Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
1.
Front Immunol ; 15: 1360140, 2024.
Article in English | MEDLINE | ID: mdl-38711513

ABSTRACT

Introduction: Modified Vaccinia Virus Ankara (MVA) is a safe vaccine vector inducing long- lasting and potent immune responses. MVA-mediated CD8+T cell responses are optimally induced, if both, direct- and cross-presentation of viral or recombinant antigens by dendritic cells are contributing. Methods: To improve the adaptive immune responses, we investigated the role of the purinergic receptor P2X7 (P2RX7) in MVA-infected feeder cells as a modulator of cross-presentation by non-infected dendritic cells. The infected feeder cells serve as source of antigen and provide signals that help to attract dendritic cells for antigen take up and to license these cells for cross-presentation. Results: We demonstrate that presence of an active P2RX7 in major histocompatibility complex (MHC) class I (MHCI) mismatched feeder cells significantly enhanced MVA-mediated antigen cross-presentation. This was partly regulated by P2RX7-specific processes, such as the increased availability of extracellular particles as well as the altered cellular energy metabolism by mitochondria in the feeder cells. Furthermore, functional P2RX7 in feeder cells resulted in a delayed but also prolonged antigen expression after infection. Discussion: We conclude that a combination of the above mentioned P2RX7-depending processes leads to significantly increased T cell activation via cross- presentation of MVA-derived antigens. To this day, P2RX7 has been mostly investigated in regards to neuroinflammatory diseases and cancer progression. However, we report for the first time the crucial role of P2RX7 for antigen- specific T cell immunity in a viral infection model.


Subject(s)
Cross-Priming , Dendritic Cells , Genetic Vectors , Receptors, Purinergic P2X7 , Vaccinia virus , Animals , Humans , Mice , Antigen Presentation/immunology , Antigens, Viral/immunology , CD8-Positive T-Lymphocytes/immunology , Cross-Priming/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Mice, Inbred C57BL , Receptors, Purinergic P2X7/immunology , Receptors, Purinergic P2X7/metabolism , Vaccinia virus/immunology
2.
Cell Rep ; 43(4): 114096, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38607919

ABSTRACT

Receptors controlling the cross-presentation of tumor antigens by macrophage subsets in cancer tissues are poorly explored. Here, we show that TIM4+ large peritoneal macrophages efficiently capture and cross-present tumor-associated antigens at early stages of peritoneal infiltration by ovarian cancer cells. The phosphatidylserine (PS) receptor TIM4 promotes maximal uptake of dead cells or PS-coated artificial targets and triggers inflammatory and metabolic gene programs in combination with cytoskeletal remodeling and upregulation of transcriptional signatures related to antigen processing. At the cellular level, TIM4-mediated engulfment induces nucleation of F-actin around nascent phagosomes, delaying the recruitment of vacuolar ATPase, acidification, and cargo degradation. In vivo, TIM4 deletion blunts induction of early anti-tumoral effector CD8 T cells and accelerates the progression of ovarian tumors. We conclude that TIM4-mediated uptake drives the formation of specialized phagosomes that prolong the integrity of ingested antigens and facilitate cross-presentation, contributing to immune surveillance of the peritoneum.


Subject(s)
Antigens, Neoplasm , Carcinogenesis , Macrophages, Peritoneal , Animals , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/immunology , Female , Mice , Carcinogenesis/pathology , Carcinogenesis/immunology , Carcinogenesis/metabolism , Humans , Antigens, Neoplasm/metabolism , Antigens, Neoplasm/immunology , Ovarian Neoplasms/immunology , Ovarian Neoplasms/pathology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/genetics , Membrane Proteins/metabolism , Mice, Inbred C57BL , Cross-Priming/immunology , Cell Line, Tumor , Phagosomes/metabolism , Antigen Presentation/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Actins/metabolism
3.
Adv Mater ; 36(25): e2402532, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38563503

ABSTRACT

Due to inherent differences in cellular composition and metabolic behavior with host cells, tumor-harbored bacteria can discriminatorily affect tumor immune landscape. However, the mechanisms by which intracellular bacteria affect antigen presentation process between tumor cells and antigen-presenting cells (APCs) are largely unknown. The invasion behavior of attenuated Salmonella VNP20009 (VNP) into tumor cells is investigated and an attempt is made to modulate this behavior by modifying positively charged polymers on the surface of VNP. It is found that non-toxic chitosan oligosaccharide (COS) modified VNP (VNP@COS) bolsters the formation of gap junction between tumor cells and APCs by enhancing the ability of VNP to infect tumor cells. On this basis, a bacterial biohybrid is designed to promote in situ antigen cross-presentation through intracellular bacteria induced gap junction. This bacterial biohybrid also enhances the expression of major histocompatibility complex class I molecules on the surface of tumor cells through the incorporation of Mdivi-1 coupled with VNP@COS. This strategic integration serves to heighten the immunogenic exposure of tumor antigens; while, preserving the cytotoxic potency of T cells. A strategy is proposed to precisely controlling the function and local effects of microorganisms within tumors.


