1
|
Zhang M, Chen W, Ju Y, Zhao H, Wang C. Polymer-Protein Nanovaccine Synthesized via Reactive Self-Assembly with Potential Application in Cancer Immunotherapy: Physicochemical and Biological Characterization In Vitro and In Vivo. Macromol Rapid Commun 2023; 44:e2300438. [PMID: 37708966 DOI: 10.1002/marc.202300438] [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: 07/20/2023] [Revised: 09/10/2023] [Indexed: 09/16/2023]
Abstract
Nanovaccines composed of polymeric nanocarriers and protein-based antigens have attracted much attention in recent years because of their enormous potential in the prevention and treatment of diseases such as viral infections and cancer. While surface-conjugated protein antigens are known to be more immunoactive than encapsulated antigens, current surface conjugation methods often result in low and insufficient protein loading. Herein, reactive self-assembly is used to prepare nanovaccine from poly(ε-caprolactone) (PCL) and ovalbumin (OVA)-a model antigen. A rapid thiol-disulfide exchange reaction between PCL with pendant pyridyl disulfide groups and thiolated OVA results in the formation of nanoparticles with narrow size distribution. High OVA loading (≈70-80 wt%) is achieved, and the native secondary structure of OVA is preserved. Compared to free OVA, the nanovaccine is much superior in enhancing antigen uptake by bone marrow-derived dendritic cells (BMDCs), promoting BMDC maturation and antigen presentation via the MHC I pathway, persisting at the injection site and draining lymph nodes, activating both Th1 and Th2 T cell immunity, and ultimately, resisting tumor challenge in mice. This is the first demonstration of reactive self-assembly for the construction of a polymer-protein nanovaccine with clear potential in advancing cancer immunotherapy.
Collapse
Affiliation(s)
- Mingming Zhang
- Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
| | - Wenjuan Chen
- Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
| | - Yuanyuan Ju
- Key Laboratory of Functional Polymer Materials, Ministry of Education, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Hanying Zhao
- Key Laboratory of Functional Polymer Materials, Ministry of Education, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Chun Wang
- Department of Biomedical Engineering, University of Minnesota, 7-105 Hasselmo Hall, 312 Church Street S. E., Minneapolis, MN, 55455, USA
| |
Collapse
|
2
|
Elashiry MM, Bergeron BE, Tay FR. Enterococcus faecalis in secondary apical periodontitis: Mechanisms of bacterial survival and disease persistence. Microb Pathog 2023; 183:106337. [PMID: 37683835 DOI: 10.1016/j.micpath.2023.106337] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/03/2023] [Accepted: 09/05/2023] [Indexed: 09/10/2023]
Abstract
Enterococcus faecalis is a commensal bacterium commonly found in the human gastrointestinal tract. However, in individuals with compromised immune systems, the pathogen can lead to severe illness. This opportunistic pathogen is associated with secondary apical diseases and is adept at resisting antibiotics and other forms of treatment because of its numerous virulence factors. Enterococcus faecalis is capable of disrupting the normal functions of immune cells, thereby hindering the body's ability to eradicate the infection. However, intensive research is needed in further understanding the adverse immunomodulatory effects of E. faecalis. Potential strategies specific for eradicating E. faecalis have proven beneficial in the treatment of persistent secondary apical periodontitis.
Collapse
Affiliation(s)
- Mohamed M Elashiry
- Department of Endodontics, Dental College of Georgia, Augusta University, Georgia, USA; Department of Endodontics, Faculty of Dentistry, Ain Shams University, Cairo, Egypt.
| | - Brian E Bergeron
- Department of Endodontics, Dental College of Georgia, Augusta University, Georgia, USA
| | - Franklin R Tay
- Department of Endodontics, Dental College of Georgia, Augusta University, Georgia, USA
| |
Collapse
|
3
|
Nyariki JN, Kimani NM, Kibet PS, Kinuthia GK, Isaac AO. Coenzyme Q10 exhibits anti-inflammatory and immune-modulatory thereby decelerating the occurrence of experimental cerebral malaria. Mol Biochem Parasitol 2023; 255:111579. [PMID: 37385350 DOI: 10.1016/j.molbiopara.2023.111579] [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: 05/14/2023] [Revised: 06/26/2023] [Accepted: 06/26/2023] [Indexed: 07/01/2023]
Abstract
Cerebral Malaria (CM) is associated with the complex neurological syndrome, whose pathology is mediated by severe inflammatory processes following infection with Plasmodium falciparum. Coenzyme-Q10 (Co-Q10) is a potent anti-inflammatory, anti-oxidant, and anti-apoptotic agent with numerous clinical applications. The aim of this study was to elucidate the role of oral administration of Co-Q10 on the initiation or regulation of inflammatory immune response during experimental cerebral malaria (ECM). For this purpose, the pre-clinical effect of Co-Q10 was evaluated in C57BL/6 J mice infected with Plasmodium berghei ANKA (PbA). Treatment with Co-Q10 resulted in the reduction of infiltrating parasite load, greatly improved the survival rate of PbA-infected mice that occurred independent of parasitaemia and prevented PbA-induced disruption of the blood-brain barrier (BBB) integrity. Exposure to Co-Q10 resulted in the reduction of infiltration of effector CD8 + T cells in the brain and secretion of cytolytic Granzyme B molecules. Notably, Co-Q10-treated mice had reduced levels of CD8 +T cell chemokines CXCR3, CCR2, and CCR5 in the brain following PbA-infection. Brain tissue analysis showed a reduction in the levels of inflammatory mediators TNF- α, CCL3, and RANTES in Co-Q10 administered mice. In addition, Co-Q10 modulated the differentiation and maturation of both splenic and brain dendritic cells and cross-presentation (CD8α+DCs) during ECM. Remarkably, Co-Q10 was very effective in decreasing levels of CD86, MHC-II, and CD40 in macrophages associated with ECM pathology. Exposure to Co-Q10 resulted in increased expression levels of Arginase-1 and Ym1/chitinase 3-like 3, which is linked to ECM protection. Furthermore, Co-Q10 supplementation prevented PbA-induced depletion of Arginase and CD206 mannose receptor levels. Co-Q10 abrogated PbA-driven elevation in pro-inflammatory cytokines IL-1β, IL-18, and IL-6 levels. In conclusion, the oral supplementation with Co-Q10 decelerates the occurrence of ECM by preventing lethal inflammatory immune responses and dampening genes associated with inflammation and immune-pathology during ECM, and offers an inimitable opening for developing an anti-inflammatory agent against cerebral malaria.
Collapse
Affiliation(s)
- James Nyabuga Nyariki
- Department of Biochemistry and Biotechnology, Technical of University of Kenya, P.O Box 52428-00200 Nairobi, Kenya.
| | - Njogu M Kimani
- Department of Physical Sciences, University of Embu, P.O Box 6-60100 Embu, Kenya
| | - Peter Shikuku Kibet
- Department of Pathology, Hematology and Blood Transfusion thematic unit, University of Nairobi, PO Box 30197-00100, Nairobi, Kenya
| | - Geoffrey K Kinuthia
- Department of Science & Public Health, Daystar University, PO Box 44400-00100, Nairobi, Kenya
| | - Alfred Orina Isaac
- Department of Pharmaceutical Sciences and Technology, School Health Sciences and Biomedical Sciences, Technical University of Kenya, P.O Box 52428-00200 Nairobi, Kenya
| |
Collapse
|
4
|
Villafán H, Gutiérrez-Ospina G. Looking beyond Self-Protection: The Eyes Instruct Systemic Immune Tolerance Early in Life. Brain Sci 2023; 13:1261. [PMID: 37759864 PMCID: PMC10526493 DOI: 10.3390/brainsci13091261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 08/15/2023] [Accepted: 08/18/2023] [Indexed: 09/29/2023] Open
Abstract
The eyes provide themselves with immune tolerance. Frequent skin inflammatory diseases in young blind people suggest, nonetheless, that the eyes instruct a systemic immune tolerance that benefits the whole body. We tested this premise by using delayed skin contact hypersensitivity (DSCH) as a tool to compare the inflammatory response developed by sighted (S) and birth-enucleated (BE) mice against oxazolone or dinitrofluorobenzene at the ages of 10, 30 and 60 days of life. Adult mice enucleated (AE) at 60 days of age were also assessed when they reached 120 days of life. BE mice displayed exacerbated DSCH at 60 but not at 10 or 30 days of age. AE mice, in contrast, show no exacerbated DSCH. Skin inflammation in 60-day-old BE mice was hapten exclusive and supported by distinct CD8+ lymphocytes. The number of intraepidermal T lymphocytes and migrating Langerhans cells was, however, similar between S and BE mice by the age of 60 days. Our observations support the idea that the eyes instruct systemic immune tolerance that benefits organs outside the eyes from an early age. The higher prevalence of inflammatory skin disorders reported in young people might then reflect reduced immune tolerance associated with the impaired functional morphology of the eyes.
Collapse
Affiliation(s)
- Horacio Villafán
- Programa de Doctorado en Ciencias Biológicas, Unidad de Posgrado, Circuito de Posgrados, Ciudad Universitaria, Edificio D, 1piso, Coyoacán, Ciudad de México 04510, Mexico
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Gabriel Gutiérrez-Ospina
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
- Coordinación de Psicobiología y Neurociencias, Facultad de Psicología, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
- Department of Zoology and Physiology and Wyoming Sensory Biology Center of Biomedical Research Excellence, University of Wyoming, Laramie, WY 82071, USA
| |
Collapse
|
5
|
Xu H, Zhu S, Govinden R, Chenia HY. Multiple Vaccines and Strategies for Pandemic Preparedness of Avian Influenza Virus. Viruses 2023; 15:1694. [PMID: 37632036 PMCID: PMC10459121 DOI: 10.3390/v15081694] [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: 06/26/2023] [Revised: 07/14/2023] [Accepted: 07/27/2023] [Indexed: 08/27/2023] Open
Abstract
Avian influenza viruses (AIV) are a continuous cause of concern due to their pandemic potential and devasting effects on poultry, birds, and human health. The low pathogenic avian influenza virus has the potential to evolve into a highly pathogenic avian influenza virus, resulting in its rapid spread and significant outbreaks in poultry. Over the years, a wide array of traditional and novel strategies has been implemented to prevent the transmission of AIV in poultry. Mass vaccination is still an economical and effective approach to establish immune protection against clinical virus infection. At present, some AIV vaccines have been licensed for large-scale production and use in the poultry industry; however, other new types of AIV vaccines are currently under research and development. In this review, we assess the recent progress surrounding the various types of AIV vaccines, which are based on the classical and next-generation platforms. Additionally, the delivery systems for nucleic acid vaccines are discussed, since these vaccines have attracted significant attention following their significant role in the fight against COVID-19. We also provide a general introduction to the dendritic targeting strategy, which can be used to enhance the immune efficiency of AIV vaccines. This review may be beneficial for the avian influenza research community, providing ideas for the design and development of new AIV vaccines.
