Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 134
Filter
1.
Proc Natl Acad Sci U S A ; 121(19): e2322822121, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38687784

ABSTRACT

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.


Subject(s)
Extracellular Matrix , Hydrogels , Muscle, Skeletal , Animals , Hydrogels/chemistry , Swine , Extracellular Matrix/metabolism , Tissue Engineering/methods , Decellularized Extracellular Matrix/chemistry , Mice , alpha-Galactosidase/immunology , alpha-Galactosidase/metabolism , Deoxycholic Acid/chemistry , Octoxynol/chemistry
2.
Mol Genet Metab ; 142(3): 108494, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38820907

ABSTRACT

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.


Subject(s)
Disease Models, Animal , Fabry Disease , Genetic Therapy , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells , Trihexosylceramides , alpha-Galactosidase , Animals , Fabry Disease/therapy , Fabry Disease/genetics , Mice , alpha-Galactosidase/genetics , alpha-Galactosidase/immunology , Hematopoietic Stem Cells/metabolism , Trihexosylceramides/metabolism , Immunoconjugates/pharmacology , Humans , Genetic Vectors/genetics , Genetic Vectors/administration & dosage , Lentivirus/genetics , Transplantation Conditioning/methods
3.
J Immunol ; 204(7): 1998-2005, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32144163

ABSTRACT

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.


Subject(s)
Antibodies/immunology , Epitopes/immunology , Galactosyltransferases/immunology , alpha-Galactosidase/immunology , Animals , Cancer Vaccines/immunology , Disease Models, Animal , Humans , Mice , Mice, Inbred C57BL , Transplantation, Heterologous/methods
4.
Scand J Immunol ; 93(5): e13008, 2021 May.
Article in English | MEDLINE | ID: mdl-33314191

ABSTRACT

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.


Subject(s)
Antibodies, Bacterial/blood , HIV Infections/immunology , Pneumococcal Vaccines/immunology , Polysaccharides, Bacterial/immunology , alpha-Galactosidase/immunology , Antibodies, Bacterial/immunology , Humans , Immunoglobulin G/blood , Immunoglobulin G/immunology , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/immunology
5.
J Allergy Clin Immunol ; 145(4): 1061-1071, 2020 04.
Article in English | MEDLINE | ID: mdl-32057766

ABSTRACT

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.


Subject(s)
Allergens/immunology , Food Hypersensitivity/immunology , Meat Proteins/immunology , Tick-Borne Diseases/immunology , alpha-Galactosidase/immunology , Animals , Food Hypersensitivity/diagnosis , Food Hypersensitivity/therapy , Immunoglobulin E/metabolism , National Institute of Allergy and Infectious Diseases (U.S.) , Tick-Borne Diseases/diagnosis , Tick-Borne Diseases/therapy , Ticks , United States
6.
Int J Mol Sci ; 22(5)2021 Mar 06.
Article in English | MEDLINE | ID: mdl-33800950

ABSTRACT

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.


Subject(s)
Antibodies, Neutralizing/immunology , Enzyme Replacement Therapy , Enzyme-Linked Immunosorbent Assay , Fabry Disease/immunology , alpha-Galactosidase/immunology , alpha-N-Acetylgalactosaminidase/immunology , Antibodies, Neutralizing/biosynthesis , Antibody Affinity , Dose-Response Relationship, Immunologic , Fabry Disease/blood , Fabry Disease/drug therapy , Humans , Immunoglobulin G/biosynthesis , Immunoglobulin G/immunology , Male , Recombinant Proteins/immunology , Recombinant Proteins/therapeutic use , Reference Standards , alpha-Galactosidase/blood , alpha-Galactosidase/therapeutic use , alpha-N-Acetylgalactosaminidase/blood , alpha-N-Acetylgalactosaminidase/therapeutic use
7.
Int J Mol Sci ; 22(3)2021 Jan 30.
Article in English | MEDLINE | ID: mdl-33573215

ABSTRACT

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.


Subject(s)
Epigenesis, Genetic/drug effects , Epitopes/metabolism , Graft Rejection/prevention & control , Hydroxamic Acids/pharmacology , Transplantation, Heterologous/adverse effects , Animals , Animals, Genetically Modified , Cell Line , Cloning, Organism/methods , Cryopreservation , Embryo, Mammalian , Epitopes/genetics , Epitopes/immunology , Fibroblasts , Fucosyltransferases/genetics , Fucosyltransferases/immunology , Fucosyltransferases/metabolism , Gene Knockout Techniques , Graft Rejection/immunology , Humans , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Skin/cytology , Swine , Transplantation, Heterologous/methods , alpha-Galactosidase/genetics , alpha-Galactosidase/immunology , alpha-Galactosidase/metabolism , Galactoside 2-alpha-L-fucosyltransferase
8.
Mol Genet Metab ; 131(1-2): 229-234, 2020.
Article in English | MEDLINE | ID: mdl-32888778

ABSTRACT

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.


