CD8(+) T lymphocytes may provide limited host defense. The cytokine interleukin-7 (IL-7) functions to enhance lymphocyte proliferation, survival, and recruitment of immune cells to sites of infection. However, there is little known about the role of IL-7 in PCP or its potential use as an immunotherapeutic agent. We hypothesized that treatment with recombinant human IL-7 (rhIL-7) would augment host defense against Pneumocystis and accelerate pathogen clearance in CD4-depleted mice. Control and CD4-depleted mice were infected with Pneumocystis, and rhIL-7 was administered via intraperitoneal injection. Our studies indicate that endogenous murine IL-7 is part of the normal host response to Pneumocystis murina and that administration of rhIL-7 markedly enhanced clearance of Pneumocystis in CD4-depleted mice. Additionally, we observed increased recruitment of CD8(+) T lymphocytes to the lungs and decreased apoptosis of pulmonary CD8(+) T lymphocytes in rhIL-7-treated animals compared to those in untreated mice. The antiapoptotic effect of rhIL-7 was associated with increased levels of Bcl-2 protein in T lymphocytes. rhIL-7 immunotherapy in CD4-depleted mice also increased the number of gamma interferon (IFN-γ)-positive CD8(+) central memory T lymphocytes in the lungs. We conclude that rhIL-7 has a potent therapeutic effect in the treatment of murine Pneumocystis pneumonia in CD4-depleted mice. This therapeutic effect is mediated through enhanced recruitment of CD8(+) T cells and decreased apoptosis of lung T lymphocytes, with a preferential action on central memory CD8(+) T lymphocytes.
Collapse
|
14
|
Dhama K, Saminathan M, Jacob SS, Singh M, Karthik K, . A, Tiwari R, Sunkara LT, Malik YS, Singh RK. Effect of Immunomodulation and Immunomodulatory Agents on Health with some Bioactive Principles, Modes of Action and Potent Biomedical Applications. INT J PHARMACOL 2015. [DOI: 10.3923/ijp.2015.253.290] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
15
|
Qiu Y, Wang W, Xiao W, Yang H. Role of the intestinal cytokine microenvironment in shaping the intraepithelial lymphocyte repertoire. J Leukoc Biol 2015; 97:849-857. [PMID: 25765675 DOI: 10.1189/jlb.3ru1014-465r] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Revised: 01/17/2015] [Accepted: 02/16/2015] [Indexed: 12/11/2022] Open
Abstract
Local resident IELs are composed of distinct subsets of T cells with potent cytolytic and immunoregulatory capacities. As IELs are located within this unique interface between the core of the body and the outside environment, the specific development and function of intestinal IELs must be tightly regulated. To accomplish this, the cytokine microenvironment of the intestine has evolved sophisticated mechanisms that modulate the phenotype, ontogeny, and function of these cells. In this review, we summarize the evidence demonstrating the origin of certain intestinal cytokines, including IL-7, IL-15, IL-2, TGF-β, and SCF and discuss what influence such cytokines may have on IELs. Moreover, we review data suggesting that the abnormal expression of cytokines that leads to the heightened activation of IELs may also contribute to immunopathological responses or exacerbate inflammatory diseases, such as IBD and celiac disease, or promote cancer development and progression.
Collapse
Affiliation(s)
- Yuan Qiu
- Department of General Surgery, Xinqiao Hospital, The Third Military Medical University, Chongqing, China
| | - Wensheng Wang
- Department of General Surgery, Xinqiao Hospital, The Third Military Medical University, Chongqing, China
| | - Weidong Xiao
- Department of General Surgery, Xinqiao Hospital, The Third Military Medical University, Chongqing, China
| | - Hua Yang
- Department of General Surgery, Xinqiao Hospital, The Third Military Medical University, Chongqing, China
| |
Collapse
|
16
|
|
17
|
Gorzin Z, Gorzin AA, Tabarraei A, Behnampour N, Irani S, Ghaemi A. Immunogenicity evaluation of a DNA vaccine expressing the hepatitis C virus non-structural protein 2 gene in C57BL/6 mice. IRANIAN BIOMEDICAL JOURNAL 2014; 18:1-7. [PMID: 24375156 DOI: 10.6091/ibj.1231.2013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUNDS Most of the hepatitis C virus (HCV) infections elicit poor immune responses and 75% to 85% of cases become chronic; therefore, the development of an effective vaccine against HCV is of paramount importance. In this study, we aimed to evaluate co-administration of HCV non-Structural Protein 2 and IL-12 DNA vaccines in C57BL/6 mice. METHODS A plasmid encoding full-length HCV NS2 protein (non-structural protein 2) was generated and used to vaccinate mice. Negative control (an empty expression vector) was also employed to evaluate the background response. To investigate immune responses against vaccine, C57BL/6 mice received three doses of the vaccine with a two-week interval. Cellular immunity was assessed using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide assay for lymphocyte proliferation, lactate dehydrogenase release for cytotoxic T lymphocyte (CTL) activity and cytokine assay. RESULTS The findings demonstrated that immunization of mice with plasmid expressing HCV NS2 induced CTL response, interferon gamma production, and lymphocyte proliferation compared to negative control. The results also demonstrated that co-administration of IL-12 with the HCV NS2 plasmid induced significantly better immune response in C57BL/6 mice. CONCLUSION DNA vaccine encoding HCV NS2 is an effective candidate that can trigger CTL-based immune response against HCV. In addition, the results suggested that combining the DNA vaccine approach with immune stimulatory cytokines may significantly enhance antigen-specific immune responses.
