Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 135
Filtrer
1.
Mol Genet Metab ; 142(3): 108494, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38820907

RÉSUMÉ

BACKGROUND: Fabry disease (FD) is characterized by deficient activity of α-galactosidase A (GLA). Consequently, globotriaosylceramide (Gb3) accumulates in various organs, causing cardiac, renal, and cerebrovascular damage. Gene therapies for FD have been investigated in humans. Strong conditioning is required for hematopoietic stem cell-targeted gene therapy (HSC-GT). However, strong conditioning leads to various side effects and should be avoided. In this study, we tested antibody-based conditioning for HSC-GT in wild-type and FD model mice. METHODS: After preconditioning with an antibody-drug conjugate, HSC-GT using a lentiviral vector was performed in wild-type and Fabry model mice. In the wild-type experiment, the EGFP gene was introduced into HSCs and transplanted into preconditioned mice, and donor chimerism and EGFP expression were analyzed. In the FD mouse model, the GLA gene was introduced into HSCs and transplanted into preconditioned Fabry mice. GLA activity and Gb3 accumulation in the organs were analyzed. RESULTS: In the wild-type mouse experiment, when anti-CD45 antibody-drug conjugate was used, the percentage of donor cells at 6 months was 64.5%, and 69.6% of engrafted donor peripheral blood expressed EGFP. When anti-CD117 antibody-drug conjugate and ATG were used, the percentage of donor cells at 6 months was 80.7%, and 73.4% of engrafted donor peripheral blood expressed EGFP. Although large variations in GLA activity among mice were observed in the FD mouse experiment for both preconditioning regimens, Gb3 was significantly reduced in many organs. CONCLUSIONS: Antibody-based preconditioning may be an alternative preconditioning strategy for HSC-GT for treating FD.


Sujet(s)
Modèles animaux de maladie humaine , Maladie de Fabry , Thérapie génétique , Transplantation de cellules souches hématopoïétiques , Cellules souches hématopoïétiques , Trihexosylcéramide , alpha-Galactosidase , Animaux , Maladie de Fabry/thérapie , Maladie de Fabry/génétique , Souris , alpha-Galactosidase/génétique , alpha-Galactosidase/immunologie , Cellules souches hématopoïétiques/métabolisme , Trihexosylcéramide/métabolisme , Immunoconjugués/pharmacologie , Humains , Vecteurs génétiques/génétique , Vecteurs génétiques/administration et posologie , Lentivirus/génétique , Conditionnement pour greffe/méthodes
2.
Proc Natl Acad Sci U S A ; 121(19): e2322822121, 2024 May 07.
Article de Anglais | MEDLINE | ID: mdl-38687784

RÉSUMÉ

Hydrogels derived from decellularized extracellular matrices (ECM) of animal origin show immense potential for regenerative applications due to their excellent cytocompatibility and biomimetic properties. Despite these benefits, the impact of decellularization protocols on the properties and immunogenicity of these hydrogels remains relatively unexplored. In this study, porcine skeletal muscle ECM (smECM) underwent decellularization using mechanical disruption (MD) and two commonly employed decellularization detergents, sodium deoxycholate (SDC) or Triton X-100. To mitigate immunogenicity associated with animal-derived ECM, all decellularized tissues were enzymatically treated with α-galactosidase to cleave the primary xenoantigen-the α-Gal antigen. Subsequently, the impact of the different decellularization protocols on the resultant hydrogels was thoroughly investigated. All methods significantly reduced total DNA content in hydrogels. Moreover, α-galactosidase treatment was crucial for cleaving α-Gal antigens, suggesting that conventional decellularization methods alone are insufficient. MD preserved total protein, collagen, sulfated glycosaminoglycan, laminin, fibronectin, and growth factors more efficiently than other protocols. The decellularization method impacted hydrogel gelation kinetics and ultrastructure, as confirmed by turbidimetric and scanning electron microscopy analyses. MD hydrogels demonstrated high cytocompatibility, supporting satellite stem cell recruitment, growth, and differentiation into multinucleated myofibers. In contrast, the SDC and Triton X-100 protocols exhibited cytotoxicity. Comprehensive in vivo immunogenicity assessments in a subcutaneous xenotransplantation model revealed MD hydrogels' biocompatibility and low immunogenicity. These findings highlight the significant influence of the decellularization protocol on hydrogel properties. Our results suggest that combining MD with α-galactosidase treatment is an efficient method for preparing low-immunogenic smECM-derived hydrogels with enhanced properties for skeletal muscle regenerative engineering and clinical applications.