Subject(s)
Antigen Presentation , Chitosan , Gap Junctions , Salmonella , Humans , Chitosan/chemistry , Cell Line, Tumor , Gap Junctions/metabolism , Salmonella/immunology , Animals , Cross-Priming , Mice , Oligosaccharides/chemistry , Neoplasms/immunology , Neoplasms/pathology , Antigen-Presenting Cells/immunology , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class I/immunology
4.
Nat Commun ; 15(1): 2280, 2024 Mar 13.
Article in English | MEDLINE | ID: mdl-38480738

ABSTRACT

Cross-presentation by type 1 DCs (cDC1) is critical to induce and sustain antitumoral CD8 T cell responses to model antigens, in various tumor settings. However, the impact of cross-presenting cDC1 and the potential of DC-based therapies in tumors carrying varied levels of bona-fide neoantigens (neoAgs) remain unclear. Here we develop a hypermutated model of non-small cell lung cancer in female mice, encoding genuine MHC-I neoepitopes to study neoAgs-specific CD8 T cell responses in spontaneous settings and upon Flt3L + αCD40 (DC-therapy). We find that cDC1 are required to generate broad CD8 responses against a range of diverse neoAgs. DC-therapy promotes immunogenicity of weaker neoAgs and strongly inhibits the growth of high tumor-mutational burden (TMB) tumors. In contrast, low TMB tumors respond poorly to DC-therapy, generating mild CD8 T cell responses that are not sufficient to block progression. scRNA transcriptional analysis, immune profiling and functional assays unveil the changes induced by DC-therapy in lung tissues, which comprise accumulation of cDC1 with increased immunostimulatory properties and less exhausted effector CD8 T cells. We conclude that boosting cDC1 activity is critical to broaden the diversity of anti-tumoral CD8 T cell responses and to leverage neoAgs content for therapeutic advantage.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Female , Mice , Animals , Dendritic Cells , Carcinoma, Non-Small-Cell Lung/therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/therapy , Lung Neoplasms/metabolism , CD8-Positive T-Lymphocytes , Cross-Priming
5.
Biomater Sci ; 12(6): 1490-1501, 2024 Mar 12.
Article in English | MEDLINE | ID: mdl-38329387

ABSTRACT

Cross-presentation, exogenous antigen presentation onto major histocompatibility complex class I molecules on antigen presenting cells, is crucially important for inducing antigen-specific cellular immune responses for cancer immunotherapy and for the treatment of infectious diseases. One strategy to induce cross-presentation is cytosolic delivery of an exogenous antigen using fusogenic or endosomolytic molecule-introduced nanocarriers. Earlier, we reported liposomes modified with pH-responsive polymers to achieve cytosolic delivery of an antigen. Polyglycidol-based or polysaccharide-based pH-responsive polymers can provide liposomes with delivery performance of antigenic proteins into cytosol via membrane fusion with endosomes responding to acidic pH, leading to induction of cross-presentation. Mannose residue was introduced to pH-responsive polysaccharides to increase uptake selectivity to antigen presenting cells and to improve cross-presentation efficiency. However, direct introduction of mannose residue into pH-responsive polysaccharides suppressed cytoplasmic delivery performance of liposomes. To avoid such interference, for this study, mannose-containing glycans were incorporated separately into pH-responsive polysaccharide-modified liposomes. Soybean agglutinin-derived glycopeptide was used as a ligand for lectins on antigen presenting cells. Incorporation of glycopeptide significantly increased the cellular uptake of liposomes by dendritic cell lines and increased cross-presentation efficiency. Liposomes incorporated both glycopeptide and pH-responsive polysaccharides exhibited strong adjuvant effects in vitro and induced the increase of dendritic cells, M1 macrophages, and effector T cells in the spleen. Subcutaneous administration of these liposomes induced antigen-specific cellular immunity, resulting in strong therapeutic effects in tumor-bearing mice. These results suggest that separate incorporation of glycopeptides and pH-responsive polysaccharides into antigen-loaded liposomes is an effective strategy to produce liposome-based nanovaccines to achieve antigen cross-presentation and induction of cellular immunity towards cancer immunotherapy.


