1
|
Kumar R, Srivastava V. Application of anti-fungal vaccines as a tool against emerging anti-fungal resistance. FRONTIERS IN FUNGAL BIOLOGY 2023; 4:1241539. [PMID: 37746132 PMCID: PMC10512234 DOI: 10.3389/ffunb.2023.1241539] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 07/31/2023] [Indexed: 09/26/2023]
Abstract
After viruses and bacteria, fungal infections remain a serious threat to the survival and well-being of society. The continuous emergence of resistance against commonly used anti-fungal drugs is a serious concern. The eukaryotic nature of fungal cells makes the identification of novel anti-fungal agents slow and difficult. Increasing global temperature and a humid environment conducive to fungal growth may lead to a fungal endemic or a pandemic. The continuous increase in the population of immunocompromised individuals and falling immunity forced pharmaceutical companies to look for alternative strategies for better managing the global fungal burden. Prevention of infectious diseases by vaccines can be the right choice. Recent success and safe application of mRNA-based vaccines can play a crucial role in our quest to overcome anti-fungal resistance. Expressing fungal cell surface proteins in human subjects using mRNA technology may be sufficient to raise immune response to protect against future fungal infection. The success of mRNA-based anti-fungal vaccines will heavily depend on the identification of fungal surface proteins which are highly immunogenic and have no or least side effects in human subjects. The present review discusses why it is essential to look for anti-fungal vaccines and how vaccines, in general, and mRNA-based vaccines, in particular, can be the right choice in tackling the problem of rising anti-fungal resistance.
Collapse
Affiliation(s)
- Ravinder Kumar
- Department of Pathology, Collage of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Vartika Srivastava
- Department of Clinical Microbiology and Infectious Diseases, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, Gauteng, South Africa
| |
Collapse
|
2
|
Lin P, Zhang J, Xie G, Li J, Guo C, Lin H, Zhang Y. Innate Immune Responses to Sporothrix schenckii: Recognition and Elimination. Mycopathologia 2022; 188:71-86. [PMID: 36329281 DOI: 10.1007/s11046-022-00683-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 09/24/2022] [Indexed: 11/06/2022]
Abstract
Sporothrix schenckii (S. schenckii), a ubiquitous thermally dimorphic fungus, is the etiological agent of sporotrichosis, affecting immunocompromised and immunocompetent individuals. Despite current antifungal regimens, sporotrichosis results in prolonged treatment and significant mortality rates in the immunosuppressed population. The innate immune system forms the host's first and primary line of defense against S. schenckii, which has a bi-layered cell wall structure. Many components act as pathogen-associated molecular patterns (PAMPs) in pathogen-host interactions. PAMPs are recognized by pattern recognition receptors (PRRs) such as toll-like receptors, C-type lectin receptors, and complement receptors, triggering innate immune cells such as neutrophils, macrophages, and dendritic cells to phagocytize or produce mediators, contributing to S. schenckii elimination. The ultrastructure of S. schenckii and pathogen-host interactions, including PRRs and innate immune cells, are summarized in this review, promoting a better understanding of the innate immune response to S. schenckii and aiding in the development of protective and therapeutic strategies to combat sporotrichosis.
Collapse
Affiliation(s)
- Peng Lin
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jianfeng Zhang
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Guinan Xie
- Department of Traditional Chinese Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Junchen Li
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Chenqi Guo
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Haiyue Lin
- Graduate School, Tianjin Medical University, Tianjin, China
| | - Yu Zhang
- Department of Dermatology, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, China.
| |
Collapse
|
3
|
Zhao H, Zhao Q, Zhu S, Huang B, Lv L, Liu G, Li Z, Wang L, Dong H, Han H. Molecular characterization and immune protection of an AN1-like zinc finger protein of Eimeria tenella. Parasitol Res 2019; 119:623-635. [PMID: 31758298 DOI: 10.1007/s00436-019-06545-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 11/06/2019] [Indexed: 11/28/2022]
Abstract
Coccidiosis is caused by multiple species of the apicomplexan protozoa Eimeria. Among them, Eimeria tenella is frequently considered to be the most pathogenic. Zinc finger proteins (ZnFPs) are a type of protein containing zinc finger domains. In the present study, a putative Eimeria tenella AN1-like ZnFP (E. tenella AN1-like zinc finger domain-containing protein, putative partial mRNA, EtAN1-ZnFP) was cloned and characterized, and its immune protective effects were evaluated. The 798-bp ORF sequence of EtAN1-ZnFP that encoded a protein of approximately 27.0 kDa was obtained. The recombinant EtAN1-ZnFP protein (rEtAN1-ZnFP) was expressed in Escherichia coli. Western blot analysis showed that the recombinant protein was recognized by the anti-GST monoclonal antibody and anti-sporozoite protein rabbit serum. qPCR analysis revealed that EtAN1-ZnFP was highly expressed in unsporulated oocysts and sporozoites. Immunostaining with an anti-rEtAN1-ZnFP antibody indicated that EtAN1-ZnFP was uniformly distributed in the cytoplasm of sporozoites, except for the refractive body; furthermore, this protein was evenly distributed in the cytoplasm of immature schizonts but seldom distributed in mature schizonts. The results of the in vitro invasion inhibition assay indicated that the antibodies against rEtAN1-ZnFP efficiently reduced the ability of E. tenella sporozoites to invade host cells. Animal challenge experiments demonstrated that the chickens immunized with rEtAN1-ZnFP protein significantly decreased mean lesion scores and fecal oocyst output compared with challenged control group. The results suggest that EtAN1-ZnFP can induce partial immune protection against infection with E. tenella and could be an effective candidate for the development of new vaccines.
Collapse
Affiliation(s)
- Huanzhi Zhao
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, CAAS, Shanghai, 200241, China
| | - Qiping Zhao
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, CAAS, Shanghai, 200241, China
| | - Shunhai Zhu
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, CAAS, Shanghai, 200241, China
| | - Bing Huang
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, CAAS, Shanghai, 200241, China
| | - Ling Lv
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, CAAS, Shanghai, 200241, China
| | - Guiling Liu
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, CAAS, Shanghai, 200241, China.,College of Life and Environment Sciences, Shanghai Normal University, Shanghai, 200234, China
| | - Zhihang Li
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, CAAS, Shanghai, 200241, China.,College of Life and Environment Sciences, Shanghai Normal University, Shanghai, 200234, China
| | - Lu Wang
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, CAAS, Shanghai, 200241, China
| | - Hui Dong
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, CAAS, Shanghai, 200241, China
| | - Hongyu Han
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, CAAS, Shanghai, 200241, China.
| |
Collapse
|
4
|
Paulovičová L, Paulovičová E, Farkaš P, Čížová A, Bystrický P, Jančinová V, Turánek J, Pericolini E, Gabrielli E, Vecchiarelli A, Hrubiško M. Bioimmunological activities of Candida glabrata cellular mannan. FEMS Yeast Res 2019; 19:5303726. [PMID: 30689830 DOI: 10.1093/femsyr/foz009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 01/25/2019] [Indexed: 12/17/2022] Open
Abstract
Candida glabrata is a second most common human opportunistic pathogen which causes superficial but also life-threatening systemic candidosis. According to the localisation of mannans and mannoproteins in the outermost layer of the cell wall, mannan detection could be one of the first steps in the cell recognition of Candida cells by the host innate immune system. Mannans from the cell wall provide important immunomodulatory activities, comprising stimulation of cytokine production, induction of dendritic cells (DCs) maturation and T-cell immunity. The model of DCs represents a promising tool to study immunomodulatory interventions throughout the vaccine development. Activated DCs induce, activate and polarise T-cell responses by expression of distinct maturation markers and cytokines regulating the adaptive immune responses. In addition, they are uniquely adept at decoding the fungus-associated information and translate it in qualitatively different T helper responses. We find out, that C. glabrata mannan is able to induce proliferation of splenocytes and to increase the production of TNF-α and IL-4. Next, increased the expression of co-stimulatory molecules CD80 and CD86 and the proportion of CD4+CD25+ and CD4+CD28+ T cells during in vitro stimulation of splenocytes. Reported results provide C. glabrata mannan capability to modulate cytokine production, DCs activation and antigen presentation activity, influencing T-cell phenotype in response to stimulation.
Collapse
Affiliation(s)
- Lucia Paulovičová
- Institute of Chemistry, Center for Glycomics, Slovak Academy of Sciences, Dúbravská cesta 9, 845 38 Bratislava, Slovakia
| | - Ema Paulovičová
- Institute of Chemistry, Center for Glycomics, Slovak Academy of Sciences, Dúbravská cesta 9, 845 38 Bratislava, Slovakia
| | - Pavol Farkaš
- Institute of Chemistry, Center for Glycomics, Slovak Academy of Sciences, Dúbravská cesta 9, 845 38 Bratislava, Slovakia
| | - Alžbeta Čížová
- Institute of Chemistry, Center for Glycomics, Slovak Academy of Sciences, Dúbravská cesta 9, 845 38 Bratislava, Slovakia
| | - Peter Bystrický
- Division of Neurosciences, Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University, Malá Hora, 10701/4A, 036 01 Martin, Slovakia
| | - Viera Jančinová
- Institute of Experimental Pharmacology and Toxicology, Centre of Experimental Medicine, Slovak Academy of Sciences, Dúbravská cesta 9, 841 04 Bratislava, Slovakia
| | - Jaroslav Turánek
- Department of Pharmacology and Immunotherapy, Veterinary Research Institute, v.v.i., Hudcova 70, 621 00 Brno, Czech Republic
| | - Eva Pericolini
- Microbiology Section, Department of Medicine,University of Perugia, Sant' Andrea delle Fratte, 061 32 Perugia, Italy
- Department of Diagnostic, Clinic and Public Health Medicine, University of Modena and Reggio Emilia, 411 25 Modena, Italy
| | - Elena Gabrielli
- Microbiology Section, Department of Medicine,University of Perugia, Sant' Andrea delle Fratte, 061 32 Perugia, Italy
| | - Anna Vecchiarelli
- Microbiology Section, Department of Medicine,University of Perugia, Sant' Andrea delle Fratte, 061 32 Perugia, Italy
| | - Martin Hrubiško
- Department of Clinical Immunology and Allergy, Oncology Institute of St. Elisabeth, Heydukova 10, 812 50 Bratislava, Slovakia
| |
Collapse
|
5
|
Nami S, Mohammadi R, Vakili M, Khezripour K, Mirzaei H, Morovati H. Fungal vaccines, mechanism of actions and immunology: A comprehensive review. Biomed Pharmacother 2018; 109:333-344. [PMID: 30399567 DOI: 10.1016/j.biopha.2018.10.075] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 10/02/2018] [Accepted: 10/14/2018] [Indexed: 11/28/2022] Open
Abstract
Fungal infections include a wide range of opportunistic and invasive diseases. Two of four major fatal diseases in patients with human immunodeficiency virus (HIV) infection are related to the fungal infections, cryptococcosis, and pneumocystosis. Disseminated candidiasis and different clinical forms of aspergillosis annually impose expensive medical costs to governments and hospitalized patients and ultimately lead to high mortality rates. Therefore, urgent implementations are necessary to prevent the expansion of these diseases. Designing an effective vaccine is one of the most important approaches in this field. So far, numerous efforts have been carried out in developing an effective vaccine against fungal infections. Some of these challenges engaged in different stages of clinical trials but none of them could be approved by the United States Food and Drug Administration (FDA). Here, in addition to have a comprehensive overview on the data from studied vaccine programs, we will discuss the immunology response against fungal infections. Moreover, it will be attempted to clarify the underlying immune mechanisms of vaccines targeting different fungal infections that are crucial for designing an effective vaccination strategy.