Sujet(s)
Matrice extracellulaire , Hydrogels , Muscles squelettiques , Animaux , Hydrogels/composition chimique , Suidae , Matrice extracellulaire/métabolisme , Ingénierie tissulaire/méthodes , Matrice extracellulaire décellularisée/composition chimique , Souris , alpha-Galactosidase/immunologie , alpha-Galactosidase/métabolisme , Acide désoxycholique/composition chimique , Octoxinol/composition chimique
5.
CEN Case Rep ; 13(4): 290-296, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38135868

RÉSUMÉ

Fabry disease is an X-linked hereditary disorder caused by deficient α-galactosidase A (GLA) activity. Patients with Fabry disease are often treated with enzyme replacement therapy (ERT). However, ERT often induces the formation of neutralizing antidrug antibodies (ADAs), which may impair the therapeutic efficacy. Here, we report the case of a 32-year-old man with Fabry disease and resultant neutralizing ADAs who was treated by switching from agalsidase-α to agalsidase-ß. We monitored biomarkers, such as plasma globotriaosylsphingosine (lyso-Gb3), urinary globotriaosylceramide (Gb3), urinary mulberry bodies, renal and cardiac parameters, and disease severity during the treatment period. Although plasma lyso-Gb3 and urinary Gb3 levels quickly decreased within two months after the initiation of ERT with agalsidase-α, they gradually increased thereafter. The urinary mulberry bodies continued to appear. Both the ADA titer and serum mediated GLA inhibition rates started to increase after two months. Moreover, 3.5 years after ERT, the vacuolated podocyte area in the renal biopsy decreased slightly from 23.1 to 18.9%. However, plasma lyso-Gb3 levels increased, and urinary Gb3, mulberry body levels, and ADA titers remained high. Therefore, we switched to agalsidase-ß which reduced, but did not normalize, plasma lyso-Gb3 levels and stabilized renal and cardiac parameters. Disease severity was attenuated. However, urinary Gb3 and mulberry body levels did not decrease noticeably in the presence of high ADA titers. The kidneys take up a small amount of the administered recombinant enzyme, and the clearance of Gb3 that has accumulated in the kidney may be limited despite the switching from agalsidase-α to agalsidase-ß.


Sujet(s)
Anticorps neutralisants , Marqueurs biologiques , Thérapie enzymatique substitutive , Maladie de Fabry , Isoenzymes , Sphingolipides , Trihexosylcéramide , alpha-Galactosidase , Humains , Maladie de Fabry/traitement médicamenteux , Maladie de Fabry/diagnostic , Mâle , Adulte , alpha-Galactosidase/usage thérapeutique , alpha-Galactosidase/administration et posologie , alpha-Galactosidase/immunologie , Marqueurs biologiques/sang , Thérapie enzymatique substitutive/méthodes , Isoenzymes/usage thérapeutique , Isoenzymes/administration et posologie , Anticorps neutralisants/sang , Trihexosylcéramide/urine , Sphingolipides/sang , Glycolipides , Rein/anatomopathologie , Indice de gravité de la maladie , Résultat thérapeutique , Protéines recombinantes
7.
Int J Mol Sci ; 22(5)2021 Mar 06.
Article de Anglais | MEDLINE | ID: mdl-33800950

RÉSUMÉ

Male patients with Fabry disease (FD) are at high risk for the formation of antibodies to recombinant α-galactosidase A (AGAL), used for enzyme replacement therapy. Due to the rapid disease progression, the identification of patients at risk is highly warranted. However, currently suitable references and standardized protocols for anti-drug antibodies (ADA) determination do not exist. Here we generate a comprehensive patient-derived antibody mixture as a reference, allowing ELISA-based quantification of antibody titers from individual blood samples. Serum samples of 22 male patients with FD and ADAs against AGAL were pooled and purified by immune adsorption. ADA-affinities against agalsidase-α, agalsidase-ß and Moss-AGAL were measured by quartz crystal microbalance with dissipation monitoring (QCM-D). AGAL-specific immune adsorption generated a polyclonal ADA mixture showing a concentration-dependent binding and inhibition of AGAL. Titers in raw sera and from purified total IgGs (r2 = 0.9063 and r2 = 0.8952, both p < 0.0001) correlated with the individual inhibitory capacities of ADAs. QCM-D measurements demonstrated comparable affinities of the reference antibody for agalsidase-α, agalsidase-ß and Moss-AGAL (KD: 1.94 ± 0.11 µM, 2.46 ± 0.21 µM, and 1.33 ± 0.09 µM, respectively). The reference antibody allows the ELISA-based ADA titer determination and quantification of absolute concentrations. Furthermore, ADAs from patients with FD have comparable affinities to agalsidase-α, agalsidase-ß and Moss-AGAL.