Subject(s)
Liposomes , Neoplasms , Animals , Mice , Liposomes/chemistry , Antigen Presentation , Cross-Priming , Glycopeptides/pharmacology , Mannose/pharmacology , Antigens/chemistry , Neoplasms/therapy , Polymers/chemistry , Hydrogen-Ion Concentration , Polysaccharides/chemistry , Dendritic Cells , Mice, Inbred C57BL
6.
J Immunol ; 212(7): 1063-1068, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38353614

ABSTRACT

Activation of naive CD8-positive T lymphocytes is mediated by dendritic cells that cross-present MHC class I (MHC-I)-associated peptides derived from exogenous Ags. The most accepted mechanism involves the translocation of Ags from phagosomes or endolysosomes into the cytosol, where antigenic peptides generated by cytosolic proteasomes are delivered by the transporter associated with Ag processing (TAP) to the endoplasmic reticulum, or an endocytic Ag-loading compartment, where binding to MHC-I occurs. We have described an alternative pathway where cross-presentation is independent of TAP but remains dependent on proteasomes. We provided evidence that active proteasomes found within the lumen of phagosomes and endolysosomal vesicles locally generate antigenic peptides that can be directly loaded onto trafficking MHC-I molecules. However, the mechanism of active proteasome delivery to the endocytic compartments remained unknown. In this study, we demonstrate that phagosome-associated LC3A/B structures deliver proteasomes into subcellular compartments containing exogenous Ags and that autophagy drives TAP-independent, proteasome-dependent cross-presentation.


Subject(s)
Cross-Priming , Proteasome Endopeptidase Complex , Proteasome Endopeptidase Complex/metabolism , Antigen Presentation , Autophagosomes , Phagosomes/metabolism , Histocompatibility Antigens Class I , Antigens , Membrane Transport Proteins/metabolism , Peptides/metabolism
7.
Vaccine ; 42(7): 1549-1560, 2024 Mar 07.
Article in English | MEDLINE | ID: mdl-38320931

ABSTRACT

Tumor subunit vaccines have great potential in personalized cancer immunotherapy. They are usually administered with adjuvant owing to their low immunogenicity. Cholera toxin (CT) is a biological adjuvant with diverse biological functions and a long history of use. Our earlier study revealed that a CT-like chimeric protein co-delivered with murine granulocyte-macrophage colony stimulating factor (mGM-CSF) and prostate cancer antigen epitope could co-stimulate dendritic cells (DCs) and enhance cross presentation of tumor epitope. To further study the molecular mechanism of CT-like chimeric protein in cross presentation, major histocompatibility complex class I (MHC I)-restricted epitope 257-264 of ovalbumin (OVAT) was used as a model antigen peptide in this study. Recombinant A subunit and pentameric B subunit of CT protein were respectively genetically constructed and purified. Then both assembled into AB5 chimeric protein in vitro. Three different chimeric biomacromolecules containing mGM-CSF and OVAT were constructed according to the different fusion sites and whether the endoplasmic reticulum (ER) retention sequence was included. It was found that A2 domain and B subunit of CT were both available for loading epitopes and retaining GM1 affinity. The binding activity of GM1 was positively correlated with antigen endocytosis. Once internalized, DCs became mature and cross-presented antigen. KDEL helped the whole molecule to be retained in the ER, and this improved the cross presentation of antigen on MHC I molecules. In conclusion, hexameric CT-like chimeric protein with dual effects of GM1 affinity and ER retention sequence were potential in improvement of cross presentation. The results laid a foundation for designing personalized tumor vaccine based on CT-like chimeric protein molecular structure.


Subject(s)
Cholera Toxin , Neoplasms , Mice , Animals , Humans , Cholera Toxin/metabolism , Cross-Priming , G(M1) Ganglioside/metabolism , G(M1) Ganglioside/pharmacology , Recombinant Proteins/pharmacology , Adjuvants, Immunologic/pharmacology , Recombinant Fusion Proteins/genetics , Epitopes , Antigen Presentation
8.
Microbiol Spectr ; 12(3): e0307423, 2024 Mar 05.
Article in English | MEDLINE | ID: mdl-38349165

ABSTRACT

Influenza virus is known to cause respiratory tract infections of varying severity in individuals of all ages. The EasyNAT Rapid Flu assay is a newly developed in vitro diagnostic test that employs cross-priming isothermal amplification (CPA) to detect and differentiate influenza A and B viruses in human nasopharyngeal (NP) swabs. The aim of this study is to determine the performance characteristics of the EasyNAT Rapid Flu assay for rapid detection of influenza virus. The limit of detection (LOD) and cross-reactivity of the EasyNAT Rapid Flu assay were assessed. The clinical performance of the assay was evaluated using NP swab samples that were tested with real-time reverse-transcription polymerase chain reaction (RT-PCR) and Xpert Xpress Flu/RSV assay. The LOD for the detection of influenza A and B using the EasyNAT Rapid Flu assay was found to be 500 copies/mL. Furthermore, the assay exhibited no cross-reactivity with other common respiratory viruses tested. For the 114 NP swab samples tested for influenza A using both the EasyNAT Rapid Flu assay and real-time RT-PCR, the two assays demonstrated a high level of agreement (κ = 0.963, P < 0.001), with a positive percentage agreement (PPA) of 97.7% and a negative percentage agreement (NPA) of 98.6%. Similarly, for the 43 NP swab samples tested for influenza A and B using both the EasyNAT Rapid Flu assay and Xpert Xpress Flu/RSV assay, the two assays showed a high level of agreement (κ = 0.933, P < 0.001), with the overall rate of agreement (ORA) of 97.7% for influenza A and 100% for influenza B. The EasyNAT Rapid Flu assay demonstrates excellent performance in the detection of influenza A, highlighted by its strong agreement with RT-PCR-based assays.IMPORTANCEThe newly developed EasyNAT Rapid Flu assay is an innovative cross-priming isothermal amplification-based method designed for detecting influenza A and B viruses at point-of-care settings. This study aims to thoroughly assess the analytical and clinical performance of the assay, offering valuable insights into its potential advantages and limitations. The findings of this research hold significant implications for clinical practice.