Collapse
Affiliation(s)
- Sanam Nami
- Department of Medical Mycology and Parasitology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Rasoul Mohammadi
- Department of Medical Parasitology and Mycology, School of Medicine/Infectious Diseases and Tropical Medicine Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mahshid Vakili
- Department of Medical Mycology and Parasitology/Invasive Fungi Research Center (IFRC), School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Kimia Khezripour
- Department of Pharmacotherapy, School of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| | - Hamid Morovati
- Department of Medical Mycology and Parasitology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
6
|
Quinello C, Souza Ferreira L, Picolli I, Loesch ML, Portuondo DL, Batista-Duharte A, Zeppone Carlos I. Sporothrix schenckii Cell Wall Proteins-Stimulated BMDCs Are Able to Induce a Th1-Prone Cytokine Profile In Vitro. J Fungi (Basel) 2018; 4:E106. [PMID: 30200530 PMCID: PMC6162427 DOI: 10.3390/jof4030106] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Revised: 08/30/2018] [Accepted: 08/31/2018] [Indexed: 02/07/2023] Open
Abstract
Sporotrichosis is a subcutaneous mycosis affecting humans and other animals. The disease can be acquired by accidental inoculation of the fungus through the skin or through the respiratory system. Sporotrichosis can also be transmitted through bites or scratches by infected cats and more rarely by other animals (zoonotic transmission). Conventional antifungal therapy is especially inefficient in immunocompromised patients, who tend to develop the most severe forms of the disease, thus prompting the search for alternative therapies. Given their antigen-presenting properties, dendritic cells (DCs) have been used in both prophylactic and therapeutic vaccination strategies. Hence, this study aims to assess the use of DCs as a prophylactic tool in sporotrichosis by evaluating the immune profile induced by Sporothrix schenckii cell wall proteins (SsCWP)-stimulated, bone-marrow-derived DCs (BMDCs). Mouse BMDCs were stimulated with SsCWP for 24 h and analyzed for the surface expression of costimulatory molecules and TLR-4, as well as for the secretion of proinflammatory cytokines and IL-10. Following that, activated BMDCs were cocultured with splenocytes for 72 h and had the same cytokines measured in the supernatant. SsCWP-stimulated BMDCs showed higher expression of CD80, CD86, and CD40, but not TLR-4, and higher secretion of IL-6, IL-17A, and TNF. On the other hand, higher levels of IFN-γ, IL-10, and IL-2 were found in the supernatants of the coculture as compared with the BMDCs alone; TNF secretion was almost completely abrogated, whereas IL-6 was only partially inhibited and IL-17A was unaffected. Our results thus suggest that SsCWP-stimulated BMDCs are able to induce a Th1-prone cytokine profile which is known to be protective against other fungal diseases. This result could lead to studies which evaluate the development of prophylactic and/or therapeutic DC-based tools against sporotrichosis.
Collapse
Affiliation(s)
- Camila Quinello
- Department of Clinical Analysis, School of Pharmaceutical Sciences, Araraquara, São Paulo State University (UNESP), Rodovia Araraquara-Jaú-Km 1, Araraquara 14800-903, SP, Brazil.
| | - Lucas Souza Ferreira
- Department of Clinical Analysis, School of Pharmaceutical Sciences, Araraquara, São Paulo State University (UNESP), Rodovia Araraquara-Jaú-Km 1, Araraquara 14800-903, SP, Brazil.
| | - Isabella Picolli
- Department of Clinical Analysis, School of Pharmaceutical Sciences, Araraquara, São Paulo State University (UNESP), Rodovia Araraquara-Jaú-Km 1, Araraquara 14800-903, SP, Brazil.
| | - Maria Luiza Loesch
- Department of Clinical Analysis, School of Pharmaceutical Sciences, Araraquara, São Paulo State University (UNESP), Rodovia Araraquara-Jaú-Km 1, Araraquara 14800-903, SP, Brazil.
| | - Deivys Leandro Portuondo
- Department of Clinical Analysis, School of Pharmaceutical Sciences, Araraquara, São Paulo State University (UNESP), Rodovia Araraquara-Jaú-Km 1, Araraquara 14800-903, SP, Brazil.
| | - Alexander Batista-Duharte
- Department of Clinical Analysis, School of Pharmaceutical Sciences, Araraquara, São Paulo State University (UNESP), Rodovia Araraquara-Jaú-Km 1, Araraquara 14800-903, SP, Brazil.
| | - Iracilda Zeppone Carlos
- Department of Clinical Analysis, School of Pharmaceutical Sciences, Araraquara, São Paulo State University (UNESP), Rodovia Araraquara-Jaú-Km 1, Araraquara 14800-903, SP, Brazil.
| |
Collapse
|
7
|
Ledur PC, Tondolo JSM, Jesus FPK, Verdi CM, Loreto ÉS, Alves SH, Santurio JM. Dendritic cells pulsed with Pythium insidiosum (1,3)(1,6)-β-glucan, Heat-inactivated zoospores and immunotherapy prime naïve T cells to Th1 differentiation in vitro. Immunobiology 2017; 223:294-299. [PMID: 29074300 DOI: 10.1016/j.imbio.2017.10.033] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 10/14/2017] [Indexed: 11/30/2022]
Abstract
Pythiosis is a life-threatening disease caused by the fungus-like microorganism Pythium insidiosum that can lead to death if not treated. Since P. insidiosum has particular cell wall characteristics, pythiosis is difficult to treat, as it does not respond well to traditional antifungal drugs. In our study, we investigated a new immunotherapeutic approach with potential use in treatment and in the acquisition of immunity against pythiosis. Dendritic cells from both human and mouse, pulsed with P. insidiosum heat-inactivated zoospore, (1,3)(1,6)-β-glucan and the immunotherapeutic PitiumVac® efficiently induced naïve T cell differentiation in a Th1 phenotype by the activation of specific Th1 cytokine production in vitro. Heat-inactivated zoospores showed the greatest Th1 response among the tested groups, with a significant increase in IL-6 and IFN-γ production in human cells. In mice cells, we also observed a Th17 pathway induction, with an increase on the IL-17A levels in lymphocytes cultured with β-glucan pulsed DCs. These results suggest a potential use of DCs pulsed with P. insidiosum antigens as a new therapeutic strategy in the treatment and acquisition of immunity against pythiosis.
Collapse
Affiliation(s)
- Pauline C Ledur
- Programa de Pós-graduação em Farmacologia, Centro de Ciências da Saúde, Universidade Federal de Santa Maria, Santa Maria, Brazil
| | - Juliana S M Tondolo
- Programa de Pós-graduação em Farmacologia, Centro de Ciências da Saúde, Universidade Federal de Santa Maria, Santa Maria, Brazil
| | - Francielli P K Jesus
- Programa de Pós-Graduação em Ciências Farmacêuticas, Centro de Ciências da Saúde, Universidade Federal de Santa Maria, RS, Brazil
| | - Camila M Verdi
- Programa de Pós-graduação em Farmacologia, Centro de Ciências da Saúde, Universidade Federal de Santa Maria, Santa Maria, Brazil
| | - Érico S Loreto
- Programa de Pós-graduação em Farmacologia, Centro de Ciências da Saúde, Universidade Federal de Santa Maria, Santa Maria, Brazil
| | - Sydney H Alves
- Programa de Pós-Graduação em Química Orgânica, Laboratório de Processos Tecnológicos e Catálise, Universidade Federal do Rio Grande do Sul, RS, Brazil
| | - Janio M Santurio
- Programa de Pós-graduação em Farmacologia, Centro de Ciências da Saúde, Universidade Federal de Santa Maria, Santa Maria, Brazil.
| |
Collapse
|
8
|
|
9
|
Rodrigues ME, Silva S, Azeredo J, Henriques M. Novel strategies to fight Candida species infection. Crit Rev Microbiol 2014; 42:594-606. [PMID: 25383647 DOI: 10.3109/1040841x.2014.974500] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
In recent years, there has been a significant increase in the incidence of human fungal infections. The increase in cases of infection caused by Candida species, and the consequent excessive use of antimicrobials, has favored the emergence of resistance to conventional antifungal agents over the past decades. Consequently, Candida infections morbidity and mortality are also increasing. Therefore, new approaches are needed to improve the outcome of patients suffering from Candida infections, because it seems unlikely that the established standard treatments will drastically lower the morbidity of mucocutaneous Candida infections and the high mortality associated with invasive candidiasis. This review aims to present the last advances in the traditional antifungal therapy, and present an overview of novel strategies that are being explored for the treatment of Candida infections, with a special focus on combined antifungal agents, antifungal therapies with alternative compounds (plant extracts and essential oils), adjuvant immunotherapy, photodynamic therapy and laser therapy.
Collapse
Affiliation(s)
- Maria Elisa Rodrigues
- a CEB -- Centre of Biological Engineering, LIBRO -- Laboratório de Investigação em Biofilmes Rosário Oliveira, University of Minho , Braga , Portugal
| | - Sónia Silva
- a CEB -- Centre of Biological Engineering, LIBRO -- Laboratório de Investigação em Biofilmes Rosário Oliveira, University of Minho , Braga , Portugal
| | - Joana Azeredo
- a CEB -- Centre of Biological Engineering, LIBRO -- Laboratório de Investigação em Biofilmes Rosário Oliveira, University of Minho , Braga , Portugal
| | - Mariana Henriques
- a CEB -- Centre of Biological Engineering, LIBRO -- Laboratório de Investigação em Biofilmes Rosário Oliveira, University of Minho , Braga , Portugal
| |
Collapse
|
10
|
Cassone A, Torosantucci A. Opportunistic fungi and fungal infections: the challenge of a single, general antifungal vaccine. Expert Rev Vaccines 2014; 5:859-67. [PMID: 17184223 DOI: 10.1586/14760584.5.6.859] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
A vaccine made up by an algal beta-glucan (laminarin), conjugated with a protein component, protects against infections by different fungi and induces antibodies capable of inhibiting fungal growth. Although taking a premium on a common molecular theme, this remains a sort of 'cross-kingdom' vaccine because the immunizing antigen and the vaccination target belong to organisms from two different kingdoms and this is certainly the first case in the field of human vaccines. Thus, it is possible to convey in a single immunological tool the potential to protect against multiple infections, in theory all those caused by beta-glucan-expressing fungi. The generation of antibodies with the potential to directly inhibit the growth of, or kill the fungal cells also opens an exciting perspective for both active and passive vaccination in immunocompromised subjects.