Sujet(s)
Anticorps neutralisants/immunologie , Thérapie enzymatique substitutive , Test ELISA , Maladie de Fabry/immunologie , alpha-Galactosidase/immunologie , alpha-N-Acetylgalactosaminidase/immunologie , Anticorps neutralisants/biosynthèse , Affinité des anticorps , Relation dose-réponse (immunologie) , Maladie de Fabry/sang , Maladie de Fabry/traitement médicamenteux , Humains , Immunoglobuline G/biosynthèse , Immunoglobuline G/immunologie , Mâle , Protéines recombinantes/immunologie , Protéines recombinantes/usage thérapeutique , Normes de référence , alpha-Galactosidase/sang , alpha-Galactosidase/usage thérapeutique , alpha-N-Acetylgalactosaminidase/sang , alpha-N-Acetylgalactosaminidase/usage thérapeutique
8.
Int J Mol Sci ; 22(3)2021 Jan 30.
Article de Anglais | MEDLINE | ID: mdl-33573215

RÉSUMÉ

This study was conducted to explore whether trichostatin A-assisted epigenomic modulation (TSA-EM) can affect the expression of not only recombinant human α1,2-fucosyltransferase (rhα1,2-FT) and α-galactosidase A (rhα-Gal A) immune system enzymes but also Galα1→3Gal epitopes in ex vivo proliferating adult cutaneous fibroblast cells (ACFCs) derived from hFUT2×hGLA bi-transgenic pigs that had been produced for the needs of future xenotransplantation efforts. The ACFC lines were treated with 50 nM TSA for 24 h and then the expression profiles of rhα1,2-FT and rhα-Gal A enzymes were analyzed by Western blot and immunofluorescence. The expression profiles of the Galα1→3Gal epitope were determined by lectin blotting and lectin fluorescence. The ACFCs derived from non-transgenic (nTG) pigs were served as the negative (TSA-) and positive (TSA+) control groups. For both hFUT2×hGLA and nTG samples, the expression levels of α1,2-FT and α-Gal A proteins in TSA+ cells were more than twofold higher in comparison to TSA- cells. Moreover, a much lower expression of the Galα1→3Gal epitopes was shown in TSA- hFUT2×hGLA cells as compared to the TSA- nTG group. Interestingly, the levels of Galα1→3Gal expression in TSA-treated hFUT2×hGLA and nTG ACFCs were significantly higher than those noticed for their TSA-untreated counterparts. Summing up, ex vivo protection of effectively selected bi-transgenic ACFC lines, in which TSA-dependent epigenetic transformation triggered the enhancements in reprogrammability and subsequent expression of hFUT2 and hGLA transgenes and their corresponding transcripts, allows for cryopreservation of nuclear donor cells, nuclear-transferred female gametes, and resultant porcine cloned embryos. The latter can be used as a cryogenically conserved genetic resource of biological materials suitable for generation of bi-transgenic cloned offspring in pigs that is targeted at biomedical research in the field of cell/tissue xenotransplantation.


Sujet(s)
Épigenèse génétique/effets des médicaments et des substances chimiques , Épitopes/métabolisme , Rejet du greffon/prévention et contrôle , Acides hydroxamiques/pharmacologie , Transplantation hétérologue/effets indésirables , Animaux , Animal génétiquement modifié , Lignée cellulaire , Clonage d'organisme/méthodes , Cryoconservation , Embryon de mammifère , Épitopes/génétique , Épitopes/immunologie , Fibroblastes , Fucosyltransferases/génétique , Fucosyltransferases/immunologie , Fucosyltransferases/métabolisme , Techniques de knock-out de gènes , Rejet du greffon/immunologie , Humains , Protéines recombinantes/génétique , Protéines recombinantes/métabolisme , Peau/cytologie , Suidae , Transplantation hétérologue/méthodes , alpha-Galactosidase/génétique , alpha-Galactosidase/immunologie , alpha-Galactosidase/métabolisme ,
9.
Scand J Immunol ; 93(5): e13008, 2021 May.
Article de Anglais | MEDLINE | ID: mdl-33314191