Collapse
Affiliation(s)
- Hai Xu
- Jiangsu Key Laboratory for High-Tech Research and Development of Veterinary Biopharmaceuticals, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou 225300, China;
- Discipline of Microbiology, School of Life Sciences, College of Agriculture, Engineering and Science, University of KwaZulu-Natal, Durban 4001, South Africa;
| | - Shanyuan Zhu
- Jiangsu Key Laboratory for High-Tech Research and Development of Veterinary Biopharmaceuticals, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou 225300, China;
| | - Roshini Govinden
- Discipline of Microbiology, School of Life Sciences, College of Agriculture, Engineering and Science, University of KwaZulu-Natal, Durban 4001, South Africa;
| | - Hafizah Y. Chenia
- Discipline of Microbiology, School of Life Sciences, College of Agriculture, Engineering and Science, University of KwaZulu-Natal, Durban 4001, South Africa;
| |
Collapse
|
6
|
Lee W, Suresh M. Vaccine adjuvants to engage the cross-presentation pathway. Front Immunol 2022; 13:940047. [PMID: 35979365 PMCID: PMC9376467 DOI: 10.3389/fimmu.2022.940047] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 07/08/2022] [Indexed: 11/13/2022] Open
Abstract
Adjuvants are indispensable components of vaccines for stimulating optimal immune responses to non-replicating, inactivated and subunit antigens. Eliciting balanced humoral and T cell-mediated immunity is paramount to defend against diseases caused by complex intracellular pathogens, such as tuberculosis, malaria, and AIDS. However, currently used vaccines elicit strong antibody responses, but poorly stimulate CD8 cytotoxic T lymphocyte (CTL) responses. To elicit potent CTL memory, vaccines need to engage the cross-presentation pathway, and this requirement has been a crucial bottleneck in the development of subunit vaccines that engender effective T cell immunity. In this review, we focus on recent insights into DC cross-presentation and the extent to which clinically relevant vaccine adjuvants, such as aluminum-based nanoparticles, water-in oil emulsion (MF59) adjuvants, saponin-based adjuvants, and Toll-like receptor (TLR) ligands modulate DC cross-presentation efficiency. Further, we discuss the feasibility of using carbomer-based adjuvants as next generation of adjuvant platforms to elicit balanced antibody- and T-cell based immunity. Understanding of the molecular mechanism of DC cross-presentation and the mode of action of adjuvants will pave the way for rational design of vaccines for infectious diseases and cancer that require balanced antibody- and T cell-based immunity.
Collapse
|
7
|
Parra-Medina R, Rocha F, Castañeda-González JP, Moreno-Lucero P, Veloza L, Romero-Rojas AE. Synchronous or collision solid neoplasms and lymphomas: A systematic review of 308 case reports. Medicine (Baltimore) 2022; 101:e28988. [PMID: 35838994 PMCID: PMC11132339 DOI: 10.1097/md.0000000000028988] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 02/13/2022] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND The presence of a lymphoma associated with a solid synchronous neoplasm or collision neoplasm has been rarely in the literature, and a detailed characterization of these cases is lacking to date. OBJECTIVE To describe the main clinicopathological features of synchronous/collision tumors. METHODS A systematic search in PubMed, Scielo, and Virtual Health Library literature databases for cases or case series of synchronous or collision lymphoma and other solid neoplasms reported up to March 2021 was performed. Three reviewers independently screened the literature, extracted data, and assessed the quality of the included studies. The systematic review was performed following the Preferred Reporting Items for Systematic Meta-Analyses guidelines. RESULTS Mean age of patients was 62.9 years (52.9% men). A total of 308 cases were included (62% synchronous and 38% collision). The most frequent location of both synchronous and collision tumors was the gastrointestinal tract with the most common solid neoplasm being adenocarcinoma, and the most frequent lymphoma diffuse large B-cell lymphoma (21.7%) and mucosa-associated lymphoid tissue lymphoma (20.4%). Of the total number of mucosa-associated lymphoid tissue lymphomas and gastric adenocarcinomas, the presence of Helicobacter pylori infection was documented in 47.3% of them. Only 2% of all cases had a previous history of lymphoma. Thus, in most cases (98%), lymphoma was discovery incidentally. In addition, nodal lymphoma was associated with metastasis in 29 (9.4%) cases as collision tumor, most commonly (90%) in locoregional lymph nodes of the solid neoplasm. CONCLUSIONS The frequent association of some type of B-cell lymphoma and adenocarcinoma in synchronous/collision tumors of the gastrointestinal tract points to common pathogenic mechanisms in both neoplasia, particularly related to chronic inflammation in this location. In most cases, lymphoma identified in locoregional lymph nodes or distant of a carcinoma seems to represent an incidental finding during the carcinoma diagnostic/therapeutic approach. A synergy between carcinoma and lymphoma (involving inflammation and immunosuppression mechanisms) may favor tumor progression and dissemination. A better understating of the interactions lymphoma/carcinoma in the setting of synchronous/collision tumors may help to improve patient management and prognosis.
Collapse
Affiliation(s)
- Rafael Parra-Medina
- Department of Pathology, Fundación Universitaria de Ciencias de la Salud, Hospital San José, Bogotá, Colombia
- Research Institute, Fundación Universtaria de Ciencias de la Salud, Bogotá, Colombia
- Department of Pathology, Instituto Nacional de Cancerología, Bogotá, Colombia
| | - Franky Rocha
- Department of Pathology, Fundación Universitaria de Ciencias de la Salud, Hospital San José, Bogotá, Colombia
| | | | - Paula Moreno-Lucero
- Department of Pathology, Fundación Universitaria de Ciencias de la Salud, Hospital San José, Bogotá, Colombia
| | - Luis Veloza
- Institute of Pathology, Lausanne University Hospital, Lausanne University, Lausanne, Switzerland
| | | |
Collapse
|
8
|
Roquilly A, Mintern JD, Villadangos JA. Spatiotemporal Adaptations of Macrophage and Dendritic Cell Development and Function. Annu Rev Immunol 2022; 40:525-557. [PMID: 35130030 DOI: 10.1146/annurev-immunol-101320-031931] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Macrophages and conventional dendritic cells (cDCs) are distributed throughout the body, maintaining tissue homeostasis and tolerance to self and orchestrating innate and adaptive immunity against infection and cancer. As they complement each other, it is important to understand how they cooperate and the mechanisms that integrate their functions. Both are exposed to commensal microbes, pathogens, and other environmental challenges that differ widely among anatomical locations and over time. To adjust to these varying conditions, macrophages and cDCs acquire spatiotemporal adaptations (STAs) at different stages of their life cycle that determine how they respond to infection. The STAs acquired in response to previous infections can result in increased responsiveness to infection, termed training, or in reduced responses, termed paralysis, which in extreme cases can cause immunosuppression. Understanding the developmental stage and location where macrophages and cDCs acquire their STAs, and the molecular and cellular players involved in their induction, may afford opportunities to harness their beneficial outcomes and avoid or reverse their deleterious effects. Here we review our current understanding of macrophage and cDC development, life cycle, function, and STA acquisition before, during, and after infection. We propose a unified framework to explain how these two cell types adjust their activities to changing conditions over space and time to coordinate their immunosurveillance functions. Expected final online publication date for the Annual Review of Immunology, Volume 40 is April 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Antoine Roquilly
- Center for Research in Transplantation and Translational Immunology, INSERM, UMR 1064, CHU Nantes, University of Nantes, Nantes, France
| | - Justine D Mintern
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
| | - Jose A Villadangos
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia.,Department of Microbiology and Immunology, Doherty Institute of Infection and Immunity, The University of Melbourne, Parkville, Victoria, Australia;
| |
Collapse
|
9
|
Biologically active lipids in the regulation of lymphangiogenesis in disease states. Pharmacol Ther 2021; 232:108011. [PMID: 34614423 DOI: 10.1016/j.pharmthera.2021.108011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 07/31/2021] [Accepted: 09/01/2021] [Indexed: 02/06/2023]
Abstract
Lymphatic vessels have crucial roles in the regulation of interstitial fluids, immune surveillance, and the absorption of dietary fat in the intestine. Lymphatic function is also closely related to the pathogenesis of various disease states such as inflammation, lymphedema, endometriosis, liver dysfunction, and tumor metastasis. Lymphangiogenesis, the formation of new lymphatic vessels from pre-existing lymphatic vessels, is a critical determinant in the above conditions. Although the effect of growth factors on lymphangiogenesis is well-characterized, and biologically active lipids are known to affect smooth muscle contractility and vasoaction, there is accumulating evidence that biologically active lipids are also important inducers of growth factors and cytokines that regulate lymphangiogenesis. This review discusses recent advances in our understanding of biologically active lipids, including arachidonic acid metabolites, sphingosine 1-phosphate, and lysophosphatidic acid, as regulators of lymphangiogenesis, and the emerging importance of the lymphangiogenesis as a therapeutic target.
Collapse
|
10
|
Wang F, Ullah A, Fan X, Xu Z, Zong R, Wang X, Chen G. Delivery of nanoparticle antigens to antigen-presenting cells: from extracellular specific targeting to intracellular responsive presentation. J Control Release 2021; 333:107-128. [PMID: 33774119 DOI: 10.1016/j.jconrel.2021.03.027] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 03/19/2021] [Accepted: 03/22/2021] [Indexed: 02/05/2023]
Abstract
An appropriate delivery system can improve the immune effects of antigens against various infections or tumors. Antigen-presenting cells (APCs) are specialized to capture and process antigens in vivo, which link the innate and adaptive immune responses. Functionalization of vaccine delivery systems with targeting moieties to APCs is a promising strategy for provoking potent immune responses. Additionally, the internalization and intracellular distribution of antigens are closely related to the initiation of downstream immune responses. With a deeper understanding of the intracellular microenvironment and the mechanisms of antigen presentation, vehicles designed to respond to endogenous and external stimuli can modulate antigen processing and presentation pathways, which are critical to the types of immune response. Here, an overview of extracellular targeting delivery of antigens to APCs and intracellular stimulus-responsiveness strategies is provided, which might be helpful for the rational design of vaccine delivery systems.
Collapse
Affiliation(s)
- Fei Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; Institute of Comparative Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Aftab Ullah
- Shantou University Medical College, Shantou 515041, China
| | - Xuelian Fan
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; Institute of Comparative Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Zhou Xu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; Institute of Comparative Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Rongling Zong
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; Institute of Comparative Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Xuewen Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; Institute of Comparative Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Gang Chen
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; Institute of Comparative Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China.
| |
Collapse
|
11
|
Pinto A, Rega A, Crother TR, Sorrentino R. Plasmacytoid dendritic cells and their therapeutic activity in cancer. Oncoimmunology 2021; 1:726-734. [PMID: 22934264 PMCID: PMC3429576 DOI: 10.4161/onci.20171] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
In the last decade several studies provided evidence that plasmacytoid dendritic cells (pDCs) infiltrate human neoplasms with poor prognosis. However, the role of tumor-associated pDCs remains controversial. Various studies indicate that pDCs play an immuno-suppressive role and facilitate tumor progression in both animal models and humans. In contrast, others found that the presence of activated tumor-associated pDCs results in tumor regression in mice. Given these findings, understanding pDC function in tumor biology is an important necessity and may pave the way for novel therapeutic strategies to fight malignancies.
Collapse
Affiliation(s)
- Aldo Pinto
- Pharmaceutical and Biomedical Sciences Department (FARMABIOMED); University of Salerno; Fisciano, Italy
| | | | | | | |
Collapse
|
12
|
Lycium barbarum Polysaccharides Promote Maturity of Murine Dendritic Cells through Toll-Like Receptor 4-Erk1/2-Blimp1 Signaling Pathway. J Immunol Res 2020; 2020:1751793. [PMID: 33344654 PMCID: PMC7725586 DOI: 10.1155/2020/1751793] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 10/11/2020] [Accepted: 10/30/2020] [Indexed: 12/04/2022] Open
Abstract
In previous studies, Lycium barbarum polysaccharides (LBP), a traditional Chinese medicine, can promote immature dendritic cells (DCs) to mature. However, the molecular mechanisms by which LBP works are not yet elucidated. Here, we found that LBP can induce DCs maturation, which is mainly characterized by the upregulation of MHCII and costimulatory molecules (CD80, CD86), and increase the production of IL-6 and IL-4. Furthermore, we found that LBP could increase the mRNA and protein expression of TLR4, p38, Erk1/2, JNK, and Blimp1 signal molecules. More interestingly, after blocking by Toll-like receptor 4 inhibitor, Resatorvid (TAK 242), the mRNA and protein expression of TLR4, Erk1/2, and Blimp1 was significantly decreased while the expression of p38 and JNK has not changed. Then, we found that after blocking by p38 inhibitor (SB203580), Erk inhibitor (PD98059), and JNK inhibitor (SP603580) separately, Blimp1 protein expression was significantly reduced; after downregulating Blimp1 by Blimp1-siRNA, the production of IL-6 was reduced. In conclusion, our results indicate that LBP can induce maturation of DCs through the TLR4-Erk1/2-Blimp1 signal pathway instead of the JNK/p38-Blimp1 pathway. Our findings may provide a novel evidence for understanding the molecular mechanisms of LBP on activating murine DCs.