Collapse
Affiliation(s)
- Antonio Cassone
- Istituto Superiore di Sanità, Department of Infectious, Parasitic and Immuno-mediated Diseases, Viale Regina Elena, Rome, Italy.
| | | |
Collapse
|
11
|
Hernández-Santos N, Huppler AR, Peterson AC, Khader SA, McKenna KC, Gaffen SL. Th17 cells confer long-term adaptive immunity to oral mucosal Candida albicans infections. Mucosal Immunol 2013; 6:900-10. [PMID: 23250275 PMCID: PMC3608691 DOI: 10.1038/mi.2012.128] [Citation(s) in RCA: 137] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Accepted: 11/13/2012] [Indexed: 02/04/2023]
Abstract
Oropharyngeal candidiasis (OPC) is an opportunistic infection caused by Candida albicans. Despite its prevalence, little is known about C. albicans-specific immunity in the oral mucosa. Vaccines against Candida generate both T helper type 1 (Th1) and Th17 responses, and considerable evidence implicates interleukin (IL)-17 in immunity to OPC. However, IL-17 is also produced by innate immune cells that are remarkably similar to Th17 cells, expressing the same markers and localizing to similar mucosal sites. To date, the relative contribution(s) of Th1, Th17, and innate IL-17-producing cells in OPC have not been clearly defined. Here, we sought to determine the nature and function of adaptive T-cell responses to OPC, using a new recall infection model. Mice subjected to infection and re-challenge with Candida mounted a robust and stable antigen-specific IL-17 response in CD4+ but not CD8+ T cells. There was little evidence for Th1 or Th1/Th17 responses. The Th17 response promoted accelerated fungal clearance, and Th17 cells could confer protection in Rag1-/- mice upon adoptive transfer. Surprisingly, CD4 deficiency did not cause OPC but was instead associated with compensatory IL-17 production by Tc17 and CD3+CD4-CD8- cells. Therefore, classic CD4+Th17 cells protect from OPC but can be compensated by other IL-17-producing cells in CD4-deficient hosts.
Collapse
Affiliation(s)
| | - Anna R. Huppler
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh
| | - Alanna C. Peterson
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh
| | | | | | - Sarah L. Gaffen
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh,Correspondence: Division of Rheumatology & Clinical Immunology, BST S703, 3500 Terrace St, Pittsburgh PA 15261, USA. 412-383-8903, Fax: 412-383-8864,
| |
Collapse
|
12
|
Huston SM, Li SS, Stack D, Timm-McCann M, Jones GJ, Islam A, Berenger BM, Xiang RF, Colarusso P, Mody CH. Cryptococcus gattii is killed by dendritic cells, but evades adaptive immunity by failing to induce dendritic cell maturation. THE JOURNAL OF IMMUNOLOGY 2013; 191:249-61. [PMID: 23740956 DOI: 10.4049/jimmunol.1202707] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
During adaptive immunity to pathogens, dendritic cells (DCs) capture, kill, process, and present microbial Ags to T cells. Ag presentation is accompanied by DC maturation driven by appropriate costimulatory signals. However, current understanding of the intricate regulation of these processes remains limited. Cryptococcus gattii, an emerging fungal pathogen in the Pacific Northwest of Canada and the United States, fails to stimulate an effective immune response in otherwise healthy hosts leading to morbidity or death. Because immunity to fungal pathogens requires intact cell-mediated immunity initiated by DCs, we asked whether C. gattii causes dysregulation of DC functions. C. gattii was efficiently bound and internalized by human monocyte-derived DCs, trafficked to late phagolysosomes, and killed. Yet, even with this degree of DC activation, the organism evaded pathways leading to DC maturation. Despite the ability to recognize and kill C. gattii, immature DCs failed to mature; there was no increased expression of MHC class II, CD86, CD83, CD80, and CCR7, or decrease of CD11c and CD32, which resulted in suboptimal T cell responses. Remarkably, no increase in TNF-α was observed in the presence of C. gattii. However, addition of recombinant TNF-α or stimulation that led to TNF-α production restored DC maturation and restored T cell responses. Thus, despite early killing, C. gattii evades DC maturation, providing a potential explanation for its ability to infect immunocompetent individuals. We have also established that DCs retain the ability to recognize and kill C. gattii without triggering TNF-α, suggesting independent or divergent activation pathways among essential DC functions.
Collapse
Affiliation(s)
- Shaunna M Huston
- Department of Microbiology and Infectious Disease, University of Calgary, Calgary, Alberta, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Abstract
Life-threatening fungal infections have increased in recent years while treatment options remain limited. The development of vaccines against fungal pathogens represents a key advance sorely needed to combat the increasing fungal disease threat. Dendritic cells (DC) are uniquely able to shape antifungal immunity by initiating and modulating naive T cell responses. Targeting DC may allow for the generation of potent vaccines against fungal pathogens. In the context of antifungal vaccine design, we describe the characteristics of the varied DC subsets, how DC recognize fungi, their function in immunity against fungal pathogens, and how DC can be targeted in order to create new antifungal vaccines. Ongoing studies continue to highlight the critical role of DC in antifungal immunity and will help guide DC-based vaccine strategies.
Collapse
|
14
|
From memory to antifungal vaccine design. Trends Immunol 2012; 33:467-74. [DOI: 10.1016/j.it.2012.04.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Revised: 04/18/2012] [Accepted: 04/23/2012] [Indexed: 01/21/2023]
|
15
|
Vecchiarelli A, Pericolini E, Gabrielli E, Pietrella D. New approaches in the development of a vaccine for mucosal candidiasis: progress and challenges. Front Microbiol 2012; 3:294. [PMID: 22905033 PMCID: PMC3417234 DOI: 10.3389/fmicb.2012.00294] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Accepted: 07/24/2012] [Indexed: 01/09/2023] Open
Abstract
The commensal fungus Candida albicans causes mucosal candidiasis in the rapidly expanding number of immunocompromised patients. Mucosal candidiasis includes oropharyngeal, esophageal, gastrointestinal, and vaginal infections. Vulvovaginal candidiasis (VVC) and antimycotic-refractory recurrent VVC is a frequent problem in healthy childbearing women. Both these mucosal infections can affect the quality of life and finding new therapeutical and preventive approaches is a challenge. A vaccine against candidal infections would be a new important tool to prevent and/or cure mucosal candidiasis and would be of benefit to many patients. Several Candida antigens have been proposed as vaccine candidates including cell wall components and virulence factors. Here we discuss the recent progress and problems associated with vaccination against mucosal candidiasis.
Collapse
Affiliation(s)
- Anna Vecchiarelli
- Microbiology Section, Department of Experimental Medicine and Biochemical Sciences, University of Perugia Perugia, Italy.
| | | | | | | |
Collapse
|
16
|
Li XL, Zheng PY, Li FG, Liu ZQ. Intestinal epithelial cell-derived integrin αVβ6 affects the function of dendritic cells. Shijie Huaren Xiaohua Zazhi 2012; 20:1553-1558. [DOI: 10.11569/wcjd.v20.i17.1553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the effect of intestinal epithelial cell (IEC)-derived integrin αVβ6 on the biological characteristics of bone marrow-derived dendritic cells (BMDCs).
METHODS: IECs and BMDCs were separated from BALB/c mice and cultured. After IECs were stimulated with ovalbumin (OVA), exosomes were prepared by multiple-step centrifugation. The expression of integrin αVβ6 in exosomes was examined by using the immune colloidal gold technique. Dendritic cells (DCs) were separated using immunomagnetic beads, and the concentration of DCs was determined by flow cytometry. DCs were then divided into five groups: blank group, OVA group, exosomes group, exosomes plus anti-αVβ6 antibody group, and exosomes plus goat anti-mouse IgG group. After these groups of DCs were treated with LPS, the expression of IL-12p70 was detected. In addition, the expression of active and total TGF-β1 was detected before LPS stimulation.
RESULTS: Compared to the blank group, the expression levels of total TGF-β1 increased (both P < 0.05) and those of active TGF-β1 showed no significant changes (both P > 0.05) in the OVA group and exosomes plus anti-αVβ6 antibody group; and the expression levels of both active and total TGF-β1 increased in the exosomes group and exosomes plus goat anti-mouse IgG group (both P < 0.05). Compared to the blank group, the expression of IL-12p70 was significantly reduced (both P < 0.05) in the exosomes group and exosomes plus goat anti-mouse IgG group, but showed no significant changes in the OVA group and exosomes plus anti-αVβ6 antibody group (both P > 0.05) 48 h after stimulation with LPS.
CONCLUSION: Intestinal epithelial cell-derived integrin αVβ6 can increase the expression of active TGF-β1 and total TGF-β1 in DCs and antagonize LPS-induced BMDC maturation.
Collapse
|
17
|
Liu M, Clemons KV, Johansen ME, Martinez M, Chen V, Stevens DA. Saccharomyces as a vaccine against systemic candidiasis. Immunol Invest 2012; 41:847-55. [PMID: 22686468 DOI: 10.3109/08820139.2012.692418] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
We have shown heat-killed Saccharomyces (HKY) is a protective vaccine against aspergillosis and coccidioidomycosis. To test the hypothesis that the efficacy of HKY- induced protection may be due to the cross-reactive antigens in the cell walls of the different fungi, we studied the effect of HKY against systemic candidiasis. Male CD-1 mice were given different regimens of HKY subcutaneously prior to intravenous challenge with Candida albicans. Compared to PBS controls, the administration of HKY (6 × 10(7)) 3, 4 or 6 times prolonged survival (all P < 0.05) and reduced fungal load in the kidney (all P < 0.05). An HKY dose of 1.2 × 10(8) given 4 times prolonged survival (P = 0.02), but showed dose-limiting toxicity. HKY given by an oral route, or by a subcutaneous route with alum as an adjuvant, did not improve survival. Overall, we found that HKY protects mice from infection by Candida albicans in a dose-and regimen-dependent manner. To understand the protection induced by HKY against different fungal species, additional studies of epitope mapping are warranted.