RÉSUMÉ

In clinical practice, the capacity for producing anti-carbohydrate antibodies is regarded as an entity, but supportive evidence is lacking. We hypothesized that the outcome of the gold standard test for clinical assessment of this capacity, antibody response to polysaccharide vaccination, correlated with the level of the abundant naturally occurring anti-carbohydrate antibody, anti-αGal. To perform an exploratory study, 47 HIV-infected adults were recruited from a vaccine trial. Participants received a 23-valent pneumococcal capsular polysaccharide vaccine. Plasma samples obtained just before and median 4 weeks after the vaccination were quantified for IgG anti-αGal antibody and IgG antibodies to polysaccharides present in the vaccine (serotypes 1, 7F and 19A) by solid-phase type immunoassays. The vaccination responses were assessed as a categorical variable (based on criteria defined by The American Academy of Allergy, Asthma & Immunology and the American College of Allergy, Asthma & Immunology) and as three different continuous variables (antibody increment, geometrical average and standard normal deviates of the achieved antibody concentrations). The baseline anti-αGal level predicted the vaccine response as a categorical variable (ROC-curve analysis, AUC = 0.71; 95%CI: 0.55-0.86) and as the three continuous variables (eg slope of linear regression of geometrical average = 0.37; 95%CI: 0.15-0.59). The correlation between the anti-αGal level and antibody responses to polysaccharide vaccination fits with a shared underlying capacity. Thus, the present study supports the notion of a measurable capacity for the production of anti-carbohydrate antibodies in each individual. Firm conclusions on the generalizability and clinical utility require further studies.


Sujet(s)
Anticorps antibactériens/sang , Infections à VIH/immunologie , Vaccins antipneumococciques/immunologie , Polyosides bactériens/immunologie , alpha-Galactosidase/immunologie , Anticorps antibactériens/immunologie , Humains , Immunoglobuline G/sang , Immunoglobuline G/immunologie , Lymphocytes T auxiliaires/cytologie , Lymphocytes T auxiliaires/immunologie
10.
Int J Mol Sci ; 21(19)2020 Sep 29.
Article de Anglais | MEDLINE | ID: mdl-33003611

RÉSUMÉ

Fabry disease (FD) is an X-linked lysosomal disorder caused by mutations in GLA gene resulting in lack of or faulty α-galactosidase A (α-GalA) enzyme. Enzyme replacement therapy (ERT) with recombinant human α-GalA enzyme (agalsidase) is the standard treatment option for FD. Infusion-related reactions (IRRs), with symptoms ranging from rigors, to fever, pain, vomiting, angioedema and diarrhea, are often seen due to immune response against the exogenous enzyme. To elucidate the mechanisms causing the IRRs in FD, eight patients who developed IRRs were investigated. All, except one, tested negative for agalsidase-specific IgE and had normal tryptase levels. Circulating dendritic cells were drastically reduced during IRRs, suggesting possible sequestration to the sites of inflammation. An increase in NK cells and a decrease in T cells were also observed. Cytokines IL-4, IL-8 and TNF-α showed a significant increase, indicating nonspecific degranulation of mast cells. All IRRs were managed successfully using a combination of standard premedications and mast cell stabilizers without any interruption of therapy. Taken together, the results indicate crosstalk between immune cells resulting in IgE-independent mast-cell-specific allergic inflammation. Mast cell stabilizers could be used to control IRRs and for safe reintroduction of agalsidase in patients previously treated with ERT.