Subject(s)
Influenza A virus , Influenza, Human , Respiratory Syncytial Virus Infections , Humans , Influenza, Human/diagnosis , Influenza A virus/genetics , Influenza B virus/genetics , Point-of-Care Systems , Cross-Priming , Sensitivity and Specificity , Nasopharynx , Molecular Diagnostic Techniques/methods , Respiratory Syncytial Virus Infections/diagnosis
9.
J Virol Methods ; 325: 114890, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38309371

ABSTRACT

Epizootics of Koi herpesvirus (KHV) cause mass mortality in koi carp (Cyprinus rubrofuscus) and common carp (Cyprinus carpio) worldwide. Rapid and accurate virus detection technology is crucial for preventing pathogen spread and minimizing damage. Although several diagnostic assays have been developed for KHV, the analytical and diagnostic performance of the detection methods has not been evaluated. In this study, we developed and validated the diagnostic performance of two molecular diagnostic assays, cross-priming amplification-based lateral flow assay (CPA-LFA) and TaqMan probe-based real-time polymerase chain reaction (PCR). To detect KHV, primers and probe were designed based on the thymidine kinase (TK) genes. The detection limits of developed CPA-LFA and real-time PCR assays were determined to be 675.69 copies/µL and 8.384 copies/µL, respectively. The diagnostic sensitivity and specificity of the developed assay were determined using fish samples (n = 179). CPA-LFA was found to be 93.67% and 100%, respectively, and real-time PCR was found to be 100% and 100%, respectively. Therefore, the newly developed CPA-LFA and real-time PCR assays accurately and rapidly detect KHV. CPA-LFA is particularly suitable for point-of-care diagnosis because of its simple diagnostic process, and real-time PCR analysis is most suitable for precise diagnosis because it can detect low viral loads.


Subject(s)
Carps , Fish Diseases , Herpesviridae Infections , Herpesviridae , Animals , Herpesviridae Infections/diagnosis , Herpesviridae Infections/veterinary , Real-Time Polymerase Chain Reaction , Cross-Priming , Fish Diseases/diagnosis , Herpesviridae/genetics
10.
Int J Biol Macromol ; 261(Pt 1): 129518, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38244740

ABSTRACT

The induction of a robust CD8+ T cell response is critical for the success of an antiviral vaccine. In this study, we incorporated a STING agonist (SA) 2'3'-cGAMP into a previously developed exosome-based CVB3 viral myocarditis vaccine (Exo-VP1) to enhance its ability to induce CD8+ T cell responses and immunoprotection. Our results showed that compared to free SA adjuvant, exosome-mediated co-delivery (ExoSA-VP1) significantly enhanced SA uptake by dendritic cells (DCs) and more potently stimulated DC maturation. Immunization of mice showed that the ExoSA-VP1 vaccine-induced higher levels of CVB3-specific T cell proliferation and cytotoxicity, significantly increased the percentage of IFN-γ+CD8+ rather than CD4+ T cells, effectively reduced cardiac viral loads, attenuated myocarditis and improved survival in mice compared to the previous Exo-VP1 vaccine. Further investigation showed that ExoSA-VP1 significantly increased both the percentage and antigen cross-presentation capacity of splenic CD8+ DCs. Depletion of these CD8+ DCs by cytochrome C administration nearly abolished the advantage of ExoSA-VP1 in dominantly inducing IFN-γ+CD8+ cytotoxic T lymphocyte (CTL) production in immunized mice. Taken together, our results demonstrated the potential of ExoSA-VP1 as a promising candidate for anti-CVB3 vaccines and provide insights into immune-enhancing strategies aiming at augmenting antigen cross-presentation by DCs and enhancing potent CTL responses.


Subject(s)
Exosomes , Myocarditis , Viral Vaccines , Animals , Mice , Cross-Priming , CD8-Positive T-Lymphocytes , Dendritic Cells
11.
J Mol Biol ; 436(4): 168446, 2024 02 15.
Article in English | MEDLINE | ID: mdl-38242283

ABSTRACT

Adjuvants are vaccine components that can boost the type, magnitude, breadth, and durability of an immune response. We have previously demonstrated that certain adjuvant combinations can act synergistically to enhance and shape immunogenicity including promotion of Th1 and cytotoxic T-cell development. These combinations also promoted protective immunity in vulnerable populations such as newborns. In this study, we employed combined antigen-specific human in vitro models to identify adjuvant combinations that could synergistically promote the expansion of vaccine-specific CD4+ cells, induce cross-presentation on MHC class I, resulting in antigen-specific activation of CD8+ cells, and direct the balance of immune response to favor the production of Th1-promoting cytokines. A screen of 78 adjuvant combinations identified several T cell-potentiating adjuvant combinations. Remarkably, a combination of TLR9 and STING agonists (CpG + 2,3-cGAMP) promoted influenza-specific CD4+ and CD8+ T cell activation and selectively favored production of Th1-polarizing cytokines TNF and IL-12p70 over co-regulated cytokines IL-6 and IL-12p40, respectively. Phenotypic reprogramming towards cDC1-type dendritic cells by CpG + 2,3-cGAMP was also observed. Finally, we characterized the molecular mechanism of this adjuvant combination including the ability of 2,3-cGAMP to enhance DC expression of TLR9 and the dependency of antigen-presenting cell activation on the Sec22b protein important to endoplasmic reticulum-Golgi vesicle trafficking. The identification of the adjuvant combination CpG + 2,3-cGAMP may therefore prove key to the future development of vaccines against respiratory viral infections tailored for the functionally distinct immune systems of vulnerable populations such as older adults and newborns.


Subject(s)
Adjuvants, Immunologic , Cross-Priming , Th1 Cells , Vaccine Development , Viral Vaccines , Humans , Infant, Newborn , Adjuvants, Immunologic/pharmacology , Cross-Priming/drug effects , Cytokines/metabolism , Dendritic Cells/immunology , Toll-Like Receptor 9 , Th1 Cells/immunology , Adolescent , Young Adult , Viral Vaccines/immunology
12.
Cancer Immunol Immunother ; 73(1): 9, 2024 Jan 17.
Article in English | MEDLINE | ID: mdl-38231450

ABSTRACT

The conventional type 1 dendritic cells (cDC1) play a pivotal role in protective immunity against pathogens and cancer. However, their low frequency in the blood and tissues limits their use in immune therapy. We have recently described a method to vaccinate against neoantigens that are induced in tumor cells by targeted delivery of a TAP siRNA to dendritic cells using a TLR9 binding CpG oligonucleotide. Since TLR9 is also expressed in immune suppressive myeloid populations TLR9 targeting could reduce the effectiveness of this approach. Here, we describe a modular multivalent antibody platform to target the TAP siRNA to resident Clec9a expressing cDC1 and show that it leads to selective and sustained TAP downregulation in cDC1 and inhibits tumor growth in mice more effectively than CpG targeted siRNA. To induce DC maturation an agonistic CD40 antibody was administered to the siRNA treated mice. To obviate the need for a second drug formulation and reduce the risk of toxicity, we exploited the multivalent nature of this targeting platform to co-deliver the TAP siRNA and a DC maturation agent, a CpG containing oligonucleotide, to cDC1 in vivo and show that it was more effective than Clec9a targeting of TAP siRNA in combination with CD40 antibody. This study describes a way to manipulate the function of cDC1 cells in vivo using a broadly applicable antibody-based targeting platform to deliver multiple biological agents to specific cells in vivo to potentiate (immune) therapy and to probe the biology of specific cell types in their natural settings.


Subject(s)
Cross-Priming , Toll-Like Receptor 9 , Animals , Mice , Antibodies , Vaccination , RNA, Small Interfering/genetics , CD40 Antigens , Oligonucleotides
13.
Biomaterials ; 305: 122470, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38228027

ABSTRACT

The efficacy of radiotherapy has not yet achieved optimal results, partially due to insufficient priming and infiltration of effector immune cells within the tumor microenvironment (TME), which often exhibits suppressive phenotypes. In particular, the infiltration of X-C motif chemokine receptor 1 (XCR1)-expressing conventional type-1 dendritic cells (cDC1s), which are critical in priming CD8+ cytotoxic T cells, within the TME is noticeably restricted. Hence, we present a facile methodology for the efficient fabrication of a calcium phosphate hydrogel loaded with X-C motif chemokine ligand 1 (XCL1) to selectively recruit cDC1s. Manganese phosphate microparticles were also loaded into this hydrogel to reprogram the TME via cGAS-STING activation, thereby facilitating the priming of cDC1s propelled specific CD8+ T cells. They also polarize tumor-associated macrophages towards the M1 phenotype and reduce the proportion of regulatory cells, effectively reversing the immunosuppressive TME into an immune-active one. The yielded XCL1@CaMnP gel exhibits significant efficacy in enhancing the therapeutic outcomes of radiotherapy, particularly when concurrently administered with postoperative radiotherapy, resulting in an impressive 60 % complete response rate. Such XCL1@CaMnP gel, which recruits cDC1s to present tumor antigens generated in situ, holds great potential as a versatile platform for enhanced cancer treatment through modulating the immunosuppressive TME.


Subject(s)
CD8-Positive T-Lymphocytes , Cross-Priming , T-Lymphocytes, Cytotoxic , Dendritic Cells , Hydrogels/pharmacology , Tumor Microenvironment
14.
J Clin Microbiol ; 62(1): e0092323, 2024 01 17.
Article in English | MEDLINE | ID: mdl-38112450

ABSTRACT

IMPORTANCE: In this study, we successfully established a new One-Pot method, named TB One-Pot, for detecting Mtb in sputum by combining CRISPR-cas12b-mediated trans-cleavage with cross-priming amplification (CPA). Our study evaluated the diagnostic performance of TB One-Pot in clinical sputum samples for tuberculosis. The findings provide evidence for the potential of TB One-Pot as a diagnostic tool for tuberculosis.


Subject(s)
Mycobacterium tuberculosis , Tuberculosis , Humans , Mycobacterium tuberculosis/genetics , Sputum/microbiology , Cross-Priming , CRISPR-Cas Systems , Sensitivity and Specificity , Nucleic Acid Amplification Techniques/methods , Tuberculosis/diagnosis , Tuberculosis/microbiology
15.
J. investig. allergol. clin. immunol ; 34(1): 38-48, 2024. tab, graf
Article in English | IBECS | ID: ibc-230813

ABSTRACT

Background: Cashew nuts often cause strong allergic reactions, which are even more severe than those of peanuts. Ana o 1 (vicilin), Ana o 2 (legumin), and Ana o 3 (2S albumin) are major cashew allergens. Cosensitization to all 3 nonhomologous cashew nut allergens has been observed. We hypothesize that this might be due to IgE cross-reactivity. Methods: IgE cross-inhibitions were performed with Ana o 1-3 using serum samples from cashew nut–allergic patients. The related hazelnut allergens Cor a 11, 9, and 14 were used as controls. For comparison, IgE cross-reactivity between the hazelnut allergens was investigated using serum samples from hazelnut-allergic patients. Results: The median percentages of cross-inhibition between Ana o 1, 2, and 3 were 84%-99%. In comparison, the median cross- inhibition values between hazelnut allergens were 33%-62%. The IC50 values revealed the highest IgE affinity to be to Ana o 3 and Cor a 14. Hazelnut legumin Cor a 9 inhibited IgE binding to Ana o 1, 2, and 3, with median percentages of 75%, 56%, and 48%, respectively. No cross-reactivity was observed between allergenic vicilins or between 2S albumins from cashew and hazelnut. Potentially cross-reactive peptides of Ana o 3 identified in silico overlapped with previously reported IgE epitopes of all 3 allergens. Conclusion: IgE with high affinity to Ana o 3 that cross-reacts with the other 2 major nonhomologous cashew nut allergens might be responsible for the high allergenic potency of cashew nut. These cross-reactive IgE types comprise the major fraction of specific IgE in cashew-allergic patients and might be responsible for cross-reactivity between unrelated tree nuts (AU)


Antecedentes: Los anacardos suelen provocar fuertes reacciones alérgicas, que son incluso más graves que las del maní. Ana o 1 (vicilina), Ana o 2 (legumina) y Ana o 3 (albúmina 2S) son los principales alérgenos del anacardo. Se ha observado cosensibilización a los 3 alérgenos no homólogos del anacardo. Nuestra hipótesis es que esto podría deberse a la reactividad cruzada de la IgE. Métodos : Se realizaron inhibiciones cruzadas de IgE con Ana o 1-3 utilizando muestras de suero de pacientes alérgicos al anacardo. Como controles se utilizaron los alérgenos de avellana relacionados Cor a 11, 9 y 14. A modo de comparación, se investigó la reactividad cruzada de IgE entre los alérgenos de la avellana utilizando muestras de suero de pacientes alérgicos a la avellana. Resultados : Los porcentajes medios de inhibición cruzada entre Ana o 1, 2 y 3 fueron del 84% al 99%. En comparación, los valores medios de inhibición cruzada entre alérgenos de avellana fueron del 33% al 62%. Los valores de IC50 revelaron que la mayor afinidad de IgE era Ana o 3 y Cor a 14. La legumina de avellana Cor a 9 inhibió la unión de IgE a Ana o 1, 2 y 3, con porcentajes medios de 75%, 56% y 48%. , respectivamente. No se observó reactividad cruzada entre vicilinas alergénicas ni entre albúminas 2S de anacardo y avellana. Los péptidos potencialmente de reacción cruzada de Ana o 3 identificados in silico se superpusieron con epítopos de IgE previamente informados de los 3 alérgenos. Conclusión : La IgE con alta afinidad por Ana o 3 que reacciona de forma cruzada con los otros 2 alérgenos principales no homólogos del anacardo podría ser responsable de la alta potencia alergénica del anacardo. Estos tipos de IgE de reacción cruzada comprenden la fracción principal de IgE específica en pacientes alérgicos al anacardo y podrían ser responsables de la reactividad cruzada entre frutos secos no relacionados (AU)