Collapse
Affiliation(s)
- Min Liu
- California Institute for Medical Research, San Jose, CA 95128, USA
| | | | | | | | | | | |
Collapse
|
18
|
Dendritic cell are able to differentially recognize Sporothrix schenckii antigens and promote Th1/Th17 response in vitro. Immunobiology 2012; 217:788-94. [PMID: 22656886 DOI: 10.1016/j.imbio.2012.04.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Revised: 04/26/2012] [Accepted: 04/27/2012] [Indexed: 12/24/2022]
Abstract
Sporotrichosis is a disease caused by the dimorphic fungus Sporothrix schenckii. The main clinical manifestations occur in the skin, however the number of systemic and visceral cases has increased, especially in immunocompromised patients. Dendritic cells (DCs) are highly capable to recognize the fungus associated data and translate it into differential T cells responses both in vivo and in vitro. Although, the mechanisms involved in the interaction between DCs and S. schenckii are not fully elucidated. The present study investigated the phenotypic and functional changes in bone marrow dendritic cells (BMDCs) stimulated in vitro with the yeast form of S. schenckii or exoantigen (ExoAg) and its ability to trigger a cellular immune response in vitro. Our results demonstrated that the live yeast of S. schenckii and its exoantigen, at a higher dose, were able to activate BMDCs and made them capable of triggering T cell responses in vitro. Whereas the yeast group promoted more pronounced IFN-γ production rather than IL-17, the Exo100 group generated similar production of both cytokines. The exoantigen stimulus suggests a capability to deviate the immune response from an effector Th1 to an inflammatory Th17 response. Interestingly, only the Exo100 group promoted the production of IL-6 and a significant increase of TGF-β, in addition to IL-23 production. Interestingly, only Exo100 group was capable to promote the production of IL-6 and a significant increase on TGF-β, in addition with IL-23 detection. Our results demonstrated the plasticity of DCs in translating the data associated with the fungus S. schenckii and ExoAg into differential T cell responses in vitro. The possibility of using ex vivo-generated DCs as vaccinal and therapeutic tools for sporotrichosis is a challenge for the future.
Collapse
|
19
|
Induction of protective immunity against Eimeria tenella, Eimeria maxima, and Eimeria acervulina infections using dendritic cell-derived exosomes. Infect Immun 2012; 80:1909-16. [PMID: 22354026 DOI: 10.1128/iai.06413-11] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
This study describes a novel immunization strategy against avian coccidiosis using exosomes derived from Eimeria parasite antigen (Ag)-loaded dendritic cells (DCs). Chicken intestinal DCs were isolated and pulsed in vitro with a mixture of sporozoite-extracted Ags from Eimeria tenella, E. maxima, and E. acervulina, and the cell-derived exosomes were isolated. Chickens were nonimmunized or immunized intramuscularly with exosomes and subsequently noninfected or coinfected with E. tenella, E. maxima, and E. acervulina oocysts. Immune parameters compared among the nonimmunized/noninfected, nonimmunized/infected, and immunized/infected groups were the numbers of cells secreting T(h)1 cytokines, T(h)2 cytokines, interleukin-16 (IL-16), and Ag-reactive antibodies in vitro and in vivo readouts of protective immunity against Eimeria infection. Cecal tonsils, Peyer's patches, and spleens of immunized and infected chickens had increased numbers of cells secreting the IL-16 and the T(h)1 cytokines IL-2 and gamma interferon, greater Ag-stimulated proliferative responses, and higher numbers of Ag-reactive IgG- and IgA-producing cells following in vitro stimulation with the sporozoite Ags compared with the nonimmunized/noninfected and nonimmunized/infected controls. In contrast, the numbers of cells secreting the T(h)2 cytokines IL-4 and IL-10 were diminished in immunized and infected chickens compared with the nonimmunized/noninfected and the nonimmunized/infected controls. Chickens immunized with Ag-loaded exosomes and infected in vivo with Eimeria oocysts had increased body weight gains, reduced feed conversion ratios, diminished fecal oocyst shedding, lessened intestinal lesion scores, and reduced mortality compared with the nonimmunized/infected controls. These results suggest that successful field vaccination against avian coccidiosis using exosomes derived from DCs incubated with Ags isolated from Eimeria species may be possible.
Collapse
|
20
|
van de Veerdonk FL, Kullberg BJ, Netea MG. Adjunctive immunotherapy with recombinant cytokines for the treatment of disseminated candidiasis. Clin Microbiol Infect 2011; 18:112-9. [PMID: 22032929 DOI: 10.1111/j.1469-0691.2011.03676.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Despite the discovery in the last decade of azoles and echinocandins as novel and potent antimycotic drugs, systemic Candida infections are still accompanied by an unacceptably high burden of morbidity and mortality. A rational novel therapeutic approach would be the use of adjuvant immunotherapy, with the aim of improving host defence against Candida. Increases in our understanding of the mechanisms that underlie the pathogenesis of Candida infections, such as the role played by pattern recognition receptors and the induction of proinflammatory cytokines during the early phases of infection, have led to the hypothesis of a potential therapeutic role of recombinant cytokines in systemic candidiasis. In the present review, we give an update of both experimental data and proof-of-principle studies in humans that argue for the use of adjunctive immunotherapy with recombinant cytokines in invasive Candida infections. Sufficiently powered studies on the role of cytokine-based treatment regimens for invasive candidiasis are needed to fully demonstrate the feasibility of this immunotherapeutic approach to improve the prognosis of severe invasive Candida infections.
Collapse
Affiliation(s)
- F L van de Veerdonk
- Nijmegen University Centre for Infectious Diseases, and Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | | | | |
Collapse
|
21
|
Kriengkauykiat J, Ito JI, Dadwal SS. Epidemiology and treatment approaches in management of invasive fungal infections. Clin Epidemiol 2011; 3:175-91. [PMID: 21750627 PMCID: PMC3130903 DOI: 10.2147/clep.s12502] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2011] [Indexed: 12/29/2022] Open
Abstract
Over the past 20 years, the number of invasive fungal infections has continued to persist, due primarily to the increased numbers of patients subjected to severe immunosuppression. Despite the development of more active, less toxic antifungal agents and the standard use of antifungal prophylaxis, invasive fungal infections (especially invasive mold infections) continue to be a significant factor in hematopoietic cell and solid organ transplantation outcomes, resulting in high mortality rates. Since the use of fluconazole as standard prophylaxis in the hematopoietic cell transplantation setting, invasive candidiasis has come under control, but no mold-active antifungal agent (except for posaconazole in the setting of acute myelogenous leukemia and myelodysplastic syndrome) has been shown to improve the survival rate over fluconazole. With the advent of new azole and echinocandin agents, we have seen the emergence of more azole-resistant and echinocandin-resistant fungi. The recent increase in zygomycosis seen in the hematopoietic cell transplantation setting may be due to the increased use of voriconazole. This has implications for the empiric approach to pulmonary invasive mold infections when zygomycosis cannot be ruled out. It is imperative that an amphotericin B product, an antifungal that has never developed resistance in over 50 years, be initiated. The clinical presentations of invasive mold infections and invasive candidiasis can be nonspecific and the diagnostic tests insensitive, so a high index of suspicion and immediate initiation of empiric therapy is required. Unfortunately, our currently available serologic tests do not predict infection ahead of disease, and, therefore cannot be used to initiate “preemptive” therapy. Also, the Aspergillus galactomannan test gives a false negative result in patients receiving antimold prophylaxis, ie, virtually all of our patients with hematologic malignancy and hematopoietic cell transplant recipients. We may eventually be able to select patients at highest risk for invasive fungal infections for prophylaxis by genetic testing. However, with our current armamentarium of antifungal agents and widespread use of prophylaxis in high-risk groups (hematologic malignancy, hematopoietic cell transplantation), we continue to see high incidence and mortality rates, and our future hope lies in reversing the immunosuppression or augmenting the immune system of these severely immunocompromised hosts by developing and utilizing immunotherapy, immunoprophylaxis, and vaccines.
Collapse
|
22
|
Induction of protective immunity against Eimeria tenella infection using antigen-loaded dendritic cells (DC) and DC-derived exosomes. Vaccine 2011; 29:3818-25. [DOI: 10.1016/j.vaccine.2011.03.022] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2011] [Revised: 02/20/2011] [Accepted: 03/03/2011] [Indexed: 11/20/2022]
|
23
|
CD4+ T cells mediate the protective effect of the recombinant Asp f3-based anti-aspergillosis vaccine. Infect Immun 2011; 79:2257-66. [PMID: 21422177 DOI: 10.1128/iai.01311-10] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The mortality and morbidity caused by invasive aspergillosis present a major obstacle to the successful treatment of blood cancers with hematopoietic cell transplants. Patients who receive hematopoietic cell transplants are usually immunosuppressed for extended periods, and infection with the ubiquitous mold Aspergillus fumigatus is responsible for most cases of aspergillosis. Previously, we demonstrated that vaccination with recombinant forms of the A. fumigatus protein Asp f3 protected cortisone acetate-immunosuppressed mice from experimentally induced pulmonary aspergillosis. Here, we investigated the vaccine's protective mechanism and evaluated in particular the roles of antibodies and T cells. After vaccination, Asp f3-specific preinfection IgG titers did not significantly differ between surviving and nonsurviving mice, and passive transfer of anti-Asp f3 antibodies did not protect immunosuppressed recipients from aspergillosis. We experimentally confirmed Asp f3's predicted peroxisomal localization in A. fumigatus hyphae. We found that fungal Asp f3 is inaccessible to antibodies, unless both cell walls and membranes have been permeabilized. Antibody-induced depletion of CD4+ T cells reduced the survival of recombinant Asp f3 (rAsp f3)-vaccinated mice to nonimmune levels, and transplantation of purified CD4+ T cells from rAsp f3-vaccinated mice into nonimmunized recipients transferred antifungal protection. In addition, residues 60 to 79 and 75 to 94 of Asp f3 contain epitopes that induce proliferation of T cells from vaccinated survivors. Vaccine-primed CD4+ T cells are not expected to clear the fungal pathogen directly; however, they may locally activate immunosuppressed phagocytes that elicit the antifungal effect.
Collapse
|
24
|
van de Veerdonk FL, Netea MG, Joosten LA, van der Meer JWM, Kullberg BJ. Novel strategies for the prevention and treatment of Candida infections: the potential of immunotherapy. FEMS Microbiol Rev 2011; 34:1063-75. [PMID: 20528948 DOI: 10.1111/j.1574-6976.2010.00232.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Infections caused by Candida spp. continue to be a substantial cause of disease burden, especially in immunocompromised patients. New approaches are needed to improve the outcome of patients suffering from Candida infections, because it seems unlikely that the established standard treatment will drastically lower the morbidity of mucocutaneous Candida infections and the high mortality associated with invasive candidiasis. New insights into the mechanisms of the anti-Candida host response have contributed to the design of novel immunotherapeutic approaches that have been proposed as adjuvant therapy in Candida infections. This review presents an overview of novel strategies in the prevention and treatment of Candida infections, with a special focus on adjuvant immunotherapy.
Collapse
Affiliation(s)
- Frank L van de Veerdonk
- Department of Medicine, Radboud University Nijmegen Medical Center, Nijmegen Institute for Infection, Inflammation and Immunity (N4i), Nijmegen, The Netherlands.
| | | | | | | | | |
Collapse
|
25
|
Tongchusak S, Leelayuwat C, Brusic V, Chaiyaroj SC. In silico prediction and immunological validation of common HLA-DRB1-restricted T cell epitopes of Candida albicans secretory aspartyl proteinase 2. Microbiol Immunol 2008; 52:231-42. [PMID: 18426398 DOI: 10.1111/j.1348-0421.2008.00032.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
ABSTRACT Sap2 is the most abundant virulence factor expressed during Candida infection, and the principal protein known to induce antibody response during Candida infection in humans. Its role in T-cell activation however, has not yet been determined. Sequence analysis revealed that Sap2 contains two variable regions: Var1 and Var2. Computational predictions by the Hotspot Hunter program identified that Var1 contains three candidate T-cell epitopes, whereas Var2 contains four. Thirty-nine overlapping peptides of Sap2 were then synthesized, and tested for their ability to induce proliferation of PBMC from 12 donors. Peptides P11, P17 and P31 exhibited significantly higher proliferative indices when compared with those of other peptides or controls. P17 and P31 are located in the areas of prediction, while P11 is not. There were other peptides outside the prediction areas that could stimulate PBMC proliferation at low levels. Nevertheless, the proliferative noise caused by such peptides was ruled out by IL-2 ELISpot analysis. Only P17 and P31 were shown to induce clonal proliferation of IFN-gamma producing lymphocytes, suggesting that these two peptides contain T cell epitopes. P11, which stimulated IL-2 producing clones, contains a known B-cell epitope. Interestingly, P17 and P31 elicited both Th1 and Th2 cell responses with significant numbers of IL-13 secreting clones in response to stimulation. Taken together, the computer-based T cell epitope prediction method could identify the immunogenic T cell epitopes of C. albicans Sap2 that promiscuously bind to the HLA-DRB1 supertype.
Collapse
Affiliation(s)
- Songsak Tongchusak
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | | | | | | |
Collapse
|
26
|
Boneberger S, Korting H. Therapeutische oder prophylaktische Vakzination bei Pilzinfektionen. Hautarzt 2008; 59:828-32. [DOI: 10.1007/s00105-008-1582-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
27
|
Cai L, Zeng T, Zeng Q, Li B, Lin X, Gong Y, Liu W, Zhang Z, Zhang S. Schistosoma japonicum: protective immunity induced by schistosomulum-derived cells in a mouse model. J Parasitol 2008; 94:395-403. [PMID: 18564740 DOI: 10.1645/ge-1315.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
We previously reported that immunization with intact live cells from schistosomula of Schistosoma japonicum (S.j) partially protected the Kunming strain of mice from challenge infection. In the present work, 2 immune protective experiments were designed to further validate the protective effect induced by this type of vaccine and to optimize the immunization protocol, including the number of inoculations and parasite stages from which immunogenic cells were derived. Three antigens derived from 18-day-old postinfection live (LLC) and dead (DLC) larval worm cells and from dead 42-day-old postinfection adult worm cells (DAC) were used as immunogens. Our results demonstrate that live cells from 18-day-old worms are capable of inducing significant protection in mice using a murine-Sj challenge model as shown by reduction rates of worm recoveries and egg burdens. The development of adult worms was stunted. A Th1-biased immune response was reflected in the protected groups as evidenced by the ratio of IgG2a/IgG1. A 38-kDa polypeptide was recognized by sera from LLC immunized animals. We demonstrate that live parasite cells are a source of novel protective antigens that can be exploited for vaccine development.
Collapse
Affiliation(s)
- Liting Cai
- Cell & Molecular Biological Experiment Center, Department of Parasitology, Xiangya School of Medicine, Central South University, Tongzipo Road 172, Changsha, Hunan 410013, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Hamad M. Antifungal Immunotherapy and Immunomodulation: A Double-hitter Approach to Deal with Invasive Fungal Infections. Scand J Immunol 2008; 67:533-43. [DOI: 10.1111/j.1365-3083.2008.02101.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
29
|
Wu K, Bi Y, Sun K, Xia J, Wang Y, Wang C. Suppression of allergic inflammation by allergen-DNA-modified dendritic cells depends on the induction of Foxp3+ Regulatory T cells. Scand J Immunol 2008; 67:140-51. [PMID: 18201369 DOI: 10.1111/j.1365-3083.2007.02050.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
CD4(+)CD25(+)Foxp3(+)Regulatory T cells (Tregs) play important roles in regulating allergic inflammation. To analyse if allergen-DNA-modified dendritic cells (DC) can suppress allergic responses and what roles Treg cells play in DC-based allergen-specific immunotherapy. Immature DC were transfected with retrovirus encoding Der p2 DNA, and administered to mice that sensitized and challenged with Der p2 protein. After Treg cells were depleted with anti-CD25 mAb, mice were re-challenged to observe the airway inflammation, and Treg cells in spleen CD4(+) T cells. And responses of spleen CD4(+) T cells to Der p2 were determined. Co-culture of naïve CD4(+) T cells with allergen-modified DC induced Foxp3+ Tregs. Sensitized and challenged mice developed allergic airway inflammation and Th2 responses, and decreased Foxp3(+) Tregs. Treatment with allergen-modified-DC suppressed airway inflammation and Th2 responses, and increased IL-10 and IFN-gamma production and Foxp3(+) Tregs significantly; and eliminated the responses of CD4(+) T cells to allergen. Administration of anit-CD25 mAb eliminated all the effects of modified-DC except for the increasing of IFN-gamma. Allergen-modified DC can induce immune tolerance to allergens and reverse the established Th2 responses induced by allergen, with dependence on the induction of Foxp3(+) Tregs.
Collapse
Affiliation(s)
- Kui Wu
- Institute of Respiratory Medicine, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | | | | | | | | | | |
Collapse
|
30
|
Fungal vaccines: real progress from real challenges. THE LANCET. INFECTIOUS DISEASES 2008; 8:114-24. [DOI: 10.1016/s1473-3099(08)70016-1] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
31
|
Role of Macrophages in Host Defense Against Aspergillosis and Strategies for Immune Augmentation. Oncologist 2007; 12 Suppl 2:7-13. [DOI: 10.1634/theoncologist.12-s2-7] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
32
|
Cassone A, De Bernardis F, Santoni G. Anticandidal immunity and vaginitis: novel opportunities for immune intervention. Infect Immun 2007; 75:4675-86. [PMID: 17562759 PMCID: PMC2044548 DOI: 10.1128/iai.00083-07] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Affiliation(s)
- Antonio Cassone
- Department of Infectious, Parasitic and Immunomediated Diseases, Istituto Superiore di Sanità, Viale Regina Elena, 299, 00161 Rome, Italy.
| | | | | |
Collapse
|
33
|
Abstract
Coccidioides causes coccidioidomycosis in the southwestern United States. Its clinical manifestations range from the primary asymptomatic to progressive pulmonary and extrapulmonary disease. Because of endemicity, frequent relapse, and virulent nature of Coccidioides, there is an urgent need for the development of effective therapy or vaccine. It has been recognized from studies in human patients and in murine models that the divergence in their susceptibility to Coccidioides infection is related to differences in T cell response. Dendritic cells (DCs) are most potent antigen-presenting cells that play a critical role in activating naïve T cells. On account of their unique immunostimulatory capacity, DCs have been used for the development of immunotherapy and vaccines against cancer and infectious diseases. We recently investigated the immunostimulatory potential of a DC-based vaccine in a murine model against Coccidioides posadasii (C. posadasii). Our results suggest that DCs act as a potent adjuvant and activate protective responses in mice against C. posadasii.
Collapse
Affiliation(s)
- Shanjana Awasthi
- Department of Pharmaceutical Sciences, 1110 North Stonewall Avenue, Oklahoma City, OK 73117, USA.
| |
Collapse
|
34
|
Cutler JE, Deepe GS, Klein BS. Advances in combating fungal diseases: vaccines on the threshold. Nat Rev Microbiol 2007; 5:13-28. [PMID: 17160002 PMCID: PMC2214303 DOI: 10.1038/nrmicro1537] [Citation(s) in RCA: 160] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The dramatic increase in fungal diseases in recent years can be attributed to the increased aggressiveness of medical therapy and other human activities. Immunosuppressed patients are at risk of contracting fungal diseases in healthcare settings and from natural environments. Increased prescribing of antifungals has led to the emergence of resistant fungi, resulting in treatment challenges. These concerns, together with the elucidation of the mechanisms of protective immunity against fungal diseases, have renewed interest in the development of vaccines against the mycoses. Most research has used murine models of human disease and, as we review in this article, the knowledge gained from these studies has advanced to the point where the development of vaccines targeting human fungal pathogens is now a realistic and achievable goal.
Collapse
Affiliation(s)
- Jim E. Cutler
- Departments of Pediatrics and Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences, and Research Institute for Children at Children’s Hospital, New Orleans, Louisiana, 70118 USA
| | - George S. Deepe
- Division of Infectious Diseases, University of Cincinnati College of Medicine, Cincinnati, Ohio, 45267–0560 USA
| | - Bruce S. Klein
- Departments of Pediatrics, Internal Medicine, and Medical Microbiology and Immunology and the University of Wisconsin Comprehensive Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, 53792 USA
| |
Collapse
|
35
|
Fungal Vaccines and Vaccination: Problems and Perspectives. IMMUNOLOGY OF FUNGAL INFECTIONS 2007. [PMCID: PMC7121605 DOI: 10.1007/1-4020-5492-0_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Vaccines against human pathogenic fungi, a rather neglected medical need until few years ago, are now gaining steps in the public health priority scale. The awareness of the rising medical threat represented by the opportunistic fungal infections among the health care-associated infections, the advances in the knowledge of fungal pathogenicity and immune response and the extraordinary progress of biotechnology have generated enthusiasm and critical new tools for active and passive anti-fungal vaccination. The discovery that antibodies play a critical role for protection against fungal infection has greatly contributed to the advancements in this field, in recognition that almost all useful vaccines against viral and bacterial pathogens owe their protective efficacy to neutralizing, opsonizing or otherwise effective antibodies. Overall, there is more hope now than few years ago about the chances of generating and having approved by the regulatory authorities one or more antifungal vaccines, be active or passive, for use in humans in the next few years. In particular, the possibility of protecting against multiple opportunistic mycoses in immuno-depressed subjects with a single, well-defined glucan-conjugate vaccine eliciting directly anti-fungal antibodies may be an important step to achieve this public health goal
Collapse
|
36
|
Deepe GS. Preventative and therapeutic vaccines for fungal infections: from concept to implementation. Expert Rev Vaccines 2006; 3:701-9. [PMID: 15606355 DOI: 10.1586/14760584.3.6.701] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Many challenges confront the development of fungal vaccines for humans including differences in host susceptibility, varied pathogenic mechanisms employed by the different species of fungi and mechanisms of host resistance. Hence, no single antigen can be expected to serve as a pan fungal vaccine. Instead, it is likely that progress for fungal vaccines will have to be made at the level of each individual organism. In recent years, tremendous strides have been made in understanding the immunopathogenesis of medically important fungal infections and identifying putative vaccine candidates. Such discoveries will facilitate the introduction of fungal vaccines into the therapeutic armamentarium of clinicians. The fungi under discussion in this review include Candida spp., Aspergillus spp., Cryptococcus neoformans, Coccidioides spp., Histoplasma capsulatum, Blastomyces dermatitidis, Paracoccidioides brasiliensis and Pneumocystis jirovecii.