Sujet(s)
Maladie de Fabry/traitement médicamenteux , Maladie de Fabry/immunologie , Inflammation/immunologie , Isoenzymes/immunologie , Protéines recombinantes/immunologie , alpha-Galactosidase/immunologie , Immunité acquise/effets des médicaments et des substances chimiques , Immunité acquise/immunologie , Adolescent , Adulte , Enfant , Thérapie enzymatique substitutive/effets indésirables , Maladie de Fabry/génétique , Maladie de Fabry/anatomopathologie , Humains , Immunité innée/effets des médicaments et des substances chimiques , Immunité innée/immunologie , Immunoglobuline E/immunologie , Inflammation/anatomopathologie , Réaction au site d'injection/génétique , Réaction au site d'injection/immunologie , Isoenzymes/administration et posologie , Mâle , Mastocytes/effets des médicaments et des substances chimiques , Mastocytes/immunologie , Adulte d'âge moyen , Protéines recombinantes/administration et posologie , Jeune adulte , alpha-Galactosidase/administration et posologie
11.
Mol Genet Metab ; 131(1-2): 229-234, 2020.
Article de Anglais | MEDLINE | ID: mdl-32888778

RÉSUMÉ

BACKGROUND: Fabry disease (FD) is a lysosomal storage disease, treatable by enzyme replacement therapy (ERT) that substitutes deficient α-galactosidase A (AGAL). The formation of neutralizing anti-drug antibodies (ADA) inhibiting AGAL activity is associated with disease progression in affected male patients. In the current study, we performed a detailed epitope mapping of ADAs from antibody-positive males against infused AGAL. METHODS: A detailed epitope mapping for 34 male FD patients with neutralizing ADAs against AGAL was performed. Based on this data, in silico analyses were used to identify potential epitope clusters and mapped surface-located or buried epitopes. ELISA-based assays against α-galactosidase B (NAGA) were performed to identify ADAs that potentially recognize shared epitopes of AGAL and NAGA. A subset of 20 patients was analyzed to assess if NAGA-recognizing ADAs against AGAL might affect long-term outcomes under ERT. RESULTS: Thirty percent of the AGAL active site was recognized by patients' ADAs. No differences between buried and surface-located epitopes were observed. Dependent on the epitopes, ADAs against AGAL were also able to recognize human NAGA. Patients with NAGA recognizing anti-AGAL antibodies presented with lower plasma NAGA activities. The presence of NAGA-recognizing ADAs had no effect on disease progression. CONCLUSION: In conclusion, our current data underline previous reports demonstrating a large variation of antibody epitopes against AGAL. Detailed epitope mapping in affected patients might be the first step for the generation of patient-specific blocking peptides and/or immune adsorption columns for an individually tailored anti-antibody strategy.


Sujet(s)
Anticorps neutralisants/immunologie , Thérapie enzymatique substitutive , Épitopes/génétique , Maladie de Fabry/traitement médicamenteux , alpha-Galactosidase/génétique , Adulte , Anticorps anti-idiotypiques/génétique , Anticorps anti-idiotypiques/immunologie , Anticorps neutralisants/administration et posologie , Anticorps neutralisants/génétique , Cartographie épitopique , Épitopes/immunologie , Maladie de Fabry/génétique , Maladie de Fabry/immunologie , Maladie de Fabry/anatomopathologie , Humains , Mâle , Protéines recombinantes/administration et posologie , Protéines recombinantes/génétique , Protéines recombinantes/immunologie , alpha-Galactosidase/administration et posologie , alpha-Galactosidase/immunologie
12.
Int J Mol Sci ; 21(16)2020 Aug 12.
Article de Anglais | MEDLINE | ID: mdl-32806627

RÉSUMÉ

Fabry Disease (FD) is a rare, X-linked, lysosomal storage disease that mainly causes renal, cardiac and cerebral complications. Enzyme replacement therapy (ERT) with recombinant alpha-galactosidase A is available, but approximately 50% of male patients with classical FD develop inhibiting anti-drug antibodies (iADAs) that lead to reduced biochemical responses and an accelerated loss of renal function. Once immunization has occurred, iADAs tend to persist and tolerization is hard to achieve. Here we developed a pre-treatment prediction model for iADA development in FD using existing data from 120 classical male FD patients from three European centers, treated with ERT. We found that nonsense and frameshift mutations in the α-galactosidase A gene (p = 0.05), higher plasma lysoGb3 at baseline (p < 0.001) and agalsidase beta as first treatment (p = 0.006) were significantly associated with iADA development. Prediction performance of a Random Forest model, using multiple variables (AUC-ROC: 0.77) was compared to a logistic regression (LR) model using the three significantly associated variables (AUC-ROC: 0.77). The LR model can be used to determine iADA risk in individual FD patients prior to treatment initiation. This helps to determine in which patients adjusted treatment and/or immunomodulatory regimes may be considered to minimize iADA development risk.