Collapse
|
13
|
DC-targeted gold nanoparticles as an efficient and biocompatible carrier for modulating allergic responses in sublingual immunotherapy. Int Immunopharmacol 2020; 86:106690. [PMID: 32585607 DOI: 10.1016/j.intimp.2020.106690] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 06/06/2020] [Accepted: 06/06/2020] [Indexed: 01/13/2023]
Abstract
BACKGROUND Sublingual immunotherapy (SLIT) was introduced to deliver allergens in an effective and non-invasive route, which can be considered as an alternative for allergen-specific subcutaneous immunotherapy (SCIT). On the other hand, the use of gold nanoparticles (AuNPs) in allergen delivery has beneficial effects on sublingual immunotherapy. In addition, the molecular targeting agents like aptamers (Apt), have been widely applied for targeted drug delivery. Therefore, the current study aimed to evaluate the effects of dendritic cells (DCs)-specific Aptamer-modified AuNPs coated with ovalbumin (OVA) on the improvement of the SLIT outcome in the mouse model of allergy. MATERIAL AND METHODS AuNPs with approximately 15 nm diameter were prepared by citrate reduction of HAuCl4. Afterward, Apt-modified AuNP complex was prepared and OVA was then loaded onto this complex. Following sensitization of Balb/c mice to OVA, SLIT was performed with Apt-AuNPs containing 5 µg OVA twice a week for a 2-month period. Allergen-specific IgE in serum, as well as cytokines secretion of spleen cells, were analyzed using ELISA. Also, nasopharyngeal lavage Fluid (NALF) was collected for total and eosinophil counts. Moreover, the lungs were removed for histopathological examination. RESULTS SLIT with Apt-modified AuNPs complex containing 5 μg OVA, decreased the IgE levels compared to the other groups. Also, IL-4 production has significantly decreased in spleen cells, while TGF-β and IFN-γ have significantly increased. The assessment of NALF in the group treated by this complex showed a decrease in total cell as well as in eosinophil count. Also, the examination of lung tissues revealed that, in the group treated by this complex, inflammation and perivascular infiltration were lesser than the other groups, which were observed in only one vessel of tissue. CONCLUSION It was shown that, Sublingual immunotherapy with DC specific Apt-modified AuNPs containing 5 μg OVA can improve the Th1 and Treg immunomodulatory responses.
Collapse
|
14
|
McDaniel MM, Kottyan LC, Singh H, Pasare C. Suppression of Inflammasome Activation by IRF8 and IRF4 in cDCs Is Critical for T Cell Priming. Cell Rep 2020; 31:107604. [PMID: 32375053 PMCID: PMC7325595 DOI: 10.1016/j.celrep.2020.107604] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 02/13/2020] [Accepted: 04/10/2020] [Indexed: 01/10/2023] Open
Abstract
Inflammasome activation leads to pyroptotic cell death, thereby eliminating the replicative niche of virulent pathogens. Although inflammasome-associated cytokines IL-1β and IL-18 have an established role in T cell function, whether inflammasome activation in dendritic cells (DCs) is critical for T cell priming is not clear. Here, we find that conventional DCs (cDCs) suppress inflammasome activation to prevent pyroptotic cell death, thus preserving their ability to prime both CD4 and CD8 T cells. Transcription factors IRF8 and IRF4, in cDC1s and cDC2s, respectively, mediate suppression of inflammasome activation by limiting the expression of inflammasome-associated genes. Overexpression of IRF4 or IRF8 inhibits inflammasome activation in macrophages, while reduced expression of IRF8 leads to aberrant inflammasome activation in cDC1s and hampers their ability to prime CD8 T cells. Thus, activation of inflammasome in DCs is detrimental to adaptive immunity, and our results reveal that cDCs use IRF4 and IRF8 to suppress this response.
Collapse
Affiliation(s)
- Margaret M McDaniel
- Immunology Graduate Program, The University of Texas Southwestern Medical Center, Dallas, TX, USA; Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Leah C Kottyan
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, OH, USA
| | - Harinder Singh
- Center for Systems Immunology, Departments of Immunology and Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Chandrashekhar Pasare
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
15
|
Toki MI, Kumar D, Ahmed FS, Rimm DL, Xu ML. Benign lymph node microenvironment is associated with response to immunotherapy. PRECISION CLINICAL MEDICINE 2020; 3:44-53. [PMID: 35693430 PMCID: PMC8985791 DOI: 10.1093/pcmedi/pbaa003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 02/03/2020] [Accepted: 02/09/2020] [Indexed: 11/12/2022] Open
Abstract
Introduction Benign lymph nodes have been considered the hubs of immune surveillance in cancer patients. The microenvironment of these lymphoid tissues can be immune suppressed, hence allowing for tumor progression. Understanding the spectrum of benign findings in bystander lymph nodes in immune checkpoint blockade therapy could prove to be key to understanding the mechanism and assessing treatment response. Methods Benign lymph nodes and spleen were evaluated from patients treated with immunotherapy who subsequently received postmortem examination. We used quantitative immunofluorescence (QIF) to assess tumor infiltrating lymphocytes (TIL) and macrophage marker expression and characterized activation status using a novel multiplexed QIF assay including CD3, GranzymeB, and Ki67. We performed immunohistochemistry to correlate results of QIF. Results Benign lymph nodes from non-responders to immunotherapy showed significantly higher expression of cytotoxic markers and proliferation index (Ki67) in T cells compared to responders. Higher expression of PD-L1 in macrophages was also observed. There was no significant difference in CD3+ expression, but higher levels of CD8+ T cells as well as CD20+ B cells were seen in lymph nodes of non-responders. No significant differences were seen between responder and non-responder splenic tissue. Findings were supported by traditional immunostaining methods. Conclusions While most studies in biomarkers for immunotherapy focus on tumor microenvironment, we show that benign lymph node microenvironment may predict response to immunotherapy. In responding patients, bystander lymph nodes appear to have been mobilized, resulting in reduced cytotoxic T cells. Conversely, patients whose disease progressed on immunotherapy demonstrate higher levels of macrophages that express increased PD-L1, and activated T cells not recruited to the tumor site.
Collapse
Affiliation(s)
- Maria I Toki
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Deepika Kumar
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Fahad S Ahmed
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - David L Rimm
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Medicine, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Mina L Xu
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06510, USA
| |
Collapse
|
16
|
Kim CG, Kye YC, Yun CH. The Role of Nanovaccine in Cross-Presentation of Antigen-Presenting Cells for the Activation of CD8 + T Cell Responses. Pharmaceutics 2019; 11:E612. [PMID: 31731667 PMCID: PMC6920862 DOI: 10.3390/pharmaceutics11110612] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 11/12/2019] [Accepted: 11/12/2019] [Indexed: 01/30/2023] Open
Abstract
Explosive growth in nanotechnology has merged with vaccine development in the battle against diseases caused by bacterial or viral infections and malignant tumors. Due to physicochemical characteristics including size, viscosity, density and electrostatic properties, nanomaterials have been applied to various vaccination strategies. Nanovaccines, as they are called, have been the subject of many studies, including review papers from a material science point of view, although a mode of action based on a biological and immunological understanding has yet to emerge. In this review, we discuss nanovaccines in terms of CD8+ T cell responses, which are essential for antiviral and anticancer therapies. We focus mainly on the role and mechanism, with particular attention to the functional aspects, of nanovaccines in inducing cross-presentation, an unconventional type of antigen-presentation that activates CD8+ T cells upon administration of exogenous antigens, in dendritic cells followed by activation of antigen-specific CD8+ T cell responses. Two major intracellular mechanisms that nanovaccines harness for cross-presentation are described; one is endosomal swelling and rupture, and the other is membrane fusion. Both processes eventually allow exogenous vaccine antigens to be exported from phagosomes to the cytosol followed by loading on major histocompatibility complex class I, triggering clonal expansion of CD8+ T cells. Advancement of nanotechnology with an enhanced understanding of how nanovaccines work will contribute to the design of more effective and safer nanovaccines.
Collapse
Affiliation(s)
- Cheol Gyun Kim
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea; (C.G.K.); (Y.-C.K.)
| | - Yoon-Chul Kye
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea; (C.G.K.); (Y.-C.K.)
| | - Cheol-Heui Yun
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea; (C.G.K.); (Y.-C.K.)
- Center for Food and Bioconvergence, Seoul National University, Seoul 08826, Korea
- Institute of Green Bio Science and Technology, Seoul National University, Pyeongchang, Gangwon-do 25354, Korea
| |
Collapse
|
17
|
Silva‐Costa LC, Garcia‐Rosa S, Smith BJ, Baldasso PA, Steiner J, Martins‐de‐Souza D. Blood plasma high abundant protein depletion unintentionally carries over 100 proteins. SEPARATION SCIENCE PLUS 2019. [DOI: 10.1002/sscp.201900057] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
- Licia C. Silva‐Costa
- Laboratory of NeuroproteomicsInstitute of BiologyDepartment of Biochemistry and Tissue BiologyUniversity of Campinas (UNICAMP) Campinas Brazil
| | - Sheila Garcia‐Rosa
- Laboratory of NeuroproteomicsInstitute of BiologyDepartment of Biochemistry and Tissue BiologyUniversity of Campinas (UNICAMP) Campinas Brazil
| | - Bradley J. Smith
- Laboratory of NeuroproteomicsInstitute of BiologyDepartment of Biochemistry and Tissue BiologyUniversity of Campinas (UNICAMP) Campinas Brazil
| | - Paulo A. Baldasso
- Laboratory of NeuroproteomicsInstitute of BiologyDepartment of Biochemistry and Tissue BiologyUniversity of Campinas (UNICAMP) Campinas Brazil
| | - Johann Steiner
- Department of PsychiatryUniversity of Magdeburg Magdeburg Germany
| | - Daniel Martins‐de‐Souza
- Laboratory of NeuroproteomicsInstitute of BiologyDepartment of Biochemistry and Tissue BiologyUniversity of Campinas (UNICAMP) Campinas Brazil
- Instituto Nacional de Biomarcadores em Neuropsiquiatria (INBION)Conselho Nacional de Desenvolvimento Científico e Tecnológico São Paulo Brazil
| |
Collapse
|
18
|
Solano-Gálvez SG, Tovar-Torres SM, Tron-Gómez MS, Weiser-Smeke AE, Álvarez-Hernández DA, Franyuti-Kelly GA, Tapia-Moreno M, Ibarra A, Gutiérrez-Kobeh L, Vázquez-López R. Human Dendritic Cells: Ontogeny and Their Subsets in Health and Disease. Med Sci (Basel) 2018; 6:medsci6040088. [PMID: 30297662 PMCID: PMC6313400 DOI: 10.3390/medsci6040088] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 09/13/2018] [Accepted: 10/04/2018] [Indexed: 12/14/2022] Open
Abstract
Dendritic cells (DCs) are a type of cells derived from bone marrow that represent 1% or less of the total hematopoietic cells of any lymphoid organ or of the total cell count of the blood or epithelia. Dendritic cells comprise a heterogeneous population of cells localized in different tissues where they act as sentinels continuously capturing antigens to present them to T cells. Dendritic cells are uniquely capable of attracting and activating naïve CD4+ and CD8+ T cells to initiate and modulate primary immune responses. They have the ability to coordinate tolerance or immunity depending on their activation status, which is why they are also considered as the orchestrating cells of the immune response. The purpose of this review is to provide a general overview of the current knowledge on ontogeny and subsets of human dendritic cells as well as their function and different biological roles.