Subject(s)
Humans , Male , Female , Child, Preschool , Child , Food Hypersensitivity , Cross Reactions , Cross-Priming , Allergens
16.
Int J Mol Sci ; 25(1)2023 Dec 22.
Article in English | MEDLINE | ID: mdl-38203367

ABSTRACT

The major histocompatibility complexes of vertebrates play a key role in the immune response. Antigen-presenting cells are loaded on MHC I molecules, which mainly present endogenous antigens; when MHC I presents exogenous antigens, this is called cross-presentation. The discovery of cross-presentation provides an important theoretical basis for the study of exogenous antigens. Cross-presentation is a complex process in which MHC I molecules present antigens to the cell surface to activate CD8+ T lymphocytes. The process of cross-representation includes many components, and this article briefly outlines the origins and development of MHC molecules, gene structures, functions, and their classical presentation pathways. The cross-presentation pathways of MHC I molecules, the cell lines that support cross-presentation, and the mechanisms of MHC I molecular transporting are all reviewed. After more than 40 years of research, the specific mechanism of cross-presentation is still unclear. In this paper, we summarize cross-presentation and anticipate the research and development prospects for cross-presentation.


Subject(s)
Cross-Priming , Histocompatibility Antigens , Animals , Major Histocompatibility Complex , Antigen-Presenting Cells , Histocompatibility Antigens Class I
17.
J. investig. allergol. clin. immunol ; 32(5): 383-392, 2022. ilus, graf
Article in English | IBECS | ID: ibc-212734

ABSTRACT

Background: Immediate and delayed-type hypersensitivity reactions to pet-borne allergens are common in atopic diseases. In atopic dermatitis (AD), controversy surrounds the contribution to the disease of cross-reactivity to self-proteins. Human cystatin A and the cat allergen Fel d 3 belong to the cystatins, an evolutionary conserved protein family. The objective of the present study was to assess crossreactivity between mammalian cystatins and to analyze T-cell responses to cystatin in AD patients sensitized to pet dander. Methods: cDNA coding for dog cystatin was cloned from dog skin. Sera from 245 patients with IgE-mediated sensitization to cat and dog dander were tested for IgE binding to recombinantly expressed feline, canine, and human cystatin. Of these, 141 were also diagnosed with AD. Results: Cystatin-specific IgE was detected in 36 patients (14.7%), of whom 19 were considerably affected by AD. Within the AD patients, 9 had measurable IgE against all 3 cystatins. Cystatin-sensitized AD patients did not differ from non–cystatin-sensitized patients in terms of disease severity, age, or total IgE levels. T-cell cytokine measurements showed elevated IL-4 levels after stimulation with feline and human cystatin. Conclusion: The humoral response suggests that in addition to Fel d 3, the homologous protein from dog might play a role in allergy. Furthermore, human cystatin appears to be capable of driving a type 2 immune response in sensitized AD patients and may therefore be considered a so-called autoallergen, as proposed for other evolutionary conserved proteins. (AU)


Antecedentes: Las reacciones de hipersensibilidad de tipo inmediato y retardado a los alérgenos que están en las mascotas son comunes en las enfermedades atópicas. En este estudio, en pacientes con dermatitis atopica (DA), se analiza la reactividad cruzada con las autoproteínas y su contribución a la enfermedad. Tanto la cistatina A humana como el alérgeno felino Fel d 3 pertenecen a la familia de las cistatinas, una familia de proteínas conservadas evolutivamente. El objetivo del presente estudio fue evaluar la reactividad cruzada entre las cistatinas de mamíferos y analizar la respuestas de las células T a la cistatina en pacientes con DA sensibilizados a la caspa de las mascotas. Métodos: El ADNc que codifica la cistatina de perro se clonó a partir de piel de perro. Se analizaron sueros de 245 pacientes con sensibilización por IgE a la caspa de gato y perro para determinar la unión de IgE a cistatina felina, canina y humana expresada de forma recombinante, respectivamente. De estos 245 pacientes, 141 fueron diagnosticados de DA. Resultados: Se detectó IgE específica frente a cistatina en el 14,7% (36) de los pacientes, de los cuales 19 padecían DA. Dentro de los pacientes con DA, 9 tenían IgE medible contra las tres cistatinas. Los pacientes con DA sensibilizados frente a cistatina no difirieron de los pacientes no sensibilizados con cistatina en términos de gravedad de la enfermedad, edad o niveles totales de IgE. El análisis de citocinas de células T reveló niveles elevados de IL-4 después de la estimulación con cistatina felina y humana. Conclusión: La respuesta humoral sugiere que, además de Fel d 3, la proteína homóloga de perro también podría desempeñar un papel en la alergia. Además, la cistatina humana parece ser capaz de promover una respuesta inmune de tipo 2 en pacientes con DA sensibilizados y, por lo tanto, puede considerarse un autoalérgeno, como se ha propuesto para otras proteínas conservadas evolutivamente. (AU)