Sujet(s)
Anticorps/immunologie , Maladie de Fabry/traitement médicamenteux , Maladie de Fabry/immunologie , Isoenzymes/immunologie , Isoenzymes/usage thérapeutique , Protéines recombinantes/immunologie , Protéines recombinantes/usage thérapeutique , alpha-Galactosidase/immunologie , alpha-Galactosidase/usage thérapeutique , Adolescent , Adulte , Algorithmes , Aire sous la courbe , Enfant , Études de cohortes , Humains , Modèles logistiques , Mâle , Adulte d'âge moyen , Courbe ROC , Facteurs de risque , Jeune adulte
13.
Biomed Res Int ; 2020: 9680474, 2020.
Article de Anglais | MEDLINE | ID: mdl-32596401

RÉSUMÉ

BACKGROUND: Animal tissues and tissue-derived biomaterials are widely used in the field of xenotransplantation and regenerative medicine. A potential immunogenic risk that affects the safety and effectiveness of xenografts is the presence of remnant α-Gal antigen (synthesized by GGTA1 or/and iGb3S). GGTA1 knockout mice have been developed as a suitable model for the analysis of anti-Gal antibody-mediated immunogenicity. However, we are yet to establish whether GGTA1/iGb3S double knockout (G/i DKO) mice are sensitive to Gal antigen-positive xenoimplants. METHODS: α-Gal antigen expression in the main organs of G/i DKO mice or bovine bone substitutes was detected via a standardized ELISA inhibition assay. Serum anti-α-Gal antibody titers of G/i DKO mice after immunization with rabbit red blood cells (RRBC) and implantation of raw lyophilized bone substitutes (Gal antigen content was 8.14 ± 3.17 × 1012/mg) or Guanhao Biotech bone substitutes (50% decrease in Gal antigen relative to the raw material) were assessed. The evaluation of total serum antibody, inflammatory cytokine, and splenic lymphocyte subtype populations and the histological analysis of implants and thymus were performed to systematically assess the immune response caused by bovine bone substitutes and bone substitute grafts in G/i DKO mice. RESULTS: α-Gal epitope expression was reduced by 100% in the main organs of G/i DKO mice, compared with their wild-type counterparts. Following immunization with RRBC, serum anti-Gal antibody titers of G/i DKO mice increased from 80- to 180-fold. After subcutaneous implantation of raw lyophilized bone substitutes and Guanhao Biotech bone substitutes into G/i DKO mice, specific anti-α-Gal IgG, anti-α-Gal IgM, and related inflammatory factors (IFN-γ and IL-6) were significantly increased in the raw lyophilized bone substitute group but showed limited changes in the Guanhao Biotech bone substitute group, compared with the control. CONCLUSION: G/i DKO mice are sensitive to Gal antigen-positive xenogeneic grafts and can be effectively utilized for evaluating the α-Gal-mediated immunogenic risk of xenogeneic grafts.


Sujet(s)
Trame osseuse , Galactosyltransferases/génétique , Hétérogreffes/immunologie , Transplantation hétérologue , Animaux , Trame osseuse/immunologie , Trame osseuse/transplantation , Substituts osseux , Bovins , Érythrocytes/immunologie , Galactosyltransferases/métabolisme , Souris , Souris knockout , Lapins , alpha-Galactosidase/immunologie
14.
J Immunol ; 204(7): 1998-2005, 2020 04 01.
Article de Anglais | MEDLINE | ID: mdl-32144163

RÉSUMÉ

Mice have been used as accepted tools for investigating complex human diseases and new drug therapies because of their shared genetics and anatomical characteristics with humans. However, the tissues in mice are different from humans in that human cells have a natural mutation in the α1,3 galactosyltransferase (α1,3GT) gene and lack α-Gal epitopes on glycosylated proteins, whereas mice and other nonprimate mammals express this epitope. The lack of α-Gal epitopes in humans results in the loss of immune tolerance to this epitope and production of abundant natural anti-Gal Abs. These natural anti-Gal Abs can be used as an adjuvant to enhance processing of vaccine epitopes to APCs. However, wild-type mice and all existing humanized mouse models cannot be used to test the efficacy of vaccines expressing α-Gal epitopes because they express α-Gal epitopes and lack anti-Gal Abs. Therefore, in an effort to bridge the gap between the mouse models and humans, we developed a new humanized mouse model that mimics humans in that it lacks α-Gal epitopes and secretes human anti-Gal Abs. The new humanized mouse model (Hu-NSG/α-Galnull) is designed to be used for preclinical evaluations of viral and tumor vaccines based on α-Gal epitopes, human-specific immune responses, xenotransplantation studies, and in vivo biomaterials evaluation. To our knowledge, our new Hu-NSG/α-Galnull is the first available humanized mouse model with such features.