Subject(s)
Antibodies, Neutralizing/immunology , Enzyme Replacement Therapy , Epitopes/genetics , Fabry Disease/drug therapy , alpha-Galactosidase/genetics , Adult , Antibodies, Anti-Idiotypic/genetics , Antibodies, Anti-Idiotypic/immunology , Antibodies, Neutralizing/administration & dosage , Antibodies, Neutralizing/genetics , Epitope Mapping , Epitopes/immunology , Fabry Disease/genetics , Fabry Disease/immunology , Fabry Disease/pathology , Humans , Male , Recombinant Proteins/administration & dosage , Recombinant Proteins/genetics , Recombinant Proteins/immunology , alpha-Galactosidase/administration & dosage , alpha-Galactosidase/immunology
9.
J Inherit Metab Dis ; 43(2): 334-347, 2020 03.
Article in English | MEDLINE | ID: mdl-31587315

ABSTRACT

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.


Subject(s)
Antibodies, Neutralizing/immunology , Enzyme Replacement Therapy , Fabry Disease/immunology , alpha-Galactosidase/immunology , Adult , Antibodies, Neutralizing/biosynthesis , Enzyme-Linked Immunosorbent Assay , Fabry Disease/blood , Fabry Disease/drug therapy , Flow Cytometry , Humans , Male , Middle Aged , alpha-Galactosidase/blood , alpha-Galactosidase/therapeutic use
10.
Ann Allergy Asthma Immunol ; 124(2): 135-142, 2020 02.
Article in English | MEDLINE | ID: mdl-31785367

ABSTRACT

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.


Subject(s)
Allergens/immunology , Anaphylaxis/etiology , alpha-Galactosidase/immunology , Animals , Food Hypersensitivity/immunology , Humans , Immunoglobulin E/immunology , Immunoglobulin G/immunology , Microbiota
13.
Allergol Immunopathol (Madr) ; 48(3): 251-258, 2020.
Article in English | MEDLINE | ID: mdl-31718865

ABSTRACT

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.


Subject(s)
Anaphylaxis/immunology , Food Hypersensitivity/immunology , Tick Bites/epidemiology , Urticaria/immunology , Adolescent , Adult , Aged , Aged, 80 and over , Allergens/immunology , Anaphylaxis/epidemiology , Female , Food Hypersensitivity/epidemiology , Humans , Male , Meat , Middle Aged , Spain/epidemiology , Urticaria/epidemiology , Young Adult , alpha-Galactosidase/immunology
14.
Int J Mol Sci ; 21(16)2020 Aug 12.
Article in English | MEDLINE | ID: mdl-32806627

ABSTRACT

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.


Subject(s)
Antibodies/immunology , Fabry Disease/drug therapy , Fabry Disease/immunology , Isoenzymes/immunology , Isoenzymes/therapeutic use , Recombinant Proteins/immunology , Recombinant Proteins/therapeutic use , alpha-Galactosidase/immunology , alpha-Galactosidase/therapeutic use , Adolescent , Adult , Algorithms , Area Under Curve , Child , Cohort Studies , Humans , Logistic Models , Male , Middle Aged , ROC Curve , Risk Factors , Young Adult
15.
Int J Mol Sci ; 21(19)2020 Sep 29.
Article in English | MEDLINE | ID: mdl-33003611

ABSTRACT

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.


Subject(s)
Fabry Disease/drug therapy , Fabry Disease/immunology , Inflammation/immunology , Isoenzymes/immunology , Recombinant Proteins/immunology , alpha-Galactosidase/immunology , Adaptive Immunity/drug effects , Adaptive Immunity/immunology , Adolescent , Adult , Child , Enzyme Replacement Therapy/adverse effects , Fabry Disease/genetics , Fabry Disease/pathology , Humans , Immunity, Innate/drug effects , Immunity, Innate/immunology , Immunoglobulin E/immunology , Inflammation/pathology , Injection Site Reaction/genetics , Injection Site Reaction/immunology , Isoenzymes/administration & dosage , Male , Mast Cells/drug effects , Mast Cells/immunology , Middle Aged , Recombinant Proteins/administration & dosage , Young Adult , alpha-Galactosidase/administration & dosage
16.
Mol Genet Metab ; 126(2): 162-168, 2019 02.
Article in English | MEDLINE | ID: mdl-30473480