Collapse
Affiliation(s)
- Sandra Georgina Solano-Gálvez
- Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico.
| | - Sonia Margarita Tovar-Torres
- Departamento de Microbiología, Centro de Investigación en Ciencias de la Salud, Facultad de Ciencias de la Salud (CICSA), Universidad Anáhuac México Campus Norte, Estado de México 52786, Mexico.
| | - María Sofía Tron-Gómez
- Departamento de Microbiología, Centro de Investigación en Ciencias de la Salud, Facultad de Ciencias de la Salud (CICSA), Universidad Anáhuac México Campus Norte, Estado de México 52786, Mexico.
| | - Ariane Estrella Weiser-Smeke
- Departamento de Microbiología, Centro de Investigación en Ciencias de la Salud, Facultad de Ciencias de la Salud (CICSA), Universidad Anáhuac México Campus Norte, Estado de México 52786, Mexico.
| | - Diego Abelardo Álvarez-Hernández
- Departamento de Microbiología, Centro de Investigación en Ciencias de la Salud, Facultad de Ciencias de la Salud (CICSA), Universidad Anáhuac México Campus Norte, Estado de México 52786, Mexico.
| | | | | | - Antonio Ibarra
- Coordinación del Centro de Investigación en Ciencias de la Salud, Facultad de Ciencias de la Salud (CICSA), Universidad Anáhuac México Campus Norte, Estado de México 52786, Mexico.
| | - Laila Gutiérrez-Kobeh
- Unidad de Investigación UNAM-INC, División Investigación, Facultad de Medicina, Universidad Nacional Autónoma de México-Instituto Nacional de Cardiología "Ignacio Chávez", Mexico City 14080, Mexico.
| | - Rosalino Vázquez-López
- Departamento de Microbiología, Centro de Investigación en Ciencias de la Salud, Facultad de Ciencias de la Salud (CICSA), Universidad Anáhuac México Campus Norte, Estado de México 52786, Mexico.
| |
Collapse
|
19
|
Pigni M, Ashok D, Stevanin M, Acha-Orbea H. Establishment and Characterization of a Functionally Competent Type 2 Conventional Dendritic Cell Line. Front Immunol 2018; 9:1912. [PMID: 30197645 PMCID: PMC6117413 DOI: 10.3389/fimmu.2018.01912] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 08/02/2018] [Indexed: 12/24/2022] Open
Abstract
Dendritic cells (DCs) are the most potent antigen presenting cells and possess an incomparable ability to activate and instruct T cells, which makes them one of the cornerstones in the regulation of the cross-talk between innate and adaptive immunity. Therefore, a deep understanding of DC biology lays the foundations to describe and to harness the mechanisms that regulate the development of the adaptive response, with clear implications in a vast array of fields such as the study of autoimmune diseases and the development of new vaccines. However, the great difficulty to obtain large quantities of viable non-activated DCs for experimentation have considerably hindered the progress of DC research. Several strategies have been proposed to overcome these limitations by promoting an increase of DC abundance in vivo, by inducing DC development from DC progenitors in vitro and by generating stable DC lines. In the past years, we have described a method to derive immortalized stable DC lines, named MutuDCs, from the spleens of Mushi1 mice, a transgenic mouse strain that express the simian virus 40 Large T-oncogene in the DCs. The comparison of these DC lines with the vast variety of DC subsets described in vivo has shown that all the MutuDC lines that we have generated so far have phenotypic and functional features of type 1 conventional DCs (cDC1s). With the purpose of deriving DC lines with characteristics of type 2 conventional DCs (cDC2s), we bred a new Batf3-/- Mushi1 murine line in which the development of the cDC1 subset is severely defective. The new MutuDC line that we generated from Batf3-/- Mushi1 mice was phenotypically and functionally characterized in this work. Our results demonstrated that all the tested characteristics of this new cell line, including the expression of subset-determining transcription factors, the profile of cytokine production and the ability to present antigens, are comparable with the features of splenic CD4- cDC2s. Therefore, we concluded that our new cell line, that we named CD4- MutuDC2 line, represents a valuable model for the CD4- cDC2 subset.
Collapse
Affiliation(s)
| | | | | | - Hans Acha-Orbea
- Department of Biochemistry CIIL, University of Lausanne, Épalinges, Switzerland
| |
Collapse
|
20
|
Toxicological and pharmacological assessment of AGEN1884, a novel human IgG1 anti-CTLA-4 antibody. PLoS One 2018; 13:e0191926. [PMID: 29617360 PMCID: PMC5884502 DOI: 10.1371/journal.pone.0191926] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 12/20/2017] [Indexed: 12/15/2022] Open
Abstract
CTLA-4 and CD28 exemplify a co-inhibitory and co-stimulatory signaling axis that dynamically sculpts the interaction of antigen-specific T cells with antigen-presenting cells. Anti-CTLA-4 antibodies enhance tumor-specific immunity through a variety of mechanisms including: blockade of CD80 or CD86 binding to CTLA-4, repressing regulatory T cell function and selective elimination of intratumoral regulatory T cells via an Fcγ receptor-dependent mechanism. AGEN1884 is a novel IgG1 antibody targeting CTLA-4. It potently enhanced antigen-specific T cell responsiveness that could be potentiated in combination with other immunomodulatory antibodies. AGEN1884 was well-tolerated in non-human primates and enhanced vaccine-mediated antigen-specific immunity. AGEN1884 combined effectively with PD-1 blockade to elicit a T cell proliferative response in the periphery. Interestingly, an IgG2 variant of AGEN1884 revealed distinct functional differences that may have implications for optimal dosing regimens in patients. Taken together, the pharmacological properties of AGEN1884 support its clinical investigation as a single therapeutic and combination agent.
Collapse
|
21
|
Soria I, Alvarez J, Manzano AI, López-Relaño J, Cases B, Mas-Fontao A, Cañada FJ, Fernández-Caldas E, Casanovas M, Jiménez-Barbero J, Palomares O, Viñals-Flórez LM, Subiza JL. Mite allergoids coupled to nonoxidized mannan from Saccharomyces cerevisae efficiently target canine dendritic cells for novel allergy immunotherapy in veterinary medicine. Vet Immunol Immunopathol 2017; 190:65-72. [PMID: 28778325 DOI: 10.1016/j.vetimm.2017.07.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 07/07/2017] [Accepted: 07/10/2017] [Indexed: 12/29/2022]
Abstract
We have recently reported that grass pollen allergoids conjugated with nonoxidized mannan of Saccharomyces cerevisae using glutaraldehyde results in a novel hypoallergenic mannan-allergen complex with improved properties for allergen vaccination. Using this approach, human dendritic cells show a better allergen uptake and cytokine profile production (higher IL-10/IL-4 ratio) for therapeutic purposes. Here we aim to address whether a similar approach can be extended to dogs using canine dendritic cells. Six healthy Spanish Greyhound dogs were used as blood donors to obtain canine dendritic cells (DC) derived from peripheral blood monocytes. Allergens from Dermatophagoides farinae mite were polymerized and conjugated with nonoxidized mannan. Nuclear magnetic resonance (NMR), gel electrophoresis (SDS-PAGE), immunoblotting and IgE-ELISA inhibition studies were conducted to evaluate the main characteristics of the allergoid obtained. Mannan-allergen conjugate and controls were assayed in vitro for canine DC uptake and production of IL-4 and IL-10. The results indicate that the conjugation of D. farinae allergens with nonoxidized mannan was feasible using glutaraldehyde. The resulting product was a polymerized structure showing a high molecular weight as detected by NMR and SDS-PAGE analysis. The mannan-allergen conjugate was hypoallergenic with a reduced reactivity with specific dog IgE. An increase in both allergen uptake and IL-10/IL-4 ratio was obtained when canine DCs were incubated with the mannan-allergen conjugate, as compared with the control allergen preparations (unmodified D. farinae allergens and oxidized mannan-allergen conjugate). We conclude that hypoallergenic D. farinae allergens coupled to nonoxidized mannan is a novel allergen preparation suitable for canine allergy immunotherapy targeting dendritic cells.
Collapse
Affiliation(s)
| | | | - Ana I Manzano
- Inmunotek S.L., Alcalá de Henares, Spain; Centro de Investigaciones Biológicas, Madrid, Spain
| | - Juan López-Relaño
- Inmunotek S.L., Alcalá de Henares, Spain; Experimental Unit, Hospital Clínico San Carlos, Madrid, Spain
| | | | | | | | - Enrique Fernández-Caldas
- Inmunotek S.L., Alcalá de Henares, Spain; Division of Allergy and Immunology, University of South Florida, College of Medicine, Tampa, USA
| | | | - Jesús Jiménez-Barbero
- Centro de Investigaciones Biológicas, Madrid, Spain; CIC bioGUNE, Parque Tecnológico de Bizkaia, Bilbao, Spain
| | - Oscar Palomares
- Dpto. Bioquímica y Biología Molecular, Universidad Complutense, Madrid, Spain
| | | | | |
Collapse
|
22
|
Reiser ML, Mosaheb MM, Lisk C, Platt A, Wetzler LM. The TLR2 Binding Neisserial Porin PorB Enhances Antigen Presenting Cell Trafficking and Cross-presentation. Sci Rep 2017; 7:736. [PMID: 28389664 PMCID: PMC5428659 DOI: 10.1038/s41598-017-00555-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 03/03/2017] [Indexed: 01/06/2023] Open
Abstract
TOLL-like receptor (TLR) ligands activate both innate and adaptive immune cells, while modulating the cellular immune response. The outer membrane protein (OMP) from Neisseria meninigitidis, PorB, is a naturally occurring TLR2 ligand and functions as an adjuvant. Here, we demonstrate that PorB increases the level of OVA in the endo-/lysosomal cellular compartment of BMDCs, increases antigen presenting cell (APC) trafficking to draining lymph nodes, and enhances antigen cross-presentation. PorB is capable of mounting an antigen specific T cell response by efficiently stimulating antigen cross-presentation in vivo and in vitro assessed by BMDC OT-I cocultivation assays. The enhanced antigen cross-presentation and the increased APC recruitment to secondary lymphoid tissues expand the scope of known adjuvant effects of PorB on the immune system. Our findings lead to a better understanding of how TLR-ligand based adjuvants can alter and modulate immune responses.
Collapse
Affiliation(s)
- Michael L Reiser
- Section of Infectious Diseases, Department of Medicine, Boston Medical Center, Boston, USA
| | - Munir M Mosaheb
- Department of Microbiology, Boston University School of Medicine, Boston, USA
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, USA
| | - Christina Lisk
- Section of Infectious Diseases, Department of Medicine, Boston Medical Center, Boston, USA
| | - Andrew Platt
- Department of Microbiology, Boston University School of Medicine, Boston, USA
| | - Lee M Wetzler
- Department of Microbiology, Boston University School of Medicine, Boston, USA.