Collapse
Affiliation(s)
- George S Deepe
- University of Cincinnati College of Medicine, Division of Infectious Diseases, Cincinnati, Ohio 45267-0560, USA.
| |
Collapse
|
37
|
Ito JI, Lyons JM, Hong TB, Tamae D, Liu YK, Wilczynski SP, Kalkum M. Vaccinations with recombinant variants of Aspergillus fumigatus allergen Asp f 3 protect mice against invasive aspergillosis. Infect Immun 2006; 74:5075-84. [PMID: 16926399 PMCID: PMC1594836 DOI: 10.1128/iai.00815-06] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
A vaccine that effectively protects immunocompromised patients against invasive aspergillosis is a novel approach to a universally fatal disease. Here we present a rationale for selection and in vivo testing of potential protein vaccine candidates, based on the modification of an immunodominant fungal allergen for which we demonstrate immunoprotective properties. Pulmonary exposure to viable Aspergillus fumigatus conidia as well as vaccination with crude hyphal extracts protects corticosteroid-immunosuppressed mice against invasive aspergillosis (J. I. Ito and J. M. Lyons, J. Infect. Dis. 186:869-871, 2002). Sera from the latter animals contain antibodies with numerous and diverse antigen specificities, whereas sera from conidium-exposed mice contain antibodies predominantly against allergen Asp f 3 (and some against Asp f 1), as identified by mass spectrometry. Subcutaneous immunization with recombinant Asp f 3 (rAsp f 3) but not with Asp f 1 was protective. The lungs of Asp f 3-vaccinated survivors were free of hyphae and showed only a patchy low-density infiltrate of mononuclear cells. In contrast, the nonimmunized animals died with invasive hyphal elements and a compact peribronchial infiltrate of predominantly polymorphonuclear leukocytes. Three truncated versions of rAsp f 3, spanning amino acid residues 15 to 168 [rAsp f 3(15-168)], 1 to 142, and 15 to 142 and lacking the known bipartite sequence required for IgE binding, were also shown to be protective. Remarkably, vaccination with either rAsp f 3(1-142) or rAsp f 3(15-168) drastically diminished the production of antigen-specific antibodies compared to vaccination with the full-length rAsp f 3(1-168) or the double-truncated rAsp f 3(15-142) version. Our findings point to a possible mechanism in which Asp f 3 vaccination induces a cellular immune response that upon infection results in the activation of lymphocytes that in turn enhances and/or restores the function of corticosteroid-suppressed macrophages to clear fungal elements in the lungs.
Collapse
Affiliation(s)
- James I Ito
- Immunology Division, City of Hope National Medical Center and Beckman Research Institute, 1500 E. Duarte Road, Duarte, CA 91010-3000, USA
| | | | | | | | | | | | | |
Collapse
|
38
|
|
39
|
Zelante T, Montagnoli C, Bozza S, Bellocchio S, Bonifazi P, De Luca A, Bistoni G, Mosci P, Romani L. Manipulating immunity against Aspergillus fumigatus. Med Mycol 2006; 44:S237-S243. [PMID: 30408910 DOI: 10.1080/13693780600778668] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
Efficient response to Aspergillusfumigatus requires different mechanisms. Polymorphonuclear neutrophils (PMNs) are the predominant immune cells in the acute stage of most fungal infections and play a crucial role in determining the type of pathology associated with fungal infections in different clinical settings. Dendritic cells (DC) are able to decode the fungus-associated information and translate it into different T helper (Th) and regulatory (Treg) cell responses. Functionally distinct Treg cells are activated after exposure to Aspergillus conidia. Early in infection, inflammation/Th1 reactivity is controlled by Treg cells suppressing PMNs and the immunogenic program of DC. The levels of IFN-γ produced in this phase set the subsequent adaptive stage by conditioning the indoleamine 2, 3-dioxygenase (IDO)-dependent tolerogenic program of DC and the subsequent activation of tolerogenic Treg cells, which inhibit Th2 cells and prevent allergy to the fungus. Knowledge of the immunopathogenesis of Aspergillus infections may pave the way to promising strategies for immunotherapy.
Collapse
Affiliation(s)
- Teresa Zelante
- Department of Experimental Medicine and Biochemical Science, University of Perugia, Perugia, Italy
| | - Claudia Montagnoli
- Department of Experimental Medicine and Biochemical Science, University of Perugia, Perugia, Italy
| | - Silvia Bozza
- Department of Experimental Medicine and Biochemical Science, University of Perugia, Perugia, Italy
| | - Silvia Bellocchio
- Department of Experimental Medicine and Biochemical Science, University of Perugia, Perugia, Italy
| | - Pierluigi Bonifazi
- Department of Experimental Medicine and Biochemical Science, University of Perugia, Perugia, Italy
| | - Antonella De Luca
- Department of Experimental Medicine and Biochemical Science, University of Perugia, Perugia, Italy
| | - Giovanni Bistoni
- Department of Experimental Medicine and Biochemical Science, University of Perugia, Perugia, Italy
| | - Paolo Mosci
- Department of Experimental Medicine and Biochemical Science, University of Perugia, Perugia, Italy
| | - Luigina Romani
- Department of Experimental Medicine and Biochemical Science, University of Perugia, Perugia, Italy
| |
Collapse
|
40
|
Dillon S, Agrawal S, Banerjee K, Letterio J, Denning TL, Oswald-Richter K, Kasprowicz DJ, Kellar K, Pare J, van Dyke T, Ziegler S, Unutmaz D, Pulendran B. Yeast zymosan, a stimulus for TLR2 and dectin-1, induces regulatory antigen-presenting cells and immunological tolerance. J Clin Invest 2006; 116:916-28. [PMID: 16543948 PMCID: PMC1401484 DOI: 10.1172/jci27203] [Citation(s) in RCA: 402] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2005] [Accepted: 01/24/2006] [Indexed: 01/08/2023] Open
Abstract
Emerging evidence suggests critical roles for APCs in suppressing immune responses. Here, we show that zymosan, a stimulus for TLR2 and dectin-1, regulates cytokine secretion in DCs and macrophages to induce immunological tolerance. First, zymosan induces DCs to secrete abundant IL-10 but little IL-6 and IL-12(p70). Induction of IL-10 is dependent on TLR2- and dectin-1-mediated activation of ERK MAPK via a mechanism independent of the activation protein 1 (AP-1) transcription factor c-Fos. Such DCs stimulate antigen-specific CD4+ T cells poorly due to IL-10 and the lack of IL-6. Second, zymosan induces F4-80+ macrophages in the splenic red pulp to secrete TGF-beta. Consistent with these effects on APCs, injection of zymosan plus OVA into mice results in OVA-specific T cells that secrete little or no Th1 or Th2 cytokines, but secrete robust levels of IL-10, and are unresponsive to challenge with OVA plus adjuvant. Finally, coinjection of zymosan with OVA plus LPS suppresses the response to OVA via a mechanism dependent on IL-10, TGF-beta, and lack of IL-6. Together, our data demonstrate that zymosan stimulates IL-10+ IL-12(p70)- IL-6low regulatory DCs and TGF-beta+ macrophages to induce immunological tolerance. These data suggest several targets for pharmacological modulation of immune responses in various clinical settings.
Collapse
Affiliation(s)
- Stephanie Dillon
- Emory Vaccine Research Center and Department of Pathology, Emory University, Atlanta, Georgia, USA.
Immunoregulation Section, Lab of Cell Regulation and Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Department of Microbiology and Immunology, Vanderbilt University Medical School, Nashville, Tennessee, USA.
Immunology Program, Benaroya Research Institute, Seattle, Washington, USA.
Scientific Resources Program, National Center for Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA.
Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.
Department of Periodontology and Oral Biology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Sudhanshu Agrawal
- Emory Vaccine Research Center and Department of Pathology, Emory University, Atlanta, Georgia, USA.
Immunoregulation Section, Lab of Cell Regulation and Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Department of Microbiology and Immunology, Vanderbilt University Medical School, Nashville, Tennessee, USA.
Immunology Program, Benaroya Research Institute, Seattle, Washington, USA.
Scientific Resources Program, National Center for Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA.
Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.
Department of Periodontology and Oral Biology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Kaustuv Banerjee
- Emory Vaccine Research Center and Department of Pathology, Emory University, Atlanta, Georgia, USA.
Immunoregulation Section, Lab of Cell Regulation and Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Department of Microbiology and Immunology, Vanderbilt University Medical School, Nashville, Tennessee, USA.
Immunology Program, Benaroya Research Institute, Seattle, Washington, USA.
Scientific Resources Program, National Center for Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA.
Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.
Department of Periodontology and Oral Biology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - John Letterio
- Emory Vaccine Research Center and Department of Pathology, Emory University, Atlanta, Georgia, USA.
Immunoregulation Section, Lab of Cell Regulation and Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Department of Microbiology and Immunology, Vanderbilt University Medical School, Nashville, Tennessee, USA.
Immunology Program, Benaroya Research Institute, Seattle, Washington, USA.
Scientific Resources Program, National Center for Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA.
Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.
Department of Periodontology and Oral Biology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Timothy L. Denning
- Emory Vaccine Research Center and Department of Pathology, Emory University, Atlanta, Georgia, USA.
Immunoregulation Section, Lab of Cell Regulation and Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Department of Microbiology and Immunology, Vanderbilt University Medical School, Nashville, Tennessee, USA.
Immunology Program, Benaroya Research Institute, Seattle, Washington, USA.
Scientific Resources Program, National Center for Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA.
Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.
Department of Periodontology and Oral Biology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Kyra Oswald-Richter
- Emory Vaccine Research Center and Department of Pathology, Emory University, Atlanta, Georgia, USA.
Immunoregulation Section, Lab of Cell Regulation and Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Department of Microbiology and Immunology, Vanderbilt University Medical School, Nashville, Tennessee, USA.
Immunology Program, Benaroya Research Institute, Seattle, Washington, USA.
Scientific Resources Program, National Center for Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA.
Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.
Department of Periodontology and Oral Biology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Deborah J. Kasprowicz
- Emory Vaccine Research Center and Department of Pathology, Emory University, Atlanta, Georgia, USA.
Immunoregulation Section, Lab of Cell Regulation and Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Department of Microbiology and Immunology, Vanderbilt University Medical School, Nashville, Tennessee, USA.
Immunology Program, Benaroya Research Institute, Seattle, Washington, USA.
Scientific Resources Program, National Center for Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA.
Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.
Department of Periodontology and Oral Biology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Kathryn Kellar
- Emory Vaccine Research Center and Department of Pathology, Emory University, Atlanta, Georgia, USA.