Subject(s)
Humans , Animals , Cats , Dogs , Dermatitis, Atopic/etiology , Pets , Cross-Priming , Cystatins/immunology , T-Lymphocytes/immunology , Dermatitis, Atopic/immunology , Enzyme-Linked Immunosorbent Assay , Electrophoresis, Polyacrylamide Gel
19.
Allergol. immunopatol ; 49(4): 155-161, jul. 2021. tab, graf
Article in English | IBECS | ID: ibc-214296

ABSTRACT

Introduction and objectives: With increasing pet allergies among pediatric patients, the need for precise environmental care is increasing. We investigated the clinical, immunological, and environmental characteristics of pediatric patients sensitized to a dog to evaluate the cross-antigenicity of canine lipocalin Can f 1 with feline lipocalin Fel d 1 and Syrian hamster extract. Materials and methods: The protein fractions of the processed and commercial Syrian hamster extracts were compared using sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE). An enzyme-linked immunosorbent assay (ELISA) inhibition test was performed on Can f 1, Fel d 1, and processed Syrian hamster extract, and the antigen-specific immunoglobulin E (IgE)-binding capacity for each antigen was analyzed using serum samples from patients. Results: Twelve of 19 patients with a median age of 40.5 months were symptomatic when exposed to dogs. Eleven (91.7%) patients showed a positive IgE response to Can f 1. Two patients were positive for Fel d 1-specific IgE antibody, and one was positive for hamster-specific IgE antibody. SDS-PAGE confirmed the presence of different patterns of protein bands between the commercial and processed hamster extracts. There was no cross-antigenicity among Can f 1, Fel d 1, and processed Syrian hamster extract. Conclusions: Since the standard commercial hamster extract did not contain Syrian hamster antigens that were diverse enough, caution should be taken when using it. In children allergic to cats and dogs, sensitization to isolated Can f 1 or Fel d 1 is unlikely to cause cross-reactivity to Syrian hamster hair and epithelium (AU)


Subject(s)
Humans , Animals , Child, Preschool , Child , Cats , Dogs , Cricetinae , Allergens/immunology , Plant Extracts , Retrospective Studies , Enzyme-Linked Immunosorbent Assay , Immunoglobulin E , Lipocalins/immunology , Cross-Priming , Dander/immunology
20.
Allergol. immunopatol ; 48(3): 265-269, mayo-jun. 2020. graf, tab
Article in English | IBECS | ID: ibc-192029

ABSTRACT

INTRODUCTION AND OBJECTIVES: A high rate of cross-reactivity has been reported between the specific proteins of hen's egg with proteins of various avian eggs by quantitative immunoelectrophoretic techniques. The aim of this study was to assess the clinical cross-reactivity of different birds' eggs in children with hen's egg allergy based on skin prick test results. MATERIAL AND METHODS: This cross-sectional study enrolled 52 infants with hen's egg allergy and 52 healthy infants with no history of food allergy from October 2018 to April 2019. Skin prick tests were performed in both patient and control groups with fresh extract of white and yolk related to pigeon, duck, goose, turkey, quail, and partridge. RESULTS: Fifty (96.1%) children with hen's egg allergy showed positive sensitization to at least one of the avian eggs. The most frequent positive skin tests were related to quail's white (36 = 69.2%) followed by duck's white (34 = 65.5%), and sensitization was the least frequent in pigeon's yolk (23 = 44.2%). Skin tests of the control group were negative to all the tested extracts. CONCLUSIÓN: Because of fewer sensitizations to some avian eggs, further research should clarify starting oral immunotherapy with the yolk of goose and pigeon in children with hen's egg allergy


No disponible


Subject(s)
Humans , Male , Female , Infant , Child, Preschool , Egg Proteins/immunology , Food Hypersensitivity/immunology , Egg Hypersensitivity/immunology , Cross-Priming/immunology , Allergens/analysis , Egg Hypersensitivity/diagnosis , Skin Tests/methods , Cross-Sectional Studies , Immunotherapy/methods , Surveys and Questionnaires
SELECTION OF CITATIONS
SEARCH DETAIL