Sujet(s)
Anticorps/immunologie , Épitopes/immunologie , Galactosyltransferases/immunologie , alpha-Galactosidase/immunologie , Animaux , Vaccins anticancéreux/immunologie , Modèles animaux de maladie humaine , Humains , Souris , Souris de lignée C57BL , Transplantation hétérologue/méthodes
16.
J Allergy Clin Immunol ; 145(4): 1061-1071, 2020 04.
Article de Anglais | MEDLINE | ID: mdl-32057766

RÉSUMÉ

The mammalian meat allergy known as the "α-Gal syndrome" relates to IgE specific for galactose-α-1,3-galactose (α-Gal), an oligosaccharide that is present in cells and tissues of nonprimate mammals. The recognition of delayed reactions to food derived from mammals in patients with IgE to α-Gal and also the association with tick bites have been increasing worldwide. In 2018, the National Institute of Allergy and Infectious Diseases, Division of Allergy, Immunology and Transplantation, sponsored a workshop on this emerging tick-related disease. International experts from the fields of tick biology, allergy, immunology, infectious disease, and dermatology discussed the current state of our understanding of this emerging medical condition. The participants provided suggestions for specific research priorities and for the development of resources to advance our knowledge of the mechanisms, diagnosis, management, and prevention of this allergic disease. This publication is a summary of the workshop and the panel's recommendations are presented herein.


Sujet(s)
Allergènes/immunologie , Hypersensibilité alimentaire/immunologie , Protéines de viande/immunologie , Maladies transmises par les tiques/immunologie , alpha-Galactosidase/immunologie , Animaux , Hypersensibilité alimentaire/diagnostic , Hypersensibilité alimentaire/thérapie , Immunoglobuline E/métabolisme , National Institute of Allergy and Infectious Diseases (USA) , Maladies transmises par les tiques/diagnostic , Maladies transmises par les tiques/thérapie , Tiques , États-Unis
17.
Ann Allergy Asthma Immunol ; 124(2): 135-142, 2020 02.
Article de Anglais | MEDLINE | ID: mdl-31785367

RÉSUMÉ

OBJECTIVE: To summarize the current understanding of anaphylaxis, with an emphasis on major findings that have been reported within the last 10 years. DATA SOURCES: Queries relating to anaphylaxis, immunoglobulin E (IgE), and mast cells were conducted with PubMed and Google Scholar, searching for primary articles and review papers. STUDY SELECTIONS: We focused on articles written in English and which were reported in major allergy and immunology journals. RESULTS: Anaphylaxis represents an extreme manifestation of a form of allergic immunity that appears to have evolved to protect against "toxic" threats that present at skin and mucosal barriers. The factors that have contributed to a rise in anaphylaxis are increasingly appreciated to relate to changes in hygiene and microbial ecology that have occurred with industrialization. Induction of allergen-specific IgG4 is often part of the allergic response and is associated with protection against anaphylaxis. The recognition of the α-Gal syndrome suggests that carbohydrates can be epitopes that are relevant to anaphylaxis and that IgE-mediated reactions do not always occur "immediately." CONCLUSION: Our understanding of anaphylaxis has advanced significantly over the past 10 years. It is anticipated that ongoing research will build on this foundation to further advance our knowledge of anaphylaxis and also translate into clinically meaningful therapies.