Collapse
Affiliation(s)
- Zahra Gorzin
- Dept. of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Ali Akbar Gorzin
- Shiraz HIV/AIDS Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Alijan Tabarraei
- Dept. of Microbiology, School of Medicine, Infectious Diseases Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Naser Behnampour
- Dept. of Statistics, Gorgan Para-Medical School, Golestan University of Medical Sciences, Gorgan, Iran
| | - Shiva Irani
- Dept. of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Amir Ghaemi
- Golestan Research Center of Gastroenterology and Hepatology-GRCGH, Dept. of Microbiology, School of Medicine, Golestan University of Medical Sciences, Gorgan, Iran.,Shefa Neuroscience Research Center, Khatam Al Anbia Hospital, Tehran, Iran
| |
Collapse
|
18
|
Deng Y, Chu J, Ren Y, Fan Z, Ji X, Mundy-Bosse B, Yuan S, Hughes T, Zhang J, Cheema B, Camardo AT, Xia Y, Wu LC, Wang LS, He X, Kinghorn AD, Li X, Caligiuri MA, Yu J. The natural product phyllanthusmin C enhances IFN-γ production by human NK cells through upregulation of TLR-mediated NF-κB signaling. THE JOURNAL OF IMMUNOLOGY 2014; 193:2994-3002. [PMID: 25122922 DOI: 10.4049/jimmunol.1302600] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Natural products are a major source for cancer drug development. NK cells are a critical component of innate immunity with the capacity to destroy cancer cells, cancer-initiating cells, and clear viral infections. However, few reports describe a natural product that stimulates NK cell IFN-γ production and unravel a mechanism of action. In this study, through screening, we found that a natural product, phyllanthusmin C (PL-C), alone enhanced IFN-γ production by human NK cells. PL-C also synergized with IL-12, even at the low cytokine concentration of 0.1 ng/ml, and stimulated IFN-γ production in both human CD56(bright) and CD56(dim) NK cell subsets. Mechanistically, TLR1 and/or TLR6 mediated PL-C's activation of the NF-κB p65 subunit that in turn bound to the proximal promoter of IFNG and subsequently resulted in increased IFN-γ production in NK cells. However, IL-12 and IL-15Rs and their related STAT signaling pathways were not responsible for the enhanced IFN-γ secretion by PL-C. PL-C induced little or no T cell IFN-γ production or NK cell cytotoxicity. Collectively, we identify a natural product with the capacity to selectively enhance human NK cell IFN-γ production. Given the role of IFN-γ in immune surveillance, additional studies to understand the role of this natural product in prevention of cancer or infection in select populations are warranted.
Collapse
Affiliation(s)
- Youcai Deng
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH 43210; Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
| | - Jianhong Chu
- The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210
| | - Yulin Ren
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210
| | - Zhijin Fan
- The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210; State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin 300071, China
| | - Xiaotian Ji
- State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin 300071, China
| | | | - Shunzong Yuan
- The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210; Department of Lymphoma, Affiliated Hospital of Academy of Military Medical Sciences, Beijing 100071, China
| | - Tiffany Hughes
- The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210
| | - Jianying Zhang
- Center for Biostatistics, The Ohio State University, Columbus, OH 43210
| | - Baljash Cheema
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210
| | - Andrew T Camardo
- The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210
| | - Yong Xia
- Department of Molecular and Cellular Biochemistry, The Ohio State University College of Medicine, Columbus, OH 43210
| | - Lai-Chu Wu
- Department of Molecular and Cellular Biochemistry, The Ohio State University College of Medicine, Columbus, OH 43210
| | - Li-Shu Wang
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI 53226; and
| | - Xiaoming He
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210
| | - A Douglas Kinghorn
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210
| | - Xiaohui Li
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing 400038, China;
| | - Michael A Caligiuri
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH 43210; The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210;
| | - Jianhua Yu
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH 43210; The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210;
| |
Collapse
|
19
|
Ballesteros NA, Saint-Jean SSR, Perez-Prieto SI, Coll JM. Trout oral VP2 DNA vaccination mimics transcriptional responses occurring after infection with infectious pancreatic necrosis virus (IPNV). FISH & SHELLFISH IMMUNOLOGY 2012; 33:1249-57. [PMID: 23041507 DOI: 10.1016/j.fsi.2012.09.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2012] [Revised: 08/26/2012] [Accepted: 09/04/2012] [Indexed: 05/12/2023]
Abstract
Time-course and organ transcriptional response profiles in rainbow trout Oncorhynchus mykiss were studied after oral DNA-vaccination with the VP2 gene of the infectious pancreatic necrosis virus (IPNV) encapsulated in alginates. The profiles were also compared with those obtained after infection with IPNV. A group of immune-related genes (stat1, ifn1, ifng, mx1, mx3, il8, il10, il11, il12b, tnf2, mhc1uda, igm and igt) previously selected from microarray analysis of successful oral vaccination of rainbow trout, were used for the RTqPCR analysis. The results showed that oral VP2-vaccination qualitatively mimicked both the time-course and organ (head kidney, spleen, intestine, pyloric ceca, and thymus) transcriptional profiles obtained after IPNV-infection. Highest transcriptional differential expression levels after oral vaccination were obtained in thymus, suggesting those might be important for subsequent protection against IPNV challenges. However, transcriptional differential expression levels of most of the genes mentioned above were lower in VP2-vaccinated than in IPNV-infected trout, except for ifn1 which were similar. Together all the results suggest that the oral-alginate VP2-vaccination procedure immunizes trout against IPNV in a similar way as IPNV-infection does while there is still room for additional improvements in the oral vaccination procedure. Some of the genes described here could be used as markers to further optimize the oral immunization method.