Immunoregulation Section, Lab of Cell Regulation and Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Department of Microbiology and Immunology, Vanderbilt University Medical School, Nashville, Tennessee, USA.
Immunology Program, Benaroya Research Institute, Seattle, Washington, USA.
Scientific Resources Program, National Center for Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA.
Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.
Department of Periodontology and Oral Biology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Jeff Pare
- Emory Vaccine Research Center and Department of Pathology, Emory University, Atlanta, Georgia, USA.
Immunoregulation Section, Lab of Cell Regulation and Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Department of Microbiology and Immunology, Vanderbilt University Medical School, Nashville, Tennessee, USA.
Immunology Program, Benaroya Research Institute, Seattle, Washington, USA.
Scientific Resources Program, National Center for Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA.
Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.
Department of Periodontology and Oral Biology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Thomas van Dyke
- Emory Vaccine Research Center and Department of Pathology, Emory University, Atlanta, Georgia, USA.
Immunoregulation Section, Lab of Cell Regulation and Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Department of Microbiology and Immunology, Vanderbilt University Medical School, Nashville, Tennessee, USA.
Immunology Program, Benaroya Research Institute, Seattle, Washington, USA.
Scientific Resources Program, National Center for Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA.
Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.
Department of Periodontology and Oral Biology, Boston University School of Medicine, Boston, Massachusetts, USA
| | | | - Derya Unutmaz
- Emory Vaccine Research Center and Department of Pathology, Emory University, Atlanta, Georgia, USA.
Immunoregulation Section, Lab of Cell Regulation and Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Department of Microbiology and Immunology, Vanderbilt University Medical School, Nashville, Tennessee, USA.
Immunology Program, Benaroya Research Institute, Seattle, Washington, USA.
Scientific Resources Program, National Center for Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA.
Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.
Department of Periodontology and Oral Biology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Bali Pulendran
- Emory Vaccine Research Center and Department of Pathology, Emory University, Atlanta, Georgia, USA.
Immunoregulation Section, Lab of Cell Regulation and Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Department of Microbiology and Immunology, Vanderbilt University Medical School, Nashville, Tennessee, USA.
Immunology Program, Benaroya Research Institute, Seattle, Washington, USA.
Scientific Resources Program, National Center for Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA.
Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.
Department of Periodontology and Oral Biology, Boston University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
41
|
Segal BH, Walsh TJ. Current Approaches to Diagnosis and Treatment of Invasive Aspergillosis. Am J Respir Crit Care Med 2006; 173:707-17. [PMID: 16387806 DOI: 10.1164/rccm.200505-727so] [Citation(s) in RCA: 208] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Filamentous fungi (moulds) are ubiquitous soil inhabitants whose conidia are inhaled into the respiratory tract, where they may cause life-threatening infections. Among these infections is invasive aspergillosis, which is a major cause of morbidity and mortality in the severely immunocompromised. Risk factors for invasive aspergillosis include prolonged and severe neutropenia, hematopoietic stem cell and solid organ transplantation, advanced AIDS, and chronic granulomatous disease. Invasive aspergillosis most commonly involves the sinopulmonary tract reflecting inhalation as the principal portal of entry. Chest computed tomography scans and new non-culture-based assays such as antigen detection and polymerase chain reaction may facilitate the early diagnosis of invasive aspergillosis, but have limitations. Reflecting an important unmet need, there has been a significant expansion in the antifungal armamentarium. The second-generation triazole, voriconazole, was superior to conventional amphotericin B as primary therapy for invasive aspergillosis, and is the new standard of care for this infection. There is significant interest in combination antifungal therapy pairing an echinocandin with either an azole or amphotericin B formulation as therapy for invasive aspergillosis. In addition, there has been an increased understanding of the immunology of Aspergillus infection, paving the way to novel immune augmentation strategies in animal models that merit evaluation in phase I clinic trials.
Collapse
Affiliation(s)
- Brahm H Segal
- Department of Medicine, SUNY at Buffalo, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA.
| | | |
Collapse
|
42
|
Carpenter KJ, Hogaboam CM. Immunosuppressive effects of CCL17 on pulmonary antifungal responses during pulmonary invasive aspergillosis. Infect Immun 2005; 73:7198-207. [PMID: 16239514 PMCID: PMC1273903 DOI: 10.1128/iai.73.11.7198-7207.2005] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Aspergillus fumigatus-sensitized CCR4-deficient (CCR4-/-) mice exhibit an accelerated clearance of conidia during fungal asthma. In the present study, we examined the roles of CCL17 and CCL22, two CCR4 ligands, during pulmonary invasive aspergillosis in neutropenic mice. Kaplan-Meier survival curve analysis revealed that wild-type C57BL/6 (CCR4+/+) mice were significantly protected from the lethal effects of Aspergillus compared with their wild-type controls following systemic neutralization with anti-CCL17 but not anti-CCL22 antibodies. Systemic neutralization of CCL17 significantly increased whole-lung CCL2 levels. Mouse survival and histological analysis revealed that the receptor mediating the deleterious effects of CCL17 was CCR4 since mice genetically deficit in CCR4 (CCR4-/-) did not develop invasive aspergillosis. Enzyme-linked immunosorbent assay analysis of whole-lung samples at day 2 after conidial challenge in neutrophil-depleted CCR4-/- and CCR4+/+ mice revealed that whole-lung IL-12 levels were significantly increased in the CCR4-/- group compared with the wild-type group. Also at day 2 after conidial challenge, significantly greater numbers of CD11c+ F4/80+ and CD11c+/CD86+ but fewer CD3/NK1.1+ cells were present in the lungs of CCR4-/- mice compared with their wild-type counterparts. Thus, CCL17-CCR4 interactions dramatically impair the pulmonary antifungal response against A. fumigatus in neutropenic mice.
Collapse
MESH Headings
- Animals
- Aspergillosis/immunology
- Bronchoalveolar Lavage
- Chemokine CCL17
- Chemokine CCL2/metabolism
- Chemokine CCL22
- Chemokine CCL3
- Chemokine CCL4
- Chemokines, CC/immunology
- Chemokines, CC/metabolism
- Cytokines/genetics
- Cytokines/metabolism
- Female
- Immune Tolerance/immunology
- Interleukin-12/metabolism
- Lung/immunology
- Lung/metabolism
- Lung/pathology
- Lung Diseases, Fungal/immunology
- Lung Diseases, Fungal/microbiology
- Lymphocytes/cytology
- Lymphocytes/immunology
- Macrophage Inflammatory Proteins/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Neutropenia/immunology
- Receptors, CCR4
- Receptors, CCR8
- Receptors, Chemokine/deficiency
- Receptors, Chemokine/genetics
- Receptors, Chemokine/immunology
- Specific Pathogen-Free Organisms
- Survival Rate
- Tumor Necrosis Factor-alpha/metabolism
Collapse
Affiliation(s)
- Kristin J Carpenter
- Department of Pathology, University of Michigan Medical School, Room 5216B, Med Sci I, 1301 Catherine Road, Ann Arbor, MI 48109-0602, USA
| | | |
Collapse
|
43
|
Abstract
In allogeneic hematopoietic stem cell transplantation (SCT), dendritic cells (DCs) as the most potent antigen-presenting cells play a central role in the development of acute and chronic graft-vs-host disease (GVHD), in graft-vs-leukemia or -malignancy reactions and in fighting infectious complications. Functional maturity and distribution of DC sub-types (DC1 and DC2) differ between the different stem cell sources used (bone marrow, granulocyte colony-stimulating factor-mobilised peripheral blood and cord blood) resulting in various rates of graft-vs-host disease and graft-vs-leukemia activity. Although DC recovery following stem cell transplantation is prompt, graft-vs-host disease and the use of immunosuppressive drugs result in qualitative and quantitative disturbances in DC homeostasis and have been observed for up to 1 year after transplantation. Complete donor DC chimerism seems to be a pre-requisite for the development of chronic GVHD and for graft-vs-leukemia activity, at least following reduced-intensity transplants, although in the early phase of acute graft-vs-host disease the presence of host antigen-presenting cells is essential. Preliminary data show promising results with DC-based immunotherapy for treatment of viral and fungal infections and of leukemic relapse following allogeneic stem cell transplantation. More information on the mechanisms and interactions between dendritic cells and regulatory T cells is needed for DC vaccination concepts for modulation of graft-vs-host disease.
Collapse
Affiliation(s)
- D Nachbaur
- Innsbruck Medical University, Bone Marrow Transplantation Unit and Tumor and Immunobiology Laboratory, Division of Hematology and Oncology, Department of Internal Medicine, Innsbruck, Austria.
| | | |
Collapse
|
44
|
Awasthi S, Awasthi V, Magee DM, Coalson JJ. Efficacy of antigen 2/proline-rich antigen cDNA-transfected dendritic cells in immunization of mice against Coccidioides posadasii. THE JOURNAL OF IMMUNOLOGY 2005; 175:3900-6. [PMID: 16148136 DOI: 10.4049/jimmunol.175.6.3900] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Coccidioides posadasii causes coccidioidomycosis, or Valley fever, in the endemic regions of the Southwestern United States. The susceptibility to C. posadasii infection has been attributed to a decreased Th1 cellular response. APCs, especially dendritic cells (DCs), play an important role in the activation of Th1 response. In this study, we investigated the efficacy of a DC-based vaccine against C. posadasii in a mouse model of coccidioidomycosis. We intranasally immunized C57BL6 mice with syngeneic, bone marrow-derived DCs (JAWS II cells) transfected with a cDNA encoding the protective Coccidioides-Ag2/proline-rich Ag. The immunized mice were lethally challenged with C. posadasii through either an i.p. or intranasal route. Upon necropsy after 10 days of infection, fungal burden in lung and spleen of immunized mice was significantly reduced as compared with the control animals. The lung tissue homogenates of immunized animals showed higher levels of IFN-gamma. Histologically, lung tissues of immunized mice were in better condition than the control mice. To further investigate, we studied the biodistribution and trafficking of injected DCs by nuclear imaging techniques. For this purpose, the transfected DCs were radiolabeled with (111)In-oxime. Scintigraphic images showed that most of the label remained in the gastrointestinal tract. A significant amount was also observed in lung, but there were negligible circulating (111)In label in blood. The results suggest that the DCs have a potent immunostimulatory activity, and immunization with DCs transfected with Ag2/proline-rich Ag-cDNA induces protective immunity against C. posadasii in C57BL6 mice.