- Section of Infectious Diseases, Department of Medicine, Boston Medical Center, Boston, USA.
| |
Collapse
|
23
|
Waight JD, Gombos RB, Wilson NS. Harnessing co-stimulatory TNF receptors for cancer immunotherapy: Current approaches and future opportunities. Hum Antibodies 2017; 25:87-109. [PMID: 28085016 DOI: 10.3233/hab-160308] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Co-stimulatory tumor necrosis factor receptors (TNFRs) can sculpt the responsiveness of T cells recognizing tumor-associated antigens. For this reason, agonist antibodies targeting CD137, CD357, CD134 and CD27 have received considerable attention for their therapeutic utility in enhancing anti-tumor immune responses, particularly in combination with other immuno-modulatory antibodies targeting co-inhibitory pathways in T cells. The design of therapeutic antibodies that optimally engage and activate co-stimulatory TNFRs presents an important challenge of how to promote effective anti-tumor immunity while avoiding serious immune-related adverse events. Here we review our current understanding of the expression, signaling and structural features of CD137, CD357, CD134 and CD27, and how this may inform the design of pharmacologically active immuno-modulatory antibodies targeting these receptors. This includes the integration of our emerging knowledge of the role of Fcγ receptors (FcγRs) in facilitating antibody-mediated receptor clustering and forward signaling, as well as promoting immune effector cell-mediated activities. Finally, we bring our current preclinical and clinical knowledge of co-stimulatory TNFR antibodies into the context of opportunities for next generation molecules with improved pharmacologic properties.
Collapse
MESH Headings
- Animals
- Antigens, CD/genetics
- Antigens, CD/immunology
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/immunology
- Antineoplastic Agents, Immunological/therapeutic use
- Gene Expression Regulation
- Humans
- Immunity, Cellular/drug effects
- Immunotherapy/methods
- Neoplasms/drug therapy
- Neoplasms/genetics
- Neoplasms/immunology
- Neoplasms/pathology
- Receptors, IgG/agonists
- Receptors, IgG/genetics
- Receptors, IgG/immunology
- Receptors, Tumor Necrosis Factor/agonists
- Receptors, Tumor Necrosis Factor/genetics
- Receptors, Tumor Necrosis Factor/immunology
- Signal Transduction
- T-Lymphocytes/drug effects
- T-Lymphocytes/immunology
- T-Lymphocytes/pathology
Collapse
|
24
|
Cell-SELEX-based selection and characterization of a G-quadruplex DNA aptamer against mouse dendritic cells. Int Immunopharmacol 2016; 36:324-332. [PMID: 27232653 DOI: 10.1016/j.intimp.2016.04.042] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 04/28/2016] [Indexed: 01/10/2023]
Abstract
Targeting of dendritic cells (DCs) by aptamers increases antigen capture and presentation to the immune system. Our aim was to produce aptamers against DC molecules using the cell-SELEX procedure. For this purpose, 18 rounds of cell-SELEX were performed on mouse macrophage J774A.1 and CT26 as target and control cells, respectively. The selected aptamers were truncated and their binding to mouse macrophages, and immature and mature DCs analyzed. Two macrophage-specific aptamers, Seq6 and Seq7, were identified. A truncated form of Seq7, Seq7-4, 33 nucleotides in length and containing the G-quadruplex, bound macrophages and immature DCs with KD values in the nanomolar range. We anticipate that Seq7-4 has potential as a therapeutic tool in targeting of mouse macrophages and immature DCs to efficiently improve different immunotherapy approaches.
Collapse
|
25
|
Antigenically Modified Human Pluripotent Stem Cells Generate Antigen-Presenting Dendritic Cells. Sci Rep 2015; 5:15262. [PMID: 26471005 PMCID: PMC4608011 DOI: 10.1038/srep15262] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 09/21/2015] [Indexed: 12/19/2022] Open
Abstract
Human pluripotent stem cells (hPSCs) provide a promising platform to produce dendritic cell (DC) vaccine. To streamline the production process, we investigated a unique antigen-loading strategy that suits this novel platform. Specifically, we stably modified hPSCs using tumour antigen genes in the form of a full-length tumour antigen gene or an artificial tumour antigen epitope-coding minigene. Such antigenically modified hPSCs were able to differentiate into tumour antigen-presenting DCs. Without conventional antigen-loading, DCs derived from the minigene-modified hPSCs were ready to prime a tumour antigen-specific T cell response and further expand these specific T cells in restimulation processes. These expanded tumour antigen-specific T cells were potent effectors with central memory or effector memory phenotype. Thus, we demonstrated that immunocompetent tumour antigen-loaded DCs can be directly generated from antigenically modified hPSCs. Using such strategy, we can completely eliminate the conventional antigen-loading step and significantly simplify the production of DC vaccine from hPSCs.
Collapse
|
26
|
Seki T, Yamamoto M, Taguchi Y, Miyauchi M, Akiyama N, Yamaguchi N, Gohda J, Akiyama T, Inoue JI. Visualization of RelB expression and activation at the single-cell level during dendritic cell maturation in Relb-Venus knock-in mice. J Biochem 2015; 158:485-95. [PMID: 26115685 DOI: 10.1093/jb/mvv064] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 05/25/2015] [Indexed: 12/19/2022] Open
Abstract
RelB is activated by the non-canonical NF-κB pathway, which is crucial for immunity by establishing lymphoid organogenesis and B-cell and dendritic cell (DC) maturation. To elucidate the mechanism of the RelB-mediated immune cell maturation, a precise understanding of the relationship between cell maturation and RelB expression and activation at the single-cell level is required. Therefore, we generated knock-in mice expressing a fusion protein between RelB and fluorescent protein (RelB-Venus) from the Relb locus. The Relb(Venus/Venus) mice developed without any abnormalities observed in the Relb(-/-) mice, allowing us to monitor RelB-Venus expression and nuclear localization as RelB expression and activation. Relb(Venus/Venus) DC analyses revealed that DCs consist of RelB(-), RelB(low) and RelB(high) populations. The RelB(high) population, which included mature DCs with projections, displayed RelB nuclear localization, whereas RelB in the RelB(low) population was in the cytoplasm. Although both the RelB(low) and RelB(-) populations barely showed projections, MHC II and co-stimulatory molecule expression were higher in the RelB(low) than in the RelB(-) splenic conventional DCs. Taken together, our results identify the RelB(low) population as a possible novel intermediate maturation stage of cDCs and the Relb(Venus/Venus) mice as a useful tool to analyse the dynamic regulation of the non-canonical NF-κB pathway.
Collapse
Affiliation(s)
- Takao Seki
- Division of Cellular and Molecular Biology, Department of Cancer Biology, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Mami Yamamoto
- Division of Cellular and Molecular Biology, Department of Cancer Biology, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Yuu Taguchi
- Division of Cellular and Molecular Biology, Department of Cancer Biology, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Maki Miyauchi
- Division of Cellular and Molecular Biology, Department of Cancer Biology, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Nobuko Akiyama
- Division of Cellular and Molecular Biology, Department of Cancer Biology, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Noritaka Yamaguchi
- Department of Molecular Cell Biology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 260-8675, Japan; and
| | - Jin Gohda
- Research Center for Asian Infectious Diseases, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Taishin Akiyama
- Division of Cellular and Molecular Biology, Department of Cancer Biology, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Jun-ichiro Inoue
- Division of Cellular and Molecular Biology, Department of Cancer Biology, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan;
| |
Collapse
|
27
|
The transcription factor TFEB acts as a molecular switch that regulates exogenous antigen-presentation pathways. Nat Immunol 2015; 16:729-36. [PMID: 26030023 DOI: 10.1038/ni.3196] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 05/05/2015] [Indexed: 12/12/2022]
Abstract
Dendritic cells (DCs) can initiate immune responses by presenting exogenous antigens to T cells via both major histocompatibility complex (MHC) class I pathways and MHC class II pathways. Lysosomal activity has an important role in modulating the balance between these two pathways. The transcription factor TFEB regulates lysosomal function by inducing lysosomal activation. Here we report that TFEB expression inhibited the presentation of exogenous antigen by MHC class I while enhancing presentation via MHC class II. TFEB promoted phagosomal acidification and protein degradation. Furthermore, we found that the activation of TFEB was regulated during DC maturation and that phagosomal acidification was impaired in DCs in which the gene encoding TFEB was silenced. Our data indicate that TFEB is a key participant in the differential regulation of the presentation of exogenous antigens by DCs.
Collapse
|
28
|
Abstract
Aptamers, as a novel class of molecular probes for diagnosis, imaging and targeting therapy, have attracted increasing attention in recent years. Aptamers are generated from libraries of single-stranded nucleic acids against different molecules via the "systematic evolution of ligands by exponential enrichment" (SELEX) method. SELEX is a repetitive process of a sequential selection procedure in which a DNA or RNA library pool is incubated separately with target and control molecules to select specific oligonucleotide aptamers with high affinities and specificities. Cell-SELEX is a modified version of the SELEX process in which whole living cells are used as targets for the aptamers. Dendritic cell (DC) targeting, as a new therapeutic approach, can improve the efficiency of immunotherapy in the treatment of allergies and cancers. DCs use various receptors to continuously induce adaptive immunity via capture and presentation of antigens to naïve T cells. DCs are considered as the best targets in modulating immune responses against cancer, autoimmunity, allergy and transplantation. Aptamers, as a new agent, can be applied in DC targeting. The purpose of this review is to present some general concepts of aptamer production and DC targeting by aptamer molecules.
Collapse
Affiliation(s)
- A Ganji
- a Student Research Committee , Mashhad University of Medical Sciences , Mashhad , Iran .,b Immunology Research Center, Medical School, Mashhad University of Medical Sciences , Mashhad , Iran , and
| | - A Varasteh
- c Allergy Research Center, Medical School, Mashhad University of Medical Sciences , Mashhad , Iran
| | - M Sankian
- b Immunology Research Center, Medical School, Mashhad University of Medical Sciences , Mashhad , Iran , and
| |
Collapse
|
29
|
Ogawa F, Amano H, Eshima K, Ito Y, Matsui Y, Hosono K, Kitasato H, Iyoda A, Iwabuchi K, Kumagai Y, Satoh Y, Narumiya S, Majima M. Prostanoid induces premetastatic niche in regional lymph nodes. J Clin Invest 2014; 124:4882-94. [PMID: 25271626 DOI: 10.1172/jci73530] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Accepted: 08/21/2014] [Indexed: 01/17/2023] Open
Abstract
The lymphatic system is an important route for cancer dissemination, and lymph node metastasis (LNM) serves as a critical prognostic determinant in cancer patients. We investigated the contribution of COX-2-derived prostaglandin E2 (PGE2) in the formation of a premetastatic niche and LNM. A murine model of Lewis lung carcinoma (LLC) cell metastasis revealed that COX-2 is expressed in DCs from the early stage in the lymph node subcapsular regions, and COX-2 inhibition markedly suppressed mediastinal LNM. Stromal cell-derived factor-1 (SDF-1) was elevated in DCs before LLC cell infiltration to the lymph nodes, and a COX-2 inhibitor, an SDF-1 antagonist, and a CXCR4 neutralizing antibody all reduced LNM. Moreover, LNM was reduced in mice lacking the PGE2 receptor EP3, and stimulation of cultured DCs with an EP3 agonist increased SDF-1 production. Compared with WT CD11c+ DCs, injection of EP3-deficient CD11c+ DCs dramatically reduced accumulation of SDF-1+CD11c+ DCs in regional LNs and LNM in LLC-injected mice. Accumulation of Tregs and lymph node lymphangiogenesis, which may influence the fate of metastasized tumor cells, was also COX-2/EP3-dependent. These results indicate that DCs induce a premetastatic niche during LNM via COX-2/EP3-dependent induction of SDF-1 and suggest that inhibition of this signaling axis may be an effective strategy to suppress premetastatic niche formation and LNM.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/pharmacology
- Carcinoma, Lewis Lung/immunology
- Carcinoma, Lewis Lung/metabolism
- Carcinoma, Lewis Lung/secondary
- Celecoxib
- Cell Line, Tumor
- Chemokine CXCL12/metabolism
- Cyclooxygenase 2/metabolism
- Cyclooxygenase 2 Inhibitors/pharmacology
- Dendritic Cells/immunology
- Dinoprostone/physiology
- Drug Screening Assays, Antitumor
- Gene Knockout Techniques
- Lung Neoplasms/immunology
- Lung Neoplasms/metabolism
- Lung Neoplasms/pathology
- Lymph Nodes/immunology
- Lymph Nodes/pathology
- Lymphangiogenesis
- Lymphatic Metastasis
- Male
- Mice, Knockout
- Neoplasm Transplantation
- Pyrazoles/pharmacology
- Receptors, CXCR4/metabolism
- Receptors, Prostaglandin E, EP3 Subtype/genetics
- Receptors, Prostaglandin E, EP3 Subtype/metabolism
- Signal Transduction
- Sulfonamides/pharmacology
- T-Lymphocytes, Regulatory/immunology
Collapse
|
30
|
Urra X, Miró F, Chamorro A, Planas AM. Antigen-specific immune reactions to ischemic stroke. Front Cell Neurosci 2014; 8:278. [PMID: 25309322 PMCID: PMC4162361 DOI: 10.3389/fncel.2014.00278] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 08/22/2014] [Indexed: 12/24/2022] Open
Abstract
Brain proteins are detected in the cerebrospinal fluid (CSF) and blood of stroke patients and their concentration is related to the extent of brain damage. Antibodies against brain antigens develop after stroke, suggesting a humoral immune response to the brain injury. Furthermore, induced immune tolerance is beneficial in animal models of cerebral ischemia. The presence of circulating T cells sensitized against brain antigens, and antigen presenting cells (APCs) carrying brain antigens in draining lymphoid tissue of stroke patients support the notion that stroke might induce antigen-specific immune responses. After stroke, brain proteins that are normally hidden from the periphery, inflammatory mediators, and danger signals can exit the brain through several efflux routes. They can reach the blood after leaking out of the damaged blood-brain barrier (BBB) or following the drainage of interstitial fluid to the dural venous sinus, or reach the cervical lymph nodes through the nasal lymphatics following CSF drainage along the arachnoid sheaths of nerves across the nasal submucosa. The route and mode of access of brain antigens to lymphoid tissue could influence the type of response. Central and peripheral tolerance prevents autoimmunity, but the actual mechanisms of tolerance to brain antigens released into the periphery in the presence of inflammation, danger signals, and APCs, are not fully characterized. Stroke does not systematically trigger autoimmunity, but under certain circumstances, such as pronounced systemic inflammation or infection, autoreactive T cells could escape the tolerance controls. Further investigation is needed to elucidate whether antigen-specific immune events could underlie neurological complications impairing recovery from stroke.
Collapse
Affiliation(s)
- Xabier Urra
- Functional Unit of Cerebrovascular Diseases, Hospital Clínic Barcelona, Spain ; August Pi i Sunyer Biomedical Research Institute (IDIBAPS) Barcelona, Spain
| | - Francesc Miró
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS) Barcelona, Spain
| | - Angel Chamorro
- Functional Unit of Cerebrovascular Diseases, Hospital Clínic Barcelona, Spain ; August Pi i Sunyer Biomedical Research Institute (IDIBAPS) Barcelona, Spain
| | - Anna M Planas
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS) Barcelona, Spain ; Department of Brain Ischemia and Neurodegeneration, Instituto de Investigaciones Biomédicas de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC) Barcelona, Spain
| |
Collapse
|
31
|
Vega-Ramos J, Roquilly A, Asehnoune K, Villadangos JA. Modulation of dendritic cell antigen presentation by pathogens, tissue damage and secondary inflammatory signals. Curr Opin Pharmacol 2014; 17:64-70. [PMID: 25128781 DOI: 10.1016/j.coph.2014.07.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Revised: 07/21/2014] [Accepted: 07/23/2014] [Indexed: 12/29/2022]
Abstract
Antigen presentation by dendritic cells (DC) is regulated directly by pathogen-associated or cell death-associated cues, or indirectly by immunomodulatory molecules produced during infection or tissue damage. DC modulation by direct encounter of pathogen-associated compounds has been thoroughly studied; the effects of molecules associated with cell death are less well characterized; modulation by secondary signals remain poorly understood. In this review we describe recent studies on the role of these three categories of immunomodulatory compounds on DC. We conclude that characterization of the role of secondary immunomodulators is an area in dare need of further study. The outcomes of this endeavor will be new opportunities for the development of better vaccines and compounds applicable to the therapeutic immunomodulation of DC function.
Collapse
Affiliation(s)
- Javier Vega-Ramos
- Department of Microbiology and Immunology, Doherty Institute of Infection and Immunity, The University of Melbourne, Pakville, Australia
| | - Antoine Roquilly
- Department of Microbiology and Immunology, Doherty Institute of Infection and Immunity, The University of Melbourne, Pakville, Australia; Laboratoire UPRES EA 3826 "Thérapeutiques cliniques et expérimentales des infections", Faculte de Médecine, Université de Nantes, France; Service d'Anesthésie Réanimation Chirurgicale, Hôtel Dieu, Nantes, France
| | - Karim Asehnoune
- Laboratoire UPRES EA 3826 "Thérapeutiques cliniques et expérimentales des infections", Faculte de Médecine, Université de Nantes, France; Service d'Anesthésie Réanimation Chirurgicale, Hôtel Dieu, Nantes, France
| | - Jose A Villadangos
- Department of Microbiology and Immunology, Doherty Institute of Infection and Immunity, The University of Melbourne, Pakville, Australia; Department of Biochemistry and Molecular Biology, Bio21 Institute, The University of Melbourne, Parkville, Australia.
| |
Collapse
|
32
|
Apostolopoulos V, Pietersz GA, Tsibanis A, Tsikkinis A, Stojanovska L, McKenzie IF, Vassilaros S. Dendritic cell immunotherapy: clinical outcomes. Clin Transl Immunology 2014; 3:e21. [PMID: 25505969 PMCID: PMC4232065 DOI: 10.1038/cti.2014.14] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Revised: 05/28/2014] [Accepted: 06/09/2014] [Indexed: 01/04/2023] Open
Abstract
The use of tumour-associated antigens for cancer immunotherapy studies is exacerbated by tolerance to these self-antigens. Tolerance may be broken by using ex vivo monocyte-derived dendritic cells (DCs) pulsed with self-antigens. Targeting tumour-associated antigens directly to DCs in vivo is an alternative and simpler strategy. The identification of cell surface receptors on DCs, and targeting antigens to DC receptors, has become a popular approach for inducing effective immune responses against cancer antigens. Many years ago, we demonstrated that targeting the mannose receptor on macrophages using the carbohydrate mannan to DCs led to appropriate immune responses and tumour protection in animal models. We conducted Phase I, I/II and II, clinical trials demonstrating the effectiveness of oxidised mannan-MUC1 in patients with adenocarcinomas. Here we summarise DC targeting approaches and their efficacy in human clinical trials.
Collapse
Affiliation(s)
- Vasso Apostolopoulos
- College of Health and Biomedicine, Centre for Chronic Disease Prevention and Management, Victoria University , Melbourne, VIC, Australia
| | - Geoffrey A Pietersz
- BioOrganic and Medicinal Chemistry Laboratory, Centre for Biomedical Research, Burnet Institute , Melbourne, VIC, Australia ; Department of Pathology, University of Melbourne , Melbourne, VIC, Australia ; Department of Immunology, Monash University , Melbourne, VIC, Australia
| | | | | | - Lily Stojanovska
- College of Health and Biomedicine, Centre for Chronic Disease Prevention and Management, Victoria University , Melbourne, VIC, Australia
| | - Ian Fc McKenzie
- Department of Pathology, University of Melbourne , Melbourne, VIC, Australia
| | | |
Collapse
|
33
|
He X, Yan J, Zhu X, Wang Q, Pang W, Qi Z, Wang M, Luo E, Parker DM, Cantorna MT, Cui L, Cao Y. Vitamin D inhibits the occurrence of experimental cerebral malaria in mice by suppressing the host inflammatory response. THE JOURNAL OF IMMUNOLOGY 2014; 193:1314-23. [PMID: 24965778 DOI: 10.4049/jimmunol.1400089] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
In animal models of experimental cerebral malaria (ECM), neuropathology is associated with an overwhelming inflammatory response and sequestration of leukocytes and parasite-infected RBCs in the brain. In this study, we explored the effect of vitamin D (VD; cholecalciferol) treatment on host immunity and outcome of ECM in C57BL/6 mice during Plasmodium berghei ANKA (PbA) infection. We observed that oral administration of VD both before and after PbA infection completely protected mice from ECM. VD administration significantly dampened the inducible systemic inflammatory responses with reduced circulating cytokines IFN-γ and TNF and decreased expression of these cytokines by the spleen cells. Meanwhile, VD also resulted in decreased expression of the chemokines CXCL9 and CXCL10 and cytoadhesion molecules (ICAM-1, VCAM-1, and CD36) in the brain, leading to reduced accumulation of pathogenic T cells in the brain and ultimately substantial improvement of the blood-brain barriers of PbA-infected mice. In addition, VD inhibited the differentiation, activation, and maturation of splenic dendritic cells. Meanwhile, regulatory T cells and IL-10 expression levels were upregulated upon VD treatment. These data collectively demonstrated the suppressive function of VD on host inflammatory responses, which provides significant survival benefits in the murine ECM model.
Collapse
Affiliation(s)
- Xiyue He
- Department of Immunology, China Medical University, Shenyang 110001, China
| | - Juan Yan
- Department of Immunology, China Medical University, Shenyang 110001, China
| | - Xiaotong Zhu
- Department of Immunology, China Medical University, Shenyang 110001, China; Department of Entomology, Pennsylvania State University, University Park, PA 16802; and
| | - Qinghui Wang
- Department of Immunology, China Medical University, Shenyang 110001, China; Institute of Pathology and Pathophysiology, China Medical University, Shenyang 110001, China
| | - Wei Pang
- Department of Immunology, China Medical University, Shenyang 110001, China
| | - Zanmei Qi
- Department of Immunology, China Medical University, Shenyang 110001, China
| | - Meilian Wang
- Department of Microbiology and Parasitology, China Medical University, Shenyang 110001, China
| | - Enjie Luo
- Department of Microbiology and Parasitology, China Medical University, Shenyang 110001, China
| | - Daniel M Parker
- Department of Entomology, Pennsylvania State University, University Park, PA 16802; and
| | - Margherita T Cantorna
- Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, PA 16802
| | - Liwang Cui
- Department of Entomology, Pennsylvania State University, University Park, PA 16802; and
| | - Yaming Cao
- Department of Immunology, China Medical University, Shenyang 110001, China;
| |
Collapse
|
34
|
Lombardi VC, Khaiboullina SF. Plasmacytoid dendritic cells of the gut: relevance to immunity and pathology. Clin Immunol 2014; 153:165-77. [PMID: 24769378 DOI: 10.1016/j.clim.2014.04.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 04/11/2014] [Accepted: 04/12/2014] [Indexed: 12/15/2022]
Abstract
Plasmacytoid dendritic cells (pDCs) are bone marrow-derived immune cells with the ability to express copious amounts of type I and III interferon (IFN) and can differentiate into antigen-presenting dendritic cells as a result of stimulation by pathogen-derived nucleic acid. These powerful combined functionalities allow pDCs to bridge the innate and adaptive immune systems resulting in a concerted pathogen response. The contribution of pDCs to gastrointestinal immunity is only now being elucidated and is proving to be a critical component in systemic immunity. This review will explore the immunology of pDCs and will discuss their involvement in human disease and tolerance with an emphasis on those in the gastrointestinal lymphoid tissue.