Sujet(s)
Allergènes/immunologie , Anaphylaxie/étiologie , alpha-Galactosidase/immunologie , Animaux , Hypersensibilité alimentaire/immunologie , Humains , Immunoglobuline E/immunologie , Immunoglobuline G/immunologie , Microbiote
18.
Allergol Immunopathol (Madr) ; 48(3): 251-258, 2020.
Article de Anglais | MEDLINE | ID: mdl-31718865

RÉSUMÉ

INTRODUCTION AND OBJECTIVES: Allergy to galactose-α-1,3-galactose (alpha-gal) is a peculiar form of food allergy generally manifesting as an anaphylactic reaction hours after mammalian meat consumption, due to the presence of specific IgE against this oligosaccharide. In addition, immediate anaphylaxis may develop after exposure to other sources of alpha-gal, such as monoclonal antibody cetuximab, vaccines, plasma expanders or anti-snake venoms. Sensitization to alpha-gal has also been implicated in the rapid degeneration of biological valve implants, and recognized as a cause of occupational disease in cattle raisers. The implication of tick bites in this type of sensitization has been accepted by all the research groups dedicated to this disease. PATIENTS AND METHOD: The present study describes the clinical and sensitization characteristics of 39 patients diagnosed with alpha-gal allergy in the hospitals of our province (Lugo, Monforte de Lemos and Burela, Spain). RESULTS: Most patients were middle-age males. Of note, is the fact that the series includes the first pediatric patient reported in Spain to date. The predominant clinical manifestations were urticaria or delayed anaphylaxis after consumption of mammalian meat. Seventy-four percent of the patients reported having suffered a previous tick bite, and the clinical presentation of anaphylaxis was significantly more prevalent in those with a persistent local reaction following the bite than in those with no such reaction (p = 0.032). CONCLUSIONS: A review is also made of the disorder which, due to its variable clinical expression, is referred to as alpha-gal syndrome. The study concludes that a diagnosis of alpha-gal allergy should be considered in patients with urticaria-anaphylaxis of uncertain origin or manifesting after the administration of vaccines or products of bovine/porcine origin.


Sujet(s)
Anaphylaxie/immunologie , Hypersensibilité alimentaire/immunologie , Morsures de tiques/épidémiologie , Urticaire/immunologie , Adolescent , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Allergènes/immunologie , Anaphylaxie/épidémiologie , Femelle , Hypersensibilité alimentaire/épidémiologie , Humains , Mâle , Viande , Adulte d'âge moyen , Espagne/épidémiologie , Urticaire/épidémiologie , Jeune adulte , alpha-Galactosidase/immunologie
19.
J Inherit Metab Dis ; 43(2): 334-347, 2020 03.
Article de Anglais | MEDLINE | ID: mdl-31587315

RÉSUMÉ

Fabry disease (FD) is a lysosomal storage disease, treatable by enzyme replacement therapy (ERT) that substitutes deficient α-galactosidase A (AGAL). The formation of neutralising anti-drug antibodies (ADA) inhibiting AGAL activity during infusion is associated with disease progression in affected male patients. In this study we analysed if ADAs also inhibit endothelial enzyme uptake as well as intracellular enzyme activity. Therefore, fluorescence-labelled AGAL in combination with ADA-positive sera from FD patients (n = 8) was used to analyse enzyme uptake in endothelial and FD-specific cells. Furthermore, immune adsorption and a comprehensive ADA epitope mapping were performed. Pre-incubation of AGAL with ADAs significantly inhibited intracellular enzyme activity, which was rescued by immune adsorption (both P < .01). ADAs from some patients also inhibited enzyme uptake. ADA epitope mapping identified an epitope at position 121 to 140 aa potentially responsible for uptake inhibition for these patients. Further analyses revealed the presence of stable AGAL/ADA-immune complexes at pH 4.5 and decreased intracellular enzyme activity in endothelial cells (P < .001). Finally, the pre-incubation of AGAL with ADAs resulted in a reduced depletion of intracellular globotriaosylceramide in patient-derived AGAL-deficient cells, demonstrating a direct negative impact of ADAs on intracellular clearance. Neutralising ADAs may not only inhibit infused AGAL activity, but according to their epitopes can also inhibit endothelial AGAL uptake. Indeed, internalised AGAL/ADA-complexes may not dissociate, underlining the importance of novel therapeutic approaches for ADA reduction and prevention to increase therapy efficiency in affected patients.


Sujet(s)
Anticorps neutralisants/immunologie , Thérapie enzymatique substitutive , Maladie de Fabry/immunologie , alpha-Galactosidase/immunologie , Adulte , Anticorps neutralisants/biosynthèse , Test ELISA , Maladie de Fabry/sang , Maladie de Fabry/traitement médicamenteux , Cytométrie en flux , Humains , Mâle , Adulte d'âge moyen , alpha-Galactosidase/sang , alpha-Galactosidase/usage thérapeutique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE