Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 689
Filter
1.
Cell Host Microbe ; 32(10): 1681-1690, 2024 Oct 09.
Article in English | MEDLINE | ID: mdl-39389032

ABSTRACT

Fungal disease poses a growing threat to public health that our current antifungal therapies are not well equipped to meet. As the population of immunocompromised hosts expands, and ecological changes favor the emergence of fungal pathogens, the development of new antifungal agents, including vaccines, becomes a global priority. Here, we summarize recent advancements in the understanding of fungal pathogenesis, key features of the host antifungal immune response, and how these findings could be leveraged to design novel approaches to deadly fungal disease.


Subject(s)
Fungal Vaccines , Fungi , Mycoses , Fungal Vaccines/immunology , Humans , Fungi/immunology , Mycoses/immunology , Mycoses/prevention & control , Mycoses/microbiology , Animals , Immunocompromised Host , Antifungal Agents/therapeutic use , Antifungal Agents/pharmacology , Host-Pathogen Interactions/immunology
2.
Hum Vaccin Immunother ; 20(1): 2406065, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-39327639

ABSTRACT

Candida albicans Is a leading cause of nosocomial bloodstream infections, particularly in immunocompromised patients. Current therapeutic strategies are insufficient, highlighting the need for effective vaccines. This study aimed to evaluate the efficacy of a dual-antigen fusion protein vaccine (AH) targeting the Als3 and Hyr1 proteins of C. albicans, using AlPO4 as an adjuvant. The AH vaccine was constructed by fusing Als317-432 and Hyr125-350 proteins, and its immunogenicity was tested in BALB/c mice and New Zealand white rabbits. Mice received three intramuscular doses of the vaccine combined with AlPO4, followed by a lethal challenge with C. albicans SC5314. Survival rates, antibody responses, cytokine production, fungal burdens, and organ pathology were assessed. The vaccine's efficacy was also validated using rabbit serum. Mice vaccinated with the AH-AlPO4 combination exhibited significantly higher antibody titers, particularly IgG and its subclasses, compared to controls (p < .001). The survival rate of vaccinated mice was 80% post-infection, significantly higher than the control group (p < .01). Vaccinated mice showed reduced fungal loads in the blood, kidneys, spleen, and liver (p < .05). Increased levels of interferon gamma and interleukin (IL)-17A were observed, indicating robust T helper (Th) 1 and Th17 cell responses. Vaccination mitigated organ damage, with kidney and liver pathology scores significantly lower than those of unvaccinated mice (p < .05). Rabbit serum with polyclonal antibodies demonstrated effective antifungal activity, confirming vaccine efficacy across species. The AH-AlPO4 vaccine effectively induced strong immune responses, reduced fungal burden, and protected against organ pathology in C. albicans infections. These findings support further development of dual-antigen vaccine strategies.


Candida, a fungus, is a major cause of bloodstream infections, especially in critical care settings. This study focused on developing a vaccine to protect against Candida infection. The vaccine targeted two key proteins, Als3p and Hyr1p, found on the surface of Candida, using a combination of these proteins. To create the vaccine, we used Als3p and Hyr1p to form a fusion protein called AH, and tested the vaccine on mice, administering it with different adjuvants (substances that enhance the immune response). The results showed that the AH vaccine, particularly when combined with the adjuvant AlPO4, induced a strong immune response in mice. This response included the production of specific antibodies and immune cells that are crucial for defending against Candida infections. Furthermore, mice receiving the AH-AlPO4 vaccine showed significantly better survival rates and lower levels of fungal infection compared to the control group or another experimental group. The vaccine also protected vital organs, such as the kidneys and liver, from Candida-induced damage. Additionally, we used rabbit serum to validate the efficacy of the vaccine, providing cross-species confirmation of its effectiveness. The study demonstrated the potential of the AH vaccine in eliciting robust immune responses and reducing the severity of Candida albicans infections. In summary, this research introduces a promising AH vaccine, which shows effectiveness in protecting against Candida infections. The study's innovative approach and positive results contribute to the ongoing efforts to develop vaccines against fungal infections, addressing a critical healthcare challenge. Further research is needed to explore the vaccine's long-term effectiveness and safety for potential use in clinical settings.


Subject(s)
Adjuvants, Immunologic , Antibodies, Fungal , Antigens, Fungal , Candida albicans , Candidiasis , Fungal Vaccines , Mice, Inbred BALB C , Recombinant Fusion Proteins , Animals , Fungal Vaccines/immunology , Fungal Vaccines/administration & dosage , Candida albicans/immunology , Candidiasis/prevention & control , Candidiasis/immunology , Antibodies, Fungal/blood , Antibodies, Fungal/immunology , Rabbits , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/genetics , Mice , Female , Antigens, Fungal/immunology , Antigens, Fungal/administration & dosage , Adjuvants, Immunologic/administration & dosage , Fungal Proteins/immunology , Fungal Proteins/administration & dosage , Cytokines , Vaccination/methods , Immunoglobulin G/blood , Disease Models, Animal , Interleukin-17/immunology , Interferon-gamma/immunology , Vaccine Efficacy , Survival Analysis , Alum Compounds
3.
ACS Infect Dis ; 10(10): 3475-3495, 2024 Oct 11.
Article in English | MEDLINE | ID: mdl-39255073