Collapse
Affiliation(s)
- Vincent C Lombardi
- Department of Biochemistry and Molecular Biology, University of Nevada School of Medicine, WPI, University of Nevada, Reno, 1664 N Virginia St. MS 0552, Reno, NV 89557, USA.
| | - Svetlana F Khaiboullina
- Department of Biochemistry and Molecular Biology, University of Nevada School of Medicine, WPI, University of Nevada, Reno, 1664 N Virginia St. MS 0552, Reno, NV 89557, USA; Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia.
| |
Collapse
|
35
|
PEG as a spacer arm markedly increases the immunogenicity of meningococcal group Y polysaccharide conjugate vaccine. J Control Release 2013; 172:382-389. [DOI: 10.1016/j.jconrel.2013.03.008] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Revised: 02/27/2013] [Accepted: 03/02/2013] [Indexed: 11/21/2022]
|
36
|
Targeting antigens to dendritic cell receptors for vaccine development. JOURNAL OF DRUG DELIVERY 2013; 2013:869718. [PMID: 24228179 PMCID: PMC3817681 DOI: 10.1155/2013/869718] [Citation(s) in RCA: 110] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Accepted: 07/11/2013] [Indexed: 12/30/2022]
Abstract
Dendritic cells (DCs) are highly specialized antigen presenting cells of the immune system which play a key role in regulating immune responses. Depending on the method of antigen delivery, DCs stimulate immune responses or induce tolerance. As a consequence of the dual function of DCs, DCs are studied in the context of immunotherapy for both cancer and autoimmune diseases. In vaccine development, a major aim is to induce strong, specific T-cell responses. This is achieved by targeting antigen to cell surface molecules on DCs that efficiently channel the antigen into endocytic compartments for loading onto MHC molecules and stimulation of T-cell responses. The most attractive cell surface receptors, expressed on DCs used as targets for antigen delivery for cancer and other diseases, are discussed.
Collapse
|
37
|
Wagner CS, Grotzke J, Cresswell P. Intracellular regulation of cross-presentation during dendritic cell maturation. PLoS One 2013; 8:e76801. [PMID: 24098562 PMCID: PMC3789698 DOI: 10.1371/journal.pone.0076801] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Accepted: 09/03/2013] [Indexed: 11/19/2022] Open
Abstract
We have investigated the effect of different maturation stimuli on the ability of mature dendritic cells (DCs) to cross-present newly acquired particulate antigens. Cross-presentation was impaired in DCs matured by treatment with TNF-α, CpG and LPS, but was less affected upon CD40L-induced maturation. The difference could not be explained by decreased antigen uptake or translocation into the cytosol, but decreased cross-presentation ability did correlate with increased phagosomal/lysosomal acidification. Nevertheless, intra-phagosomal degradation of OVA was not increased in matured samples, suggesting that decreasing phagosomal pH may also regulate cross-presentation by a mechanism other than enhancing degradation.
Collapse
Affiliation(s)
- Claudia S. Wagner
- Department of Immunobiology, Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Jeff Grotzke
- Department of Immunobiology, Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Peter Cresswell
- Department of Immunobiology, Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, United States of America
- * E-mail:
| |
Collapse
|
38
|
|
39
|
Sheng KC, Day S, Wright MD, Stojanovska L, Apostolopoulos V. Enhanced Dendritic Cell-Mediated Antigen-Specific CD4+ T Cell Responses: IFN-Gamma Aids TLR Stimulation. JOURNAL OF DRUG DELIVERY 2013; 2013:516749. [PMID: 23781340 PMCID: PMC3679806 DOI: 10.1155/2013/516749] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Accepted: 02/18/2013] [Indexed: 12/20/2022]
Abstract
Phenotypic maturation and T cell stimulation are two functional attributes of DCs critical for immune induction. The combination of antigens, including those from cancer, with Toll-like receptor (TLR) ligands induces far superior cellular immune responses compared to antigen alone. In this study, IFN-gamma treatment of bone marrow-derived DC, followed by incubation with the TLR2, TLR4, or TLR9 agonists, enhanced DC activation compared to TLR ligation alone. Most notably, the upregulation of CD40 with LPS stimulation and CD86 with CpG stimulation was observed in in vitro cultures. Similarly, IFN-gamma coinjected with TLR ligands was able to promote DC activation in vivo, with DCs migrating from the site of immunization to the popliteal lymph nodes demonstrating increased expression of CD80 and CD86. The heightened DC activation translated to a drastic increase in T cell stimulatory capacity in both antigen independent and antigen dependent fashions. This is the first time that IFN-gamma has been shown to have a combined effect with TLR ligation to enhance DC activation and function. The results demonstrate the novel use of IFN-gamma together with TLR agonists to enhance antigen-specific T cell responses, for applications in the development of enhanced vaccines and drug targets against diseases including cancer.
Collapse
Affiliation(s)
- Kuo-Ching Sheng
- Immunology and Vaccine Laboratory, Burnet Institute, Melbourne, VIC 3004, Australia
- Institute for Glycomics, Griffith University, Gold Coast, QLD 4215, Australia
| | - Stephanie Day
- Immunology and Vaccine Laboratory, Burnet Institute, Melbourne, VIC 3004, Australia
- Department of Immunology, Monash University, Melbourne, VIC 3004, Australia
| | - Mark D. Wright
- Department of Immunology, Monash University, Melbourne, VIC 3004, Australia
| | - Lily Stojanovska
- College of Health and Biomedicine, Victoria University, VIC 3021, Australia
| | - Vasso Apostolopoulos
- Immunology and Vaccine Laboratory, Burnet Institute, Melbourne, VIC 3004, Australia
- VA Consulting Services, Melbourne, VIC 3030, Australia
| |
Collapse
|
40
|
Sun X, Jones HP, Dobbs N, Bodhankar S, Simecka JW. Dendritic cells are the major antigen presenting cells in inflammatory lesions of murine Mycoplasma respiratory disease. PLoS One 2013; 8:e55984. [PMID: 23390557 PMCID: PMC3563630 DOI: 10.1371/journal.pone.0055984] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2011] [Accepted: 01/08/2013] [Indexed: 11/18/2022] Open
Abstract
Mycoplasmas cause chronic respiratory diseases in animals and humans, and to date, development of vaccines have been problematic. Using a murine model of mycoplasma pneumonia, lymphocyte responses, specifically T cells, were shown to confer protection as well as promote immunopathology in mycoplasma disease. Because T cells play such a critical role, it is important to define the role of antigen presenting cells (APC) as these cells may influence either exacerbation of mycoplasma disease pathogenesis or enhancement of protective immunity. The roles of APC, such as dendritic cells and/or macrophages, and their ability to modulate adaptive immunity in mycoplasma disease are currently unknown. Therefore, the purpose of this study was to identify individual pulmonary APC populations that may contribute to the activation of T cell responses during mycoplasma disease pathogenesis. The present study indeed demonstrates increasing numbers of CD11c− F4/80+ cells, which contain macrophages, and more mature/activated CD11c+ F4/80− cells, containing DC, in the lungs after infection. CD11c− F4/80+ macrophage-enriched cells and CD11c+ F4/80− dendritic cell-enriched populations showed different patterns of cytokine mRNA expression, supporting the idea that these cells have different impacts on immunity in response to infection. In fact, DC containing CD11c+ F4/80− cell populations from the lungs of infected mice were most capable of stimulating mycoplasma-specific CD4+ Th cell responses in vitro. In vivo, these CD11c+F4/80− cells were co-localized with CD4+ Th cells in inflammatory infiltrates in the lungs of mycoplasma-infected mice. Thus, CD11c+F4/80− dendritic cells appear to be the major APC population responsible for pulmonary T cell stimulation in mycoplasma-infected mice, and these dendritic cells likely contribute to responses impacting disease pathogenesis.
Collapse
Affiliation(s)
- Xiangle Sun
- Department of Molecular Biology and Immunology, University of North Texas Health Science Center in Fort Worth, Fort Worth, Texas, United States of America
| | - Harlan P. Jones
- Department of Molecular Biology and Immunology, University of North Texas Health Science Center in Fort Worth, Fort Worth, Texas, United States of America
| | - Nicole Dobbs
- Department of Molecular Biology and Immunology, University of North Texas Health Science Center in Fort Worth, Fort Worth, Texas, United States of America
| | - Sheetal Bodhankar
- Department of Molecular Biology and Immunology, University of North Texas Health Science Center in Fort Worth, Fort Worth, Texas, United States of America
| | - Jerry W. Simecka
- Department of Molecular Biology and Immunology, University of North Texas Health Science Center in Fort Worth, Fort Worth, Texas, United States of America
- * E-mail:
| |
Collapse
|
41
|
Andersson LI, Cirkic E, Hellman P, Eriksson H. Myeloid blood dendritic cells and monocyte-derived dendritic cells differ in their endocytosing capability. Hum Immunol 2012; 73:1073-81. [DOI: 10.1016/j.humimm.2012.08.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2011] [Revised: 07/23/2012] [Accepted: 08/09/2012] [Indexed: 01/14/2023]
|
42
|
Tel J, van der Leun AM, Figdor CG, Torensma R, de Vries IJM. Harnessing human plasmacytoid dendritic cells as professional APCs. Cancer Immunol Immunother 2012; 61:1279-88. [PMID: 22294456 PMCID: PMC3401502 DOI: 10.1007/s00262-012-1210-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2011] [Accepted: 01/19/2012] [Indexed: 12/23/2022]
Abstract
The plasmacytoid dendritic cell (pDC) constitutes a unique DC subset that links the innate and adaptive arm of the immune system. Whereas the unique capability of pDCs to produce large amounts of type I IFNs in response to pathogen recognition is generally accepted, their antigen-presenting function is often neglected since most studies on antigen presentation are aimed at other DC subsets. Recently, pDCs were demonstrated capable to present antigen leading to protective tumor immunity. In this review, we discuss how pDCs could be exploited in the fight against cancer by analyzing their capacity to capture, process and (cross-) present antigen.
Collapse
Affiliation(s)
- Jurjen Tel
- Department of Tumor Immunology, Radboud University Nijmegen Medical Centre and Nijmegen Centre for Molecular Life Sciences, Nijmegen, The Netherlands
| | | | | | | | | |
Collapse
|
43
|
Kumar S, Gowda NM, Wu X, Gowda RN, Gowda DC. CD36 modulates proinflammatory cytokine responses to Plasmodium falciparum glycosylphosphatidylinositols and merozoites by dendritic cells. Parasite Immunol 2012; 34:372-82. [PMID: 22486596 DOI: 10.1111/j.1365-3024.2012.01367.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Studies have shown that glycosylphosphatidylinositols (GPIs) of Plasmodium falciparum activate macrophages mainly through Toll-like receptor 2 (TLR2)-mediated signalling and to certain extent through TLR4-mediated signalling to induce proinflammatory cytokine production. However, the ability of parasite GPIs to activate dendritic cells (DCs) has not been reported. Here, we show that parasite GPIs efficiently activate DCs through TLR2-mediated signalling mechanism and induce the production of TNF-α and IL-12. We also studied the role of scavenger receptor CD36 in P. falciparum GPI- and merozoite-induced cytokine responses by DCs. The results indicate that CD36 modulates the cytokine-inducing activity of the parasite GPIs by collaborating with TLR2 in DCs. Furthermore, our data reveal that CD36 modulates the activity of P. falciparum merozoites, likely by the contribution of phagocytosis-coupled CD36-mediated signalling to the signalling induced by merozoites. Altogether, these results contribute towards understanding of signalling mechanisms in malaria parasite-induced activation of the innate immune system.