Collapse
Affiliation(s)
- Shanjana Awasthi
- Department of Pathology, University of Texas Health Science Center, San Antonio, TX 78229, USA
| | | | | | | |
Collapse
|
45
|
Grünebach F, Brossart P. The therapeutic use of dendritic cells transfected with tumour RNA. Expert Opin Ther Pat 2005. [DOI: 10.1517/13543776.15.12.1703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
46
|
Casadevall A, Pirofski LA. The potential of antibody-mediated immunity in the defence against biological weapons. Expert Opin Biol Ther 2005; 5:1359-72. [PMID: 16197341 DOI: 10.1517/14712598.5.10.1359] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Antibody-mediated immunity (AMI) has been used for the treatment and prevention of infectious diseases for > 100 years, and has a remarkable record of safety, efficacy and versatility. AMI can be used for defence against a wide variety of biological weapons, and passive antibody (Ab) therapy has the potential to provide immediate immunity to susceptible individuals. Recent advances in the Ab field make it possible to generate Abs with enhanced antimicrobial functions. There are significant gaps in our understanding of Ab function, such that the development of Ab-based strategies remains a largely empirical exercise. Nevertheless, the advantages inherent in the therapeutic and prophylactic use of AMI provide a powerful rationale for continued development that will undoubtedly yield many new vaccines and therapeutic Abs.
Collapse
Affiliation(s)
- Arturo Casadevall
- Division of Infectious Diseases, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | | |
Collapse
|
47
|
Bellocchio S, Bozza S, Montagnoli C, Perruccio K, Gaziano R, Pitzurra L, Romani L. Immunity to Aspergillus fumigatus: the basis for immunotherapy and vaccination. Med Mycol 2005; 43 Suppl 1:S181-8. [PMID: 16110810 DOI: 10.1080/14789940500051417] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Efficient responses to fungi require different mechanisms of immunity. Dendritic cells (DCs) are uniquely able to decode the fungus-associated information and translate it into qualitatively different T helper (Th) immune responses. Murine and human DCs phagocytose conidia and hyphae of Aspergillus fumigatus through distinct recognition receptors. The engagement of distinct receptors translates into disparate downstream signaling events, ultimately affecting cytokine production and co-stimulation. Adoptive transfer of different types of DCs activates protective and non-protective Th cells as well as regulatory T cells, ultimately affecting the outcome of the infection in mice with invasive aspergillosis. The infusion of fungus-pulsed or RNA-transfected DCs also accelerates recovery of functional antifungal Th 1 responses in mice with allogeneic hematopoietic stem cell transplantation. Patients receiving T cell-depleted allogeneic hematopoietic stem cell transplantation are unable to develop antigen-specific T cell responses soon after transplant due to defective DC functions. Our results suggest that the adoptive transfer of DCs may restore immunocompetence in hematopoietic stem cell transplantation by contributing to the educational program of T cells. Thus, the remarkable furictional plasticity of DCs can be exploited for the deliberate targeting of cells and pathways of cell-mediated immunity in response to the fungus.
Collapse
Affiliation(s)
- S Bellocchio
- Microbiology Section, Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy
| | | | | | | | | | | | | |
Collapse
|
48
|
Ibisch C, Pradal G, Bach JM, Lieubeau B. Functional canine dendritic cells can be generated in vitro from peripheral blood mononuclear cells and contain a cytoplasmic ultrastructural marker. J Immunol Methods 2005; 298:175-82. [PMID: 15847807 DOI: 10.1016/j.jim.2005.02.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2004] [Revised: 12/10/2004] [Accepted: 02/16/2005] [Indexed: 01/06/2023]
Abstract
For physiological and practical reasons the dog is a large animal model used increasingly to study the pathogenesis of human diseases and new therapeutic approaches, in particular for immune disorders. However, some immunological resources are lacking in this model, especially concerning dendritic cells. The aim of our study was to develop an efficient method to generate dendritic cells (DC) in vitro from dog peripheral blood mononuclear cells (PBMC) and to characterize their functional, structural and ultrastructural properties. PBMC were cultured in vitro with IL-4 and GM-CSF. After 1 week of culture, a great proportion of non-adherent cells displayed typical cytoplasmic processes, as evidenced both by optical and electron microscopy. Cytometric analysis revealed the presence of 41.7+/-24.6% CD14+ cells expressing both CD11c and MHC class II molecules. Allogeneic mixed lymphocyte reactions confirmed the ability of these cultures to stimulate the proliferation of allogeneic lymphocytes as already reported as a characteristic of DC in other species. In addition, we describe for the first time the presence in canine DC of cytoplasmic periodic microstructures (PMS) that could represent ultrastructural markers of canine DC. In conclusion, our study provides an easy method to generate DC from PBMC in sufficient numbers for immunological in vitro investigations in dogs, a pre-clinical model for many human diseases.
Collapse
Affiliation(s)
- C Ibisch
- Immuno-Endocrinology Unit, ENVN/INRA/University, ENVN, Atlanpole, la Chantrerie, BP 40706, F-44307 Nantes Cedex 03, France
| | | | | | | |
Collapse
|
49
|
Rementeria A, López-Molina N, Ludwig A, Vivanco AB, Bikandi J, Pontón J, Garaizar J. Genes and molecules involved in Aspergillus fumigatus virulence. Rev Iberoam Micol 2005; 22:1-23. [PMID: 15813678 DOI: 10.1016/s1130-1406(05)70001-2] [Citation(s) in RCA: 184] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Aspergillus fumigatus causes a wide range of diseases that include mycotoxicosis, allergic reactions and systemic diseases (invasive aspergillosis) with high mortality rates. Pathogenicity depends on immune status of patients and fungal strain. There is no unique essential virulence factor for development of this fungus in the patient and its virulence appears to be under polygenetic control. The group of molecules and genes associated with the virulence of this fungus includes many cell wall components, such as beta-(1-3)-glucan, galactomannan, galactomannanproteins (Afmp1 and Afmp2), and the chitin synthetases (Chs; chsE and chsG), as well as others. Some genes and molecules have been implicated in evasion from the immune response, such as the rodlets layer (rodA/hyp1 gene) and the conidial melanin-DHN (pksP/alb1 gene). The detoxifying systems for Reactive Oxygen Species (ROS) by catalases (Cat1p and Cat2p) and superoxide dismutases (MnSOD and Cu, ZnSOD), had also been pointed out as essential for virulence. In addition, this fungus produces toxins (14 kDa diffusible substance from conidia, fumigaclavin C, aurasperon C, gliotoxin, helvolic acid, fumagilin, Asp-hemolysin, and ribotoxin Asp fI/mitogilin F/restrictocin), allergens (Asp f1 to Asp f23), and enzymatic proteins as alkaline serin proteases (Alp and Alp2), metalloproteases (Mep), aspartic proteases (Pep and Pep2), dipeptidyl-peptidases (DppIV and DppV), phospholipase C and phospholipase B (Plb1 and Plb2). These toxic substances and enzymes seems to be additive and/or synergistic, decreasing the survival rates of the infected animals due to their direct action on cells or supporting microbial invasion during infection. Adaptation ability to different trophic situations is an essential attribute of most pathogens. To maintain its virulence attributes A. fumigatus requires iron obtaining by hydroxamate type siderophores (ornitin monooxigenase/SidA), phosphorous obtaining (fos1, fos2, and fos3), signal transductional falls that regulate morphogenesis and/or usage of nutrients as nitrogen (rasA, rasB, rhbA), mitogen activated kinases (sakA codified MAP-kinase), AMPc-Pka signal transductional route, as well as others. In addition, they seem to be essential in this field the amino acid biosynthesis (cpcA and homoaconitase/lysF), the activation and expression of some genes at 37 degrees C (Hsp1/Asp f12, cgrA), some molecules and genes that maintain cellular viability (smcA, Prp8, anexins), etc. Conversely, knowledge about relationship between pathogen and immune response of the host has been improved, opening new research possibilities. The involvement of non-professional cells (endothelial, and tracheal and alveolar epithelial cells) and professional cells (natural killer or NK, and dendritic cells) in infection has been also observed. Pathogen Associated Molecular Patterns (PAMP) and Patterns Recognizing Receptors (PRR; as Toll like receptors TLR-2 and TLR-4) could influence inflammatory response and dominant cytokine profile, and consequently Th response to infec tion. Superficial components of fungus and host cell surface receptors driving these phenomena are still unknown, although some molecules already associated with its virulence could also be involved. Sequencing of A. fumigatus genome and study of gene expression during their infective process by using DNA microarray and biochips, promises to improve the knowledge of virulence of this fungus.
Collapse
Affiliation(s)
- Aitor Rementeria
- Departamento Inmunología, Microbiología y Parasitología, Facultad de Ciencia y Tecnología, Universidad del País Vasco, Spain.
| | | | | | | | | | | | | |
Collapse
|
50
|
Perruccio K, Bozza S, Montagnoli C, Bellocchio S, Aversa F, Martelli M, Bistoni F, Velardi A, Romani L. Prospects for dendritic cell vaccination against fungal infections in hematopoietic transplantation. Blood Cells Mol Dis 2005; 33:248-55. [PMID: 15528139 DOI: 10.1016/j.bcmd.2004.08.011] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2004] [Indexed: 11/20/2022]
Abstract
Dendritic cells (DCs) are uniquely able to initiate and control the immune response to fungi. DCs function at three levels in the manipulation of the immune response to these pathogens. First, they mount an immediate or innate response to them, for example, by producing inflammatory mediators upon capture and phagocytosis; second, through these preceding innate functions, they decode the fungus-associated information and translate it in qualitatively different Th responses, and third, they are key in containing and dampening inflammatory responses by tolerization through the induction of regulatory T cells (Treg). DCs sense fungi in a morphotype-specific manner, through the engagement of distinct recognition receptors ultimately affecting cytokine production and costimulation. Both myeloid and plasmacytoid murine and human DCs phagocytose fungi and undergo functional maturation in response to them. However, their activation program for cytokine production was different, being IL-12 mainly produced by myeloid DCs and IL-12, IL-10 and IFN-alpha mainly produced by plasmacytoid DCs. This resulted in a distinct ability for T cell priming, being Th1, Th2, and Treg differently activated by the different DC subsets. The ability of fungus-pulsed DCs to prime for Th1 and Th2 cell activation upon adoptive transfer in vivo correlated with the occurrence of resistance and susceptibility to the infections, respectively. Antifungal protective immunity was also induced upon adoptive transfer of DCs transfected with fungal RNA. The efficacy was restricted to DCs transfected with RNA from yeasts or conidia but not with RNA from fungal hyphae. The effect was fungus-specific, as no cross-protection was observed upon adoptive transfer of DCs pulsed with either fungal species. The infusion of fungus-pulsed or RNA-transfected DCs accelerated the recovery of functional antifungal Th1 responses in mice with allogeneic hematopoietic stem cell transplantation (HSCT) and affected the outcome of the infections. As the ability of phagocytose fungi was defective in peripheral DCs from patients with HSCT, soon after the transplant, our findings suggest that the adoptive transfer of DCs may restore immunocompetence in HSCT by contributing to the educational program of T cells. Thus, the remarkable functional plasticity of DCs in response to fungi can be exploited for the deliberate targeting of cells and pathways of cell-mediated immunity in response to fungal vaccines.
Collapse
Affiliation(s)
- Katia Perruccio
- Division of Hematology, Clinical Immunology, Department of Clinical and Experimental Medicine, University of Perugia, Perugia, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|