ABSTRACT

In impoverished nations, the COVID-19 pandemic has led to a widespread occurrence of deadly fungal diseases like mucormycosis. The limited availability of effective antifungal treatments and the emergence of drug-resistant fungal strains further exacerbate the situation. Factors such as systemic steroid use, intravenous drug misuse, and overutilization of broad-spectrum antimicrobials contribute to the prevalence of hospital-acquired infections caused by drug-resistant fungi. Fungal infections exploit compromised immune status and employ intricate mechanisms to evade immune surveillance. The immune response involves the innate and adaptive immune systems, leading to phagocytic and complement-mediated elimination of fungi. However, resistance to antifungals poses a challenge, highlighting the importance of antifungal prophylaxis and therapeutic vaccination. Understanding the host-fungal immunological interactions and developing vaccines are vital in combating fungal infections. Further research is needed to address the high mortality and morbidity associated with multidrug-resistant fungal pathogens and to develop innovative treatment drugs and vaccines. This review focuses on the global epidemiological burden of fungal infections, host-fungal immunological interactions, recent advancements in vaccine development and the road ahead.


Subject(s)
COVID-19 , Fungal Vaccines , Mycoses , SARS-CoV-2 , Humans , COVID-19/prevention & control , COVID-19/epidemiology , COVID-19/immunology , Fungal Vaccines/immunology , Mycoses/prevention & control , Mycoses/epidemiology , Mycoses/immunology , SARS-CoV-2/immunology , SARS-CoV-2/drug effects , Antifungal Agents/pharmacology , Antifungal Agents/therapeutic use , Vaccine Development , Drug Resistance, Fungal
4.
J Immunol ; 213(6): 843-852, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-39109925

ABSTRACT

Candida spp. are the fourth leading cause of bloodstream infections in hospitalized patients and the most common cause of invasive fungal infection. No vaccine against Candida spp. or other fungal pathogens of humans is available. We recently discovered the Blastomyces Dectin-2 ligand endoglucanase 2 that harbors antigenic and adjuvant functions and can function as a protective vaccine against that fungus. We also reported that the adjuvant activity, which is mediated by O-mannans decorating the C terminus of Blastomyces Dectin-2 ligand endoglucanase 2, can augment peptide Ag-induced vaccine immunity against heterologous agents, including Cryptococcus, Candida, and influenza. In this article, we report that the O-linked mannans alone, in the absence of any antigenic peptide, can also protect against systemic candidiasis, reducing kidney fungal load and increasing survival in a Dectin-2-dependent manner. We found that this long-term glycan-induced protection is mediated by innate lymphocyte populations including TCR-γδ+ T cells, innate lymphoid cells, and NK cells that subsequently activate and release reactive oxygen species from neutrophils and monocytes. Our findings suggest that Blastomyces O-mannan displayed by Eng2 induces a form of protective trained immunity mediated by innate lymphocyte populations.


Subject(s)
Candidiasis , Fungal Vaccines , Immunity, Innate , Mannans , Mice , Animals , Fungal Vaccines/immunology , Immunity, Innate/immunology , Candidiasis/immunology , Candidiasis/prevention & control , Mannans/immunology , Blastomyces/immunology , Lectins, C-Type/immunology , Mice, Inbred C57BL , Vaccination , Killer Cells, Natural/immunology , Humans , Mice, Knockout
5.
Med Mycol ; 62(8)2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39122658

ABSTRACT

Cryptococcus neoformans is a widely distributed opportunistic pathogenic fungus. While C. neoformans commonly infects immunocompromised individuals, it can also affect those who are immunocompetent. Transmission of C. neoformans primarily occurs through the respiratory tract, leading to the development of meningitis. The mortality rate of Cryptococcal meningitis is high, and treatment options are limited. Cryptococcus neoformans infections pose a significant public health threat and currently lack targeted and effective response strategies. This study aimed to screen T lymphocyte (cytotoxic T lymphocyte and helper T lymphocyte) and B lymphocyte epitopes derived from four C. neoformans antigens and develop two multi-epitope vaccines by combining them with various adjuvants. Molecular docking results demonstrated that the vaccines bind stably to Toll-like receptor 4 ( and induce innate immunity. The credibility of the molecular docking results was validated through subsequent molecular dynamics simulations. Furthermore, the results of immune simulation analyses underscored the multi-epitope vaccine's capability to effectively induce robust humoral and cellular immune responses within the host organism. These two vaccines have demonstrated theoretical efficacy against C. neoformans infection as indicated by computer analysis. Nevertheless, additional experimental validation is essential to substantiate the protective efficacy of the vaccines.


A multi-epitope Cryptococcus neoformans vaccine covering the most common A and D phenotypes was designed using bioinformatics methods.