Collapse
Affiliation(s)
- Natalia A Ballesteros
- Centro de Investigaciones Biológicas-CSIC, Dpto. Microbiología Molecular y Biología de la Infección, c/Ramiro de Maeztu 9, 28040 Madrid, Spain
| | | | | | | |
Collapse
|
20
|
Wang NX, Bazdar DA, Sieg SF, von Recum HA. Microparticle delivery of Interleukin-7 to boost T-cell proliferation and survival. Biotechnol Bioeng 2012; 109:1835-43. [DOI: 10.1002/bit.24431] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2011] [Revised: 12/14/2011] [Accepted: 12/27/2011] [Indexed: 11/10/2022]
|
21
|
Abstract
Advances in the engineering of peptides, adjuvants and delivery systems have renewed the enthusiasm for peptide-based vaccination regimens in the setting of cancer, and there are a variety of clinical trials being conducted by pharmaceutical companies based on the use of peptides. The challenges to successful cancer immunotherapy are common to all immunotherapeutic strategies and not unique to peptide-based vaccination regimens. This review will describe the advances in the identification, design and delivery of peptides, the challenges to successful immunotherapy and will discuss potential options for the future.
Collapse
Affiliation(s)
- Shreya Kanodia
- Department of Molecular Microbiology and Immunology, Norris Comprehensive Cancer Center, NRT 7517, University of Southern California, 1450 Biggy Street, Los Angeles, CA 90033, USA.
| | | |
Collapse
|
22
|
Secombes C. Will advances in fish immunology change vaccination strategies? FISH & SHELLFISH IMMUNOLOGY 2008; 25:409-416. [PMID: 18562212 DOI: 10.1016/j.fsi.2008.05.001] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2008] [Revised: 05/01/2008] [Accepted: 05/07/2008] [Indexed: 05/26/2023]
Abstract
This review will discuss some of the recent advances in discovering immune genes in fish, in terms of their relevance to vaccine design and development. Particular emphasis will be placed on the many cytokine and costimulatory molecules now known, with examples drawn from the mammalian literature as to their potential value for fish vaccinology. A new area of vaccine research will also be touched upon, where efficacious responses are elicited by inhibiting the natural negative regulators of immune responses, such as Treg cell products and SOCS proteins.
Collapse
Affiliation(s)
- Chris Secombes
- Scottish Fish Immunology Research Centre, University of Aberdeen, Tillydrone Avenue, Aberdeen AB24 2TZ, United Kingdom.
| |
Collapse
|
23
|
Jalah R, Rosati M, Kulkarni V, Patel V, Bergamaschi C, Valentin A, Zhang GM, Sidhu MK, Eldridge JH, Weiner DB, Pavlakis GN, Felber BK. Efficient systemic expression of bioactive IL-15 in mice upon delivery of optimized DNA expression plasmids. DNA Cell Biol 2008; 26:827-40. [PMID: 17979522 DOI: 10.1089/dna.2007.0645] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Efficient expression vectors for interleukin 15 (IL-15) were developed combining RNA/codon optimization and modification of the IL-15 native long signal peptide. These changes resulted in elevated cytoplasmic levels of the optimized mRNA and more than 100-fold improved production of secreted human IL-15 protein. Similar modifications have also led to greatly increased rhesus macaque and murine IL-15 production. Comparison of different heterologous secretory signals showed that the tissue plasminogen activator signal is most efficient for the production of extracellular IL-15. Upon intramuscular injection of the fully optimized expression vectors in mice, IL-15 was readily detected in the serum. Serum levels represented <1% of intramuscular IL-15 and were sufficient in causing some systemic effects, such as increasing the frequency of natural killer (NK) cells in the liver. Upon hydrodynamic DNA delivery in mice, very high levels of IL-15 were produced, which increased the frequency of NK cells in liver as well as in spleen and lung. These optimized expression vectors have potential applications in vaccine and immunotherapy approaches against AIDS and cancer.
Collapse
Affiliation(s)
- Rashmi Jalah
- Human Retrovirus Pathogenesis Section, Vaccine Branch, National Cancer Institute-Frederick, Frederick, Maryland 21702-1201, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|