Collapse
Affiliation(s)
- S Kumar
- Department of Biochemistry and Molecular Biology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | | | | | | | | |
Collapse
|
44
|
Pilzner C, Bühling F, Reinheckel T, Chwieralski C, Rathinasamy A, Lauenstein HD, Wex T, Welte T, Braun A, Groneberg DA. Allergic airway inflammation in mice deficient for the antigen-processing protease cathepsin E. Int Arch Allergy Immunol 2012; 159:367-83. [PMID: 22846634 DOI: 10.1159/000338288] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2011] [Accepted: 03/14/2012] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Allergic asthma is a Th2-type chronic inflammatory disease of the lung. It is characterized by infiltration of eosinophils, neutrophils, mast cells and T lymphocytes into the airways. Th2 cytokines like interleukin (IL)-4, IL-5 and chemokines like eotaxin are increased in the asthmatic response. The processing and presentation of exogenous antigens is important in the sensitization to an allergen. Cathepsin E (Ctse) is an intracellular aspartic endoprotease which is expressed in immune cells like dendritic cells (DCs). It was found to play an essential role in the processing and presentation of ovalbumin (OVA). The aim of the present study was to investigate the inhibition of Ctse in two different experimental models of allergic airway inflammation. METHODS Ctse wild-type (Ctse(+/+)) and Ctse-deficient (Ctse(-/-)) bone marrow-derived DCs (BMDCs) were pulsed with OVA/OVA peptide and cocultured with OVA transgenic T II (OT II) cells whose proliferation was subsequently analyzed. Two different in vivo asthma models with Ctse(+/+) and Ctse(-/-) mice were performed: an acute OVA-induced and a subchronic Phleum pratense-induced airway inflammation. RESULTS Proliferation of OT II cells was decreased when cocultured with BMDCs of Ctse(-/-) mice as compared to cells cocultured with BMDCs of Ctse(+/+) mice. In vivo, Ctse deficiency led to reduced lymphocyte influx after allergen sensitization and challenge in both investigated airway inflammation models, compared to their control groups. CONCLUSION Ctse deficiency leads to a reduced antigen presentation in vitro. This is followed by a distinct effect on lymphocyte influx in states of allergic airway inflammation in vivo.
Collapse
Affiliation(s)
- Carolin Pilzner
- Department of Pneumology, Hannover Medical School, Hannover, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Total synthesis of grassystatin A, a probe for cathepsin E function. Bioorg Med Chem 2012; 20:4774-80. [PMID: 22748705 DOI: 10.1016/j.bmc.2012.05.077] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Revised: 05/30/2012] [Accepted: 05/30/2012] [Indexed: 11/21/2022]
Abstract
The linear depsipeptide grassystatin A, a valuable probe for the study of cathepsin E function, has been synthesized by a [4+6] strategy. It exhibited specific inhibitory activity against cathepsin E with an IC(50) value of 0.8 nM. Our studies indicated that inhibition of cathepsin E did not have an impact on ovalbumin antigen processing and peptide presentation, unique from studies of other aspartic protease inhibitors.
Collapse
|
46
|
Wagner CS, Grotzke JE, Cresswell P. Intracellular events regulating cross-presentation. Front Immunol 2012; 3:138. [PMID: 22675326 PMCID: PMC3366438 DOI: 10.3389/fimmu.2012.00138] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2012] [Accepted: 05/14/2012] [Indexed: 01/07/2023] Open
Abstract
Cross-presentation plays a fundamental role in the induction of CD8-T cell immunity. However, although more than three decades have passed since its discovery, surprisingly little is known about the exact mechanisms involved. Here we give an overview of the components involved at different stages of this process. First, antigens must be internalized into the cross-presenting cell. The involvement of different receptors, method of antigen uptake, and nature of the antigen can influence intracellular trafficking and access to the cross-presentation pathway. Once antigens access the endocytic system, different requirements for endosomal/phagosomal processing arise, such as proteolysis and reduction of disulfide bonds. The majority of cross-presented peptides are generated by proteasomal degradation. Therefore, antigens must cross a membrane barrier in a manner analogous to the fate of misfolded proteins in the endoplasmic reticulum (ER) that are retrotranslocated into the cytosol for degradation. Indeed, some components of the ER-associated degradation machinery have been implicated in cross-presentation. Further complicating the matter, endosomal and phagosomal compartments have been suggested as alternative sites to the ER for loading of peptides on major histocompatibility complex class I molecules. Finally, the antigen presenting cells involved, particularly dendritic cell subsets and their state of maturation, influence the efficiency of cross-presentation.
Collapse
Affiliation(s)
- Claudia S Wagner
- Department of Immunobiology, Yale University Medical Center, New Haven, CT, USA
| | | | | |
Collapse
|
47
|
Zhu X, Pan Y, Li Y, Jiang Y, Shang H, Gowda DC, Cui L, Cao Y. Targeting Toll-like receptors by chloroquine protects mice from experimental cerebral malaria. Int Immunopharmacol 2012; 13:392-7. [PMID: 22659438 DOI: 10.1016/j.intimp.2012.05.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Revised: 05/15/2012] [Accepted: 05/21/2012] [Indexed: 10/28/2022]
Abstract
Excessive production of proinflammatory cytokines, elicited mostly by Th1 cells, is an important cause of cerebral malaria (CM). Dendritic cells (DCs), a critical link between innate and adaptive immune responses, rely heavily on Toll-like receptor (TLR) signaling. Using C57BL/6 mice infected with Plasmodium berghei ANKA (PbA) as an experimental CM model, we first confirmed that inhibition of TLR9 by suppressive oligodeoxynucleotides protected mice from CM. In addition to being a well-known antimalarial, chloroquine (CQ) has been used as an immunomodulator of endocytic TLRs because it inhibits endosomal acidification. We found that immediately before and shortly after infection by PbA, treatment with a single dose of 50 mg/kg of CQ protected mice from experimental CM. Both CQ treatments significantly inhibited expression of TLR9 and MHC-II on DCs, and reduced the number of myeloid and plasmatocytoid DCs at 3 and 5 days after infection. Consequently, activation of CD4+ T cells, especially the expansion of the Th1 subsets, was dramatically inhibited in CQ treated groups, which was accompanied by a remarkable decline in the production of Th1 type proinflammatory mediators IFN-γ, TNF-α, and nitric oxide. Taken together, these results corroborated the involvement of TLR9 in CM pathogenesis and suggest that interference with the activation of this receptor is a promising strategy to prevent deleterious inflammatory response mediating pathogenesis and severity of malaria.
Collapse
Affiliation(s)
- Xiaotong Zhu
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, China
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Eisenlohr LC, Luckashenak N, Apcher S, Miller MA, Sinnathamby G. Beyond the classical: influenza virus and the elucidation of alternative MHC class II-restricted antigen processing pathways. Immunol Res 2012; 51:237-48. [PMID: 22101673 DOI: 10.1007/s12026-011-8257-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
CD4+ T cells (T(CD4+)) are activated by peptides, generally 13-17 amino acids in length, presented at the cell surface in combination with highly polymorphic MHC class II molecules. According to the classical model, these peptides are generated by endosomal digestion of internalized antigen and loaded onto MHC class II molecules in the late endosome. Historically, this "exogenous" pathway has been defined through the extensive use of purified proteins. However, the relatively recent use of clinically relevant antigens, those of influenza virus in our case, has revealed several additional pathways of peptide production, including some that are truly "endogenous", entailing synthesis of the protein within the infected cell. Indeed, some peptides appear to be created only via endogenous processing. The cell biology that underlies these alternative pathways remains poorly understood as do their relative contributions to defence against infectious agents and cancer, and the triggering of autoimmune diseases.
Collapse
Affiliation(s)
- Laurence C Eisenlohr
- Department of Microbiology and Immunology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA.
| | | | | | | | | |
Collapse
|
49
|
Gowda NM, Wu X, Gowda DC. TLR9 and MyD88 are crucial for the development of protective immunity to malaria. THE JOURNAL OF IMMUNOLOGY 2012; 188:5073-85. [PMID: 22516959 DOI: 10.4049/jimmunol.1102143] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Effective resolution of malaria infection by avoiding pathogenesis requires regulated pro- to anti-inflammatory responses and the development of protective immunity. TLRs are known to be critical for initiating innate immune responses, but their roles in the regulation of immune responses and development of protective immunity to malaria remain poorly understood. In this study, using wild-type, TLR2(-/-), TLR4(-/-), TLR9(-/-), and MyD88(-/-) mice infected with Plasmodium yoelii, we show that TLR9 and MyD88 regulate pro/anti-inflammatory cytokines, Th1/Th2 development, and cellular and humoral responses. Dendritic cells from TLR9(-/-) and MyD88(-/-) mice produced significantly lower levels of proinflammatory cytokines and higher levels of anti-inflammatory cytokines than dendritic cells from wild-type mice. NK and CD8(+) T cells from TLR9(-/-) and MyD88(-/-) mice showed markedly impaired cytotoxic activity. Furthermore, mice deficient in TLR9 and MyD88 showed higher Th2-type and lower Th1-type IgGs. Consequently, TLR9(-/-) and MyD88(-/-) mice exhibited compromised ability to control parasitemia and were susceptible to death. Our data also show that TLR9 and MyD88 distinctively regulate immune responses to malaria infection. TLR9(-/-) but not MyD88(-/-) mice produced significant levels of both pro- and anti-inflammatory cytokines, including IL-1β and IL-18, by other TLRs/inflammasome- and/or IL-1R/IL-18R-mediated signaling. Thus, whereas MyD88(-/-) mice completely lacked cell-mediated immunity, TLR9(-/-) mice showed low levels of cell-mediated immunity and were slightly more resistant to malaria infection than MyD88(-/-) mice. Overall, our findings demonstrate that TLR9 and MyD88 play central roles in the immune regulation and development of protective immunity to malaria, and have implications in understanding immune responses to other pathogens.
Collapse
Affiliation(s)
- Nagaraj M Gowda
- Department of Biochemistry and Molecular Biology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA.
| | | | | |
Collapse
|
50
|
Tse SW, Radtke AJ, Zavala F. Induction and maintenance of protective CD8+ T cells against malaria liver stages: implications for vaccine development. Mem Inst Oswaldo Cruz 2012; 106 Suppl 1:172-8. [PMID: 21881772 DOI: 10.1590/s0074-02762011000900022] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2011] [Accepted: 05/20/2011] [Indexed: 11/22/2022] Open
Abstract
CD8+ T cells against malaria liver stages represent a major protective immune mechanism against infection. Following induction in the peripheral lymph nodes by dendritic cells (DCs), these CD8+ T cells migrate to the liver and eliminate parasite infected hepatocytes. The processing and presentation of sporozoite antigen requires TAP mediated transport of major histocompatibility complex class I epitopes to the endoplasmic reticulum. Importantly, in DCs this process is also dependent on endosome-mediated cross presentation while this mechanism is not required for epitope presentation on hepatocytes. Protective CD8+ T cell responses are strongly dependent on the presence of CD4+ T cells and the capacity of sporozoite antigen to persist for a prolonged period of time. While human trials with subunit vaccines capable of inducing antibodies and CD4+ T cell responses have yielded encouraging results, an effective anti-malaria vaccine will likely require vaccine constructs designed to induce protective CD8+ T cells against malaria liver stages.
Collapse
Affiliation(s)
- Sze-Wah Tse
- W Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins Malaria Research Institute, Johns Hopkins University, Baltimore, Maryland, USA
| | | | | |
Collapse
|