Subject(s)
Computational Biology , Cryptococcus neoformans , Epitopes, B-Lymphocyte , Epitopes, T-Lymphocyte , Fungal Vaccines , Molecular Docking Simulation , Cryptococcus neoformans/immunology , Cryptococcus neoformans/chemistry , Fungal Vaccines/immunology , Epitopes, T-Lymphocyte/immunology , Epitopes, B-Lymphocyte/immunology , Humans , Cryptococcosis/immunology , Cryptococcosis/prevention & control , Toll-Like Receptor 4/immunology , Antigens, Fungal/immunology , Molecular Dynamics Simulation , Adjuvants, Immunologic , Immunoinformatics
6.
mSphere ; 9(8): e0046724, 2024 Aug 28.
Article in English | MEDLINE | ID: mdl-39037263

ABSTRACT

Systemic candidiasis remains a significant public health concern worldwide, with high mortality rates despite available antifungal drugs. Drug-resistant strains add to the urgency for alternative therapies. In this context, vaccination has reemerged as a prominent immune-based strategy. Extracellular vesicles (EVs), nanosized lipid bilayer particles, carry a diverse array of native fungal antigens, including proteins, nucleic acids, lipids, and glycans. Previous studies from our laboratory demonstrated that Candida albicans EVs triggered the innate immune response, activating bone marrow-derived dendritic cells (BMDCs) and potentially acting as a bridge between innate and adaptive immunity. Vaccination with C. albicans EVs induced the production of specific antibodies, modulated cytokine production, and provided protection in immunosuppressed mice infected with lethal C. albicans inoculum. To elucidate the mechanisms underlying EV-induced immune activation, our study investigated pathogen-associated molecular patterns (PAMPs) and pattern recognition receptors (PRRs) involved in EVs-phagocyte engagement. EVs from wild-type and mutant C. albicans strains with truncated mannoproteins were compared for their ability to stimulate BMDCs. Our findings revealed that EV decoration with O- and N-linked mannans and the presence of ß-1,3-glucans and chitin oligomers may modulate the activation of specific PRRs, in particular Toll-like receptor 4 (TLR4) and dectin-1. The protective effect of vaccination with wild-type EVs was found to be dependent on TLR4. These results suggest that fungal EVs can be harnessed in vaccine formulations to selectively activate PRRs in phagocytes, offering potential avenues for combating or preventing candidiasis.IMPORTANCESystemic candidiasis is a serious global health concern with high mortality rates and growing drug resistance. Vaccination offers a promising solution. A unique approach involves using tiny lipid-coated particles called extracellular vesicles (EVs), which carry various fungal components. Previous studies found that Candida albicans EVs activate the immune response and may bridge the gap between innate and adaptive immunity. To understand this better, we investigated how these EVs activate immune cells. We demonstrated that specific components on EV surfaces, such as mannans and glucans, interact with receptors on immune cells, including Toll-like receptor 4 (TLR4) and dectin-1. Moreover, vaccinating with these EVs led to strong immune responses and full protection in mice infected with Candida. This work shows how harnessing fungal EVs might lead to effective vaccines against candidiasis.


Subject(s)
Candida albicans , Candidiasis , Dendritic Cells , Extracellular Vesicles , Fungal Vaccines , Receptors, Pattern Recognition , Toll-Like Receptor 4 , Animals , Candida albicans/immunology , Extracellular Vesicles/immunology , Toll-Like Receptor 4/immunology , Toll-Like Receptor 4/metabolism , Mice , Candidiasis/immunology , Candidiasis/prevention & control , Candidiasis/microbiology , Fungal Vaccines/immunology , Fungal Vaccines/administration & dosage , Dendritic Cells/immunology , Receptors, Pattern Recognition/immunology , Mice, Inbred C57BL , Female , Immunity, Innate , Disease Models, Animal
7.
mBio ; 15(8): e0174624, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-38980038

ABSTRACT

The global burden of infections due to the pathogenic fungus Cryptococcus is substantial in persons with low CD4+ T-cell counts. Previously, we deleted three chitin deacetylase genes from Cryptococcus neoformans to create a chitosan-deficient, avirulent strain, designated as cda1∆2∆3∆, which, when used as a vaccine, protected mice from challenge with virulent C. neoformans strain KN99. Here, we explored the immunological basis for protection. Vaccine-mediated protection was maintained in mice lacking B cells or CD8+ T cells. In contrast, protection was lost in mice lacking α/ß T cells or CD4+ T cells. Moreover, CD4+ T cells from vaccinated mice conferred protection upon adoptive transfer to naive mice. Importantly, while monoclonal antibody-mediated depletion of CD4+ T cells just prior to vaccination resulted in complete loss of protection, significant protection was retained in mice depleted of CD4+ T cells after vaccination but prior to challenge. Vaccine-mediated protection was lost in mice genetically deficient in interferon-γ (IFNγ), tumor necrosis factor alpha (TNFα), or interleukin (IL)-23p19. A robust influx of leukocytes and IFNγ- and TNFα-expressing CD4+ T cells was seen in the lungs of vaccinated and challenged mice. Finally, a higher level of IFNγ production by lung cells stimulated ex vivo correlated with lower fungal burden in the lungs. Thus, while B cells and CD8+ T cells are dispensable, IFNγ and CD4+ T cells have overlapping roles in generating protective immunity prior to cda1∆2∆3∆ vaccination. However, once vaccinated, protection becomes less dependent on CD4+ T cells, suggesting a strategy for vaccinating HIV+ persons prior to loss of CD4+ T cells. IMPORTANCE: The fungus Cryptococcus neoformans is responsible for >100,000 deaths annually, mostly in persons with impaired CD4+ T-cell function such as AIDS. There are no approved human vaccines. We previously created a genetically engineered avirulent strain of C. neoformans, designated as cda1∆2∆3∆. When used as a vaccine, cda1∆2∆3∆ protects mice against a subsequent challenge with a virulent C. neoformans strain. Here, we defined components of the immune system responsible for vaccine-mediated protection. We found that while B cells and CD8+ T cells were dispensible, protection was lost in mice genetically deficient in CD4+ T cells and the cytokines IFNγ, TNFα, or IL-23. A robust influx of cytokine-producing CD4+ T cells was seen in the lungs of vaccinated mice following infection. Importantly, protection was retained in mice depleted of CD4+ T cells following vaccination, suggesting a strategy to protect persons who are at risk of future CD4+ T-cell dysfunction.


Subject(s)
CD4-Positive T-Lymphocytes , Chitosan , Cryptococcosis , Cryptococcus neoformans , Fungal Vaccines , Animals , Cryptococcus neoformans/immunology , Cryptococcus neoformans/genetics , Cryptococcosis/immunology , Cryptococcosis/prevention & control , Cryptococcosis/microbiology , Fungal Vaccines/immunology , Fungal Vaccines/administration & dosage , Fungal Vaccines/genetics , Chitosan/immunology , Mice , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Mice, Inbred C57BL , Interferon-gamma/immunology , Interferon-gamma/metabolism , Female
8.
Front Biosci (Landmark Ed) ; 29(7): 246, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-39082330

ABSTRACT

BACKGROUND: Pneumocystis jirovecii is the most emerging life-threating health problem that causes acute and fatal pneumonia infection. It is rare and more contagious for patients with leukemia and immune-deficiency disorders. Until now there is no treatment available for this infection therefore, it is needed to develop any treatment against this pathogen. METHODS: In this work, we used comparative proteomics, robust immune-informatics, and reverse vaccinology to create an mRNA vaccine against Pneumocystis jirovecii by targeting outer and transmembrane proteins. Using a comparative subtractive proteomic analysis of two Pneumocystis jirovecii proteomes, a distinct non-redundant Pneumocystis jirovecii (strain SE8) proteome was chosen. Seven Pneumocystis jirovecii transmembrane proteins were chosen from this proteome based on hydrophilicity, essentiality, virulence, antigenicity, pathway interaction, protein-protein network analysis, and allergenicity. OBJECTIVE: The reverse vaccinology approach was used to predict the immunogenic and antigenic epitopes of major histocompatibility complex (MHC) I, II and B-cells from the selected proteins on the basis of their antigenicity, toxicity and allergenicity. These immunogenic epitopes were linked together to construct the mRNA-based vaccine. To enhance the immunogenicity, suitable adjuvant, linkers (GPGPG, KK, and CYY), and PRDRE sequences were used. RESULTS: Through predictive modeling and confirmation via the Ramachandran plot, we assessed secondary and 3D structures. The adjuvant RpfE was incorporated to enhance the vaccine construct's immunogenicity (GRAVY index: -0.271, instability index: 39.53, antigenicity: 1.0428). The physiochemical profiling of vaccine construct was predicted it an antigenic, efficient, and potential vaccine. Notably, strong interactions were observed between the vaccine construct and TLR-3/TLR-4 (-1301.7 kcal/mol-1 and -1374.7 kcal/mol-1). CONCLUSIONS: The results predicted that mRNA-based vaccines trigger a cellular and humoral immune response, making the vaccine potential candidate against Pneumocystis jirovecii and it is more suitable for in-vitro analysis and validation to prove its effectiveness.


Subject(s)
Pneumocystis carinii , Pneumonia, Pneumocystis , Proteomics , Vaccinology , mRNA Vaccines , Proteomics/methods , Pneumocystis carinii/immunology , Pneumocystis carinii/genetics , Humans , Vaccinology/methods , mRNA Vaccines/immunology , Pneumonia, Pneumocystis/prevention & control , Pneumonia, Pneumocystis/immunology , Pneumonia, Pneumocystis/microbiology , Fungal Vaccines/immunology , Fungal Proteins/immunology , Fungal Proteins/genetics , Proteome/immunology , RNA, Messenger/genetics , RNA, Messenger/immunology , Vaccine Development/methods , Vaccines, Synthetic/immunology
9.
PLoS Pathog ; 20(7): e1012220, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38976694

ABSTRACT

The fungal infection, cryptococcosis, is responsible for >100,000 deaths annually. No licensed vaccines are available. We explored the efficacy and immune responses of subunit cryptococcal vaccines adjuvanted with Cationic Adjuvant Formulation 01 (CAF01). CAF01 promotes humoral and T helper (Th) 1 and Th17 immune responses and has been safely used in human vaccine trials. Four subcutaneous vaccines, each containing single recombinant Cryptococcus neoformans protein antigens, partially protected mice from experimental cryptococcosis. Protection increased, up to 100%, in mice that received bivalent and quadrivalent vaccine formulations. Vaccinated mice that received a pulmonary challenge with C. neoformans had an influx of leukocytes into the lung including robust numbers of polyfunctional CD4+ T cells which produced interferon gamma (IFNγ), tumor necrosis factor alpha (TNFα), and interleukin (IL)-17 upon ex vivo antigenic stimulation. Cytokine-producing lung CD8+ T cells were also found, albeit in lesser numbers. A significant, durable IFNγ response was observed in the lungs, spleen, and blood. Moreover, IFNγ secretion following ex vivo stimulation directly correlated with fungal control in the lungs. Thus, we have developed multivalent cryptococcal vaccines which protect mice from experimental cryptococcosis using an adjuvant which has been safely tested in humans. These preclinical studies suggest a path towards human cryptococcal vaccine trials.


Subject(s)
Adjuvants, Immunologic , Cryptococcosis , Cryptococcus neoformans , Fungal Vaccines , Vaccines, Subunit , Cryptococcosis/immunology , Cryptococcosis/prevention & control , Animals , Mice , Fungal Vaccines/immunology , Fungal Vaccines/administration & dosage , Cryptococcus neoformans/immunology , Vaccines, Subunit/immunology , Vaccines, Subunit/administration & dosage , Adjuvants, Immunologic/administration & dosage , Female , Mice, Inbred C57BL , Adjuvants, Vaccine/administration & dosage , Antigens, Fungal/immunology , Disease Models, Animal
10.
Immunotherapy ; 16(11): 733-748, 2024.
Article in English | MEDLINE | ID: mdl-38940276

ABSTRACT

Aim: Cryptococcus gattii causes a severe fungal infection with high mortality rate among immunosuppressed and immunocompetent individuals. Due to limitation of current antifungal treatment, new immunotherapeutic approaches are explored.Methods: This study investigated an immunization strategy utilizing heat-inactivated C. gattii with ArtinM as an adjuvant. C57BL/6 mice were intranasally immunized with heat-killed C. gattii and ArtinM was administrated either before immunization or along with HK-C. gattii. Mice were infected with C. gattii and the efficacy of the immunization protocol was evaluated.Results: Mice that received ArtinM exhibited increased levels of IL-10 and relative expression of IL-23 in the lungs, reduced fungal burden and preserved tissue integrity post-infection.Conclusion: Adjuvant ArtinM improved immunization against C. gattii infection in C57BL/6 mice.


Cryptococcus gattii is a fungus that can make lungs sick. Right now, there are no good treatments for it, so scientists are trying to find new ways to fight it. In a recent study, they tested a type of immunotherapy called ArtinM to see if it could help. When they gave ArtinM to mice, the mice got healthier and had less fungus in their lungs. This means ArtinM might be able to help fight this fungus.


Subject(s)
Adjuvants, Immunologic , Cryptococcosis , Cryptococcus gattii , Mice, Inbred C57BL , Animals , Cryptococcus gattii/immunology , Mice , Cryptococcosis/immunology , Adjuvants, Immunologic/administration & dosage , Fungal Vaccines/immunology , Disease Models, Animal , Female , Humans
11.
mSphere ; 9(7): e0018924, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-38904363

ABSTRACT

Epitopes from the Candida cell surface proteins Fba and Met6 are putative vaccine targets for invasive candidiasis. Here, we describe a Candida vaccine approach in which short peptides derived from Fba and Met6 are used in spontaneous nanoliposome antigen particle (SNAP) format. SNAP was enabled by the interaction of cobalt porphyrin phospholipid in liposomes with three histidine residues on the N-terminus of synthetic short peptide immunogens from Fba (F-SNAP), Met6 (M-SNAP), or bivalent Fba and Met6 (FM-SNAP). Liposomes were adjuvanted with synthetic monophosphoryl lipid and QS-21. In mice, immunization with F-SNAP, M-SNAP, or FM-SNAP induced antigen-specific IgG responses and mixed Th1/Th2 immunity. The duplex FM-SNAP vaccine elicited stronger antibody responses against each peptide, even at order-of-magnitude lower peptide dosing than a comparable adjuvanted, conjugate vaccine. Enzyme-linked immunosorbent spot analysis revealed the induction of antigen-specific, cytokine-producing T cells. Compared to F-SNAP or M-SNAP, higher production of TNFα, IL-2, and IFNγ was observed with re-stimulation of splenocytes from bivalent FM-SNAP-immunized mice. When vaccinated BALB/c mice were challenged with Candida auris, analysis of the fungal burden in the kidneys showed that SNAP vaccination protected from disseminated candidiasis. In a lethal fungal exposure model in A/J mice, F-SNAP, M-SNAP, and FM-SNAP vaccination protected mice from candidiasis challenge. Together, these results show that further investigation into the SNAP adjuvant platform is warranted using Fba and Met6 epitopes for a pan-Candida peptide vaccine that provides multifaceted protective immune responses. IMPORTANCE: This study introduces a promising vaccine strategy against invasive candidiasis, a severe fungal infection, by targeting specific peptides on the surface of Candida. Using a novel approach called spontaneous nanoliposome antigen particle (SNAP), we combined peptides from two key Candida proteins, Fba and Met6, into a vaccine. This vaccine induced robust immune responses in mice, including the production of protective antibodies and the activation of immune cells. Importantly, mice vaccinated with SNAP were shielded from disseminated candidiasis in experiments. These findings highlight a potential avenue for developing a broad-spectrum vaccine against Candida infections, which could significantly improve outcomes for patients at risk of these often deadly fungal diseases.


Subject(s)
Antibodies, Fungal , Candidiasis , Fungal Vaccines , Liposomes , Mice, Inbred BALB C , Animals , Mice , Fungal Vaccines/immunology , Fungal Vaccines/administration & dosage , Liposomes/immunology , Candidiasis/prevention & control , Candidiasis/immunology , Female , Antibodies, Fungal/immunology , Antigens, Fungal/immunology , Vaccines, Subunit/immunology , Vaccines, Subunit/administration & dosage , Cytokines/immunology , Vaccination , Fungal Proteins/immunology , Fungal Proteins/administration & dosage , Immunoglobulin G/blood , Immunoglobulin G/immunology , Adjuvants, Immunologic/administration & dosage , Candida albicans/immunology , Candida/immunology , Disease Models, Animal
12.
Arch Microbiol ; 206(7): 293, 2024 Jun 08.
Article in English | MEDLINE | ID: mdl-38850421

ABSTRACT

Fungal infections are incurring high risks in a range from superficial mucosal discomforts (such as oropharyngeal candidiasis and vulvovaginal candidiasis) to disseminated life-threatening diseases (such as invasive pulmonary aspergillosis and cryptococcal meningitis) and becoming a global health problem in especially immunodeficient population. The major obstacle to conquer fungal harassment lies in the presence of increasing resistance to conventional antifungal agents used in newly clinically isolated strains. Although recombinant cytokines and mono-/poly-clonal antibodies are added into antifungal armamentarium, more effective antimycotic drugs are exceedingly demanded. It is comforting that the development of fungal vaccines and adjuvants opens up a window to brighten the prospective way in the diagnosis, prevention and treatment of fungal assaults. In this review, we focus on the progression of several major fungal vaccines devised for the control of Candida spp., Aspergillus spp., Cryptococcus spp., Coccidioides spp., Paracoccidioides spp., Blastomyces spp., Histoplasma spp., Pneumocystis spp. as well as the adjuvants adopted. We then expound the interaction between fungal vaccines/adjuvants and host innate (macrophages, dendritic cells, neutrophils), humoral (IgG, IgM and IgA) and cellular (Th1, Th2, Th17 and Tc17) immune responses which generally experience immune recognition of pattern recognition receptors, activation of immune cells, and clearance of invaded fungi. Furthermore, we anticipate an in-depth understanding of immunomodulatory properties of univalent and multivalent vaccines against diverse opportunistic fungi, providing helpful information in the design of novel fungal vaccines and adjuvants.


Subject(s)
Adjuvants, Immunologic , Fungal Vaccines , Mycoses , Fungal Vaccines/immunology , Humans , Mycoses/prevention & control , Mycoses/immunology , Animals , Fungi/immunology
13.
EMBO Mol Med ; 16(6): 1254-1283, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38783167

ABSTRACT

Disseminated fungal infections account for ~1.5 million deaths per year worldwide, and mortality may increase further due to a rise in the number of immunocompromised individuals and drug-resistance fungal species. Since an approved antifungal vaccine is yet to be available, this study explored the immunogenicity and vaccine efficacy of a DNA polymerase mutant strain of Candida albicans. CNA25 is a pol32ΔΔ strain that exhibits growth defects and does not cause systemic candidiasis in mice. Immunized mice with live CNA25 were fully protected against C. albicans and C. parapsilosis but partially against C. tropicalis and C. glabrata infections. CNA25 induced steady expression of TLR2 and Dectin-1 receptors leading to a faster recognition and clearance by the immune system associated with the activation of protective immune responses mostly mediated by neutrophils, macrophages, NK cells, B cells, and CD4+ and CD8+ T cells. Molecular blockade of Dectin-1, IL-17, IFNγ, and TNFα abolished resistance to reinfection. Altogether, this study suggested that CNA25 collectively activates innate, adaptive, and trained immunity to be a promising live whole-cell vaccine against systemic candidiasis.


Subject(s)
Candida albicans , Candidiasis , Fungal Vaccines , Animals , Candidiasis/immunology , Candidiasis/prevention & control , Candidiasis/microbiology , Fungal Vaccines/immunology , Fungal Vaccines/administration & dosage , Mice , Candida albicans/immunology , Lectins, C-Type/metabolism , Lectins, C-Type/genetics , Female , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 2/immunology , Disease Models, Animal , Mice, Inbred C57BL
14.
Vaccine ; 42(20): 125990, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-38789371

ABSTRACT

Candida albicans can cause superficial or systemic infections in humans, particularly in immunocompromised individuals. Vaccination strategies targeting specific antigens of C. albicans have shown promise in providing protection against invasive candidiasis. This study aimed to evaluate the immuno-protective capacity of a KLH conjugated complex peptide, 3P-KLH, containing epitopes from C. albicans antigens Als3, Hwp1, and Met6 in a murine model of hematogenously induced candidiasis. Mice immunized with 3P-KLH raised a specific antibody response, and protection against C. albicans infection was assessed. Immunized mice exhibited significantly lower fungal load in their kidneys compared to the control group. Moreover, 37.5 % of immunized mice survived 21 days after the infection, while all control animals died within the first nine days. These findings suggest that the 3P-KLH complex peptide, targeting C. albicans key antigens, elicits a protective immune response and reduces the severity of systemic Candida infection. In addition, the high binding affinity of the selected epitopes with MHC II alleles further supports the potential immunogenicity of this peptide in humans. This research provides insights into the development of novel immunotherapeutic approaches against invasive candidiasis.


Subject(s)
Antibodies, Fungal , Antigens, Fungal , Candida albicans , Candidiasis , Fungal Proteins , Fungal Vaccines , Animals , Candida albicans/immunology , Fungal Vaccines/immunology , Fungal Vaccines/administration & dosage , Antigens, Fungal/immunology , Fungal Proteins/immunology , Fungal Proteins/genetics , Mice , Candidiasis/prevention & control , Candidiasis/immunology , Antibodies, Fungal/immunology , Female , Disease Models, Animal , Mice, Inbred BALB C , Epitopes/immunology , Peptides/immunology , Kidney/immunology , Kidney/microbiology , Kidney/pathology
15.
Methods Mol Biol ; 2775: 393-410, 2024.
Article in English | MEDLINE | ID: mdl-38758333

ABSTRACT

Creating a safe and effective vaccine against infection by the fungal pathogen Cryptococcus neoformans is an appealing option that complements the discovery of new small molecule antifungals. Recent animal studies have yielded promising results for a variety of vaccines that include live-attenuated and heat-killed whole-cell vaccines, as well as subunit vaccines formulated around recombinant proteins. Some of the recombinantly engineered cryptococcal mutants in the chitosan biosynthesis pathway are avirulent and very effective at conferring protective immunity. Mice vaccinated with these avirulent chitosan-deficient strains are protected from a lethal pulmonary infection with C. neoformans strain KN99. Heat-killed derivatives of the vaccination strains are likewise effective in a murine model of infection. The efficacy of these whole-cell vaccines, however, is dependent on a number of factors, including the inoculation dose, route of vaccination, frequency of vaccination, and the specific mouse strain used in the study. Here, we present detailed methods for identifying and optimizing various factors influencing vaccine potency and efficacy in various inbred mouse strains using a chitosan-deficient cda1Δcda2Δcda3Δ strain as a whole-cell vaccine candidate. This chapter describes the protocols for immunizing three different laboratory mouse strains with vaccination regimens that use intranasal, orotracheal, and subcutaneous vaccination routes after the animals were sedated using two different types of anesthesia.


Subject(s)
Chitosan , Cryptococcosis , Cryptococcus neoformans , Fungal Vaccines , Animals , Chitosan/chemistry , Mice , Fungal Vaccines/immunology , Fungal Vaccines/genetics , Fungal Vaccines/administration & dosage , Cryptococcosis/immunology , Cryptococcosis/prevention & control , Cryptococcosis/microbiology , Cryptococcus neoformans/immunology , Cryptococcus neoformans/genetics , Disease Models, Animal , Vaccination/methods , Female , Vaccines, Attenuated/immunology , Vaccines, Attenuated/genetics
16.
Methods Mol Biol ; 2775: 411-422, 2024.
Article in English | MEDLINE | ID: mdl-38758334

ABSTRACT

Cryptococcus neoformans infections are a major worldwide concern as current treatment strategies are becoming less effective in alleviating the infection. The most extreme and fatal cases are those of immunocompromised individuals. Clinical treatments for cryptococcosis are limited to a few classes of approved drugs, and due to a rise in drug resistance, these drugs are becoming less effective. Therefore, it is essential to develop innovative ways to control this infection. Vaccinations have emerged as a safe, viable, and cost-effective solution to treat a number of diseases over the years. Currently, there are no clinically available vaccines to treat cryptococcal infections, but a number of studies have shown promising results in animal models. Here, we present step-by-step experimental protocols using live-attenuated or heat-killed C. neoformans cells as a vaccination strategy in a preventive or in a therapeutic murine model of cryptococcosis.


Subject(s)
Cryptococcosis , Cryptococcus neoformans , Disease Models, Animal , Fungal Vaccines , Cryptococcus neoformans/immunology , Cryptococcosis/immunology , Cryptococcosis/prevention & control , Animals , Fungal Vaccines/immunology , Mice , Vaccination/methods , Vaccines, Attenuated/immunology , Humans
17.
ACS Infect Dis ; 10(6): 2089-2100, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38819951

ABSTRACT

Cryptococcus neoformans is a fungus classified by the World Health Organization as a critically important pathogen, which poses a significant threat to immunocompromised individuals. In this study, we present the chemical synthesis and evaluation of two semisynthetic vaccine candidates targeting the capsular polysaccharide glucuronoxylomannan (GXM) of C. neoformans. These semisynthetic glycoconjugate vaccines contain an identical synthetic decasaccharide (M2 motif) antigen. This antigen is present in serotype A strains, which constitute 95% of the clinical cryptococcosis cases. This synthetic oligosaccharide was conjugated to two proteins (CRM197 and Anthrax 63 kDa PA) and tested for immunogenicity in mice. The conjugates elicited a specific antibody response that bound to the M2 motif but also exhibited additional cross-reactivity toward M1 and M4 GXM motifs. Both glycoconjugates produced antibodies that bound to GXM in ELISA assays and to live fungal cells. Mice immunized with the CRM197 glycoconjugate produced weakly opsonic antibodies and displayed trends toward increased median survival relative to mice given a mock PBS injection (18 vs 15 days, p = 0.06). These findings indicate promise, achieving a successful vaccine demands further optimization of the glycoconjugate. This antigen could serve as a component in a multivalent GXM motif vaccine.


Subject(s)
Antibodies, Fungal , Cryptococcosis , Cryptococcus neoformans , Fungal Vaccines , Glycoconjugates , Vaccines, Conjugate , Cryptococcus neoformans/immunology , Animals , Fungal Vaccines/immunology , Mice , Cryptococcosis/prevention & control , Cryptococcosis/immunology , Glycoconjugates/immunology , Glycoconjugates/chemistry , Vaccines, Conjugate/immunology , Antibodies, Fungal/immunology , Female , Polysaccharides/immunology , Polysaccharides/chemistry , Mice, Inbred BALB C , Bacterial Proteins/immunology , Bacterial Proteins/chemistry , Antigens, Fungal/immunology
18.
Int Immunopharmacol ; 135: 112242, 2024 Jun 30.
Article in English | MEDLINE | ID: mdl-38772296

ABSTRACT

The emergence of Cryptococcus neoformans has posed an undeniable burden to many regions worldwide, with its strains mainly entering the lungs through the respiratory tract and spreading throughout the body. Limitations of drug regimens, such as high costs and limited options, have directed our attention toward the promising field of vaccine development. In this study, the subtractive proteomics approach was employed to select target proteins from databases that can accurately cover serotypes A and D of the Cryptococcus neoformans. Further, two multi-epitope vaccines consisting of T and B cell epitopes were demonstrated that they have good structural stability and could bind with immune receptor to induce desired immune responses in silico. After further evaluation, these vaccines show the potential for large-scale production and applicability to the majority of the population of the world. In summary, these two vaccines have been theoretically proven to combat Cryptococcus neoformans infections, awaiting further experimental validation of their actual protective effects.


Subject(s)
Computational Biology , Cryptococcosis , Cryptococcus neoformans , Epitopes, B-Lymphocyte , Fungal Vaccines , Proteomics , Cryptococcus neoformans/immunology , Fungal Vaccines/immunology , Proteomics/methods , Cryptococcosis/immunology , Cryptococcosis/prevention & control , Humans , Computational Biology/methods , Epitopes, B-Lymphocyte/immunology , Epitopes, T-Lymphocyte/immunology , Animals , Antigens, Fungal/immunology , Fungal Proteins/immunology , Fungal Proteins/chemistry , Vaccine Development , Immunoinformatics
19.
Elife ; 132024 May 24.
Article in English | MEDLINE | ID: mdl-38787374

ABSTRACT

Despite current antifungal therapy, invasive candidiasis causes >40% mortality in immunocompromised individuals. Therefore, developing an antifungal vaccine is a priority. Here, we could for the first time successfully attenuate the virulence of Candida albicans by treating it with a fungistatic dosage of EDTA and demonstrate it to be a potential live whole cell vaccine by using murine models of systemic candidiasis. EDTA inhibited the growth and biofilm formation of C. albicans. RNA-seq analyses of EDTA-treated cells (CAET) revealed that genes mostly involved in metal homeostasis and ribosome biogenesis were up- and down-regulated, respectively. Consequently, a bulky cell wall with elevated levels of mannan and ß-glucan, and reduced levels of total monosomes and polysomes were observed. CAET was eliminated faster than the untreated strain (Ca) as found by differential fungal burden in the vital organs of the mice. Higher monocytes, granulocytes, and platelet counts were detected in Ca- vs CAET-challenged mice. While hyper-inflammation and immunosuppression caused the killing of Ca-challenged mice, a critical balance of pro- and anti-inflammatory cytokines-mediated immune responses are the likely reasons for the protective immunity in CAET-infected mice.


Subject(s)
Candida albicans , Candidiasis , Animals , Candida albicans/immunology , Mice , Candidiasis/immunology , Candidiasis/prevention & control , Fungal Vaccines/immunology , Disease Models, Animal , Virulence , Female , Cytokines/metabolism , Biofilms/drug effects , Biofilms/growth & development
20.
Dtsch Med Wochenschr ; 149(10): 560-568, 2024 May.
Article in German | MEDLINE | ID: mdl-38657595

ABSTRACT

Invasive fungal diseases (IFD) are difficult to treat and pose a significant threat to immunocompromised individuals. Current antifungal agents face limitations, including antifungal resistance and adverse effects. This review aims to give a comprehensive overview of emerging treatment strategies.Novel drugs in development are Ibrexafungerp, an orally available triterpenoid inhibiting glucan synthesis, and Rezafungin representing the echinocandins with extended half-life and improved tissue penetration, both recently licensed for certain indications. Fosmanogepix targets glycosylphosphatidylinositol biosynthesis, while Olorofim, an orotomide, inhibits fungal nucleic acid synthesis, both currently assessed in advanced clinical trials.Immunotherapeutic approaches include immune checkpoint inhibitors to enhance immune response in immunosuppressed individuals and fungal-specific allogeneic CAR-T cell therapy. For prophylactic purpose in high-risk populations to develop IFD, monoclonal antibodies against different virulence factors of Candida spp. have been discovered but are not yet seen in clinical trials. Vaccines against distinct fungal antigens as well as pan fungal vaccines to prevent IFD are under development in preclinical stages, notably for Candida spp., Cryptococcus spp., and Aspergillus spp., however, their clinical value is still discussed.In summary, major advances to treat IFD have been observed, but challenges for their establishment in the clinical routine persist.


Subject(s)
Antifungal Agents , Invasive Fungal Infections , Humans , Invasive Fungal Infections/prevention & control , Invasive Fungal Infections/drug therapy , Antifungal Agents/therapeutic use , Fungal Vaccines/therapeutic use , Fungal Vaccines/immunology , Immunotherapy/methods , Therapies, Investigational
SELECTION OF CITATIONS
SEARCH DETAIL