ABSTRACT

BACKGROUND: Treatment of Fabry disease (FD) with recombinant alpha-galactosidase A (r-αGAL A) is complicated by the formation of anti-drug antibodies in the majority of male patients with the classical disease phenotype. Detailed information regarding antibody subtypes, onset and persistence of antibody development and their effect on treatment efficacy is sparse. METHODS: A retrospective study was carried out in 39 male patients with classical FD, treated with either agalsidase-alfa or agalsidase-beta (mean follow up of 10 years). With six to twelve months intervals plasma-induced in vitro inhibition of enzyme activity, lysoglobotriaosylsphingosine (lysoGb3) levels and renal function were assessed. In a subset of 12 patients, additionally anti- r-αGAL A IgM, IgA and IgG1, 2, 3 and 4 levels were analyzed. RESULTS: In 23 out of 39 patients, plasma-induced in vitro inhibition of r-αGAL A activity was observed (inhibition-positive). The inhibition titer was strongly negatively correlated to the decrease in lysoGb3: agalsidase-alfa (FElog10(inhibition) = -10.3, P ≤.001), agalsidase-beta (FElog10(inhibition) = -4.7, P ≤.001). Inhibition-positive patients had an accelerated decline in renal function (FE = 1.21, p = .042). During treatment IgG1 anti-r-αGAL A levels increased only in inhibition-positive patients (p = .0045). IgG4 anti-r-αGAL A antibodies developed in 7 out of 9 inhibition-positive patients. Other antibody subclasses were either not present or too low to quantify. CONCLUSION: Development of inhibiting antibodies against r-αGAL A negatively affects the biochemical response to ERT and resulted in an accelerated decline in renal function. The presence of IgG1 and IgG4 anti-r-αGAL A antibodies is associated with in vitro αGAL A activity inhibition.


Subject(s)
Antibodies/classification , Fabry Disease/drug therapy , Isoenzymes/immunology , Recombinant Proteins/immunology , alpha-Galactosidase/immunology , Adolescent , Adult , Antibodies/immunology , Follow-Up Studies , Humans , Immunoglobulin G/immunology , Isoenzymes/therapeutic use , Male , Middle Aged , Recombinant Proteins/therapeutic use , Retrospective Studies , Treatment Outcome , Young Adult , alpha-Galactosidase/therapeutic use
17.
Immunity ; 33(2): 216-28, 2010 Aug 27.
Article in English | MEDLINE | ID: mdl-20727792

ABSTRACT

Natural Killer T (NKT) cells are lipid-reactive, CD1d-restricted T lymphocytes important in infection, cancer, and autoimmunity. In addition to foreign antigens, NKT cells react with endogenous self lipids. However, in the face of stimulating self antigen, it remains unclear how overstimulation of NKT cells is avoided. We hypothesized that constantly degraded endogenous antigen only accumulates upon inhibition of alpha-galactosidase A (alpha-Gal-A) in lysosomes. Here, we show that alpha-Gal-A deficiency caused vigorous activation of NKT cells. Moreover, microbes induced inhibition of alpha-Gal-A activity in antigen-presenting cells. This temporary enzyme block depended on Toll-like receptor (TLR) signaling and ultimately triggered lysosomal lipid accumulation. Thus, we present TLR-dependent negative regulation of alpha-Gal-A as a mechanistic link between pathogen recognition and self lipid antigen induction for NKT cells.


Subject(s)
Autoantigens/immunology , Lipids/immunology , Lysosomes/immunology , Natural Killer T-Cells/immunology , alpha-Galactosidase/immunology , Animals , Antigen Presentation , Cells, Cultured , Coculture Techniques , Dendritic Cells/immunology , Homeostasis , Lymphocyte Activation , Lysosomes/enzymology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/immunology , Myeloid Differentiation Factor 88/metabolism , Natural Killer T-Cells/enzymology , Signal Transduction , Toll-Like Receptors/immunology , Toll-Like Receptors/metabolism , alpha-Galactosidase/metabolism
18.
Xenotransplantation ; 26(5): e12534, 2019 09.
Article in English | MEDLINE | ID: mdl-31342586

ABSTRACT

BACKGROUND: Xenografts are an attractive alternative to traditional bone grafts because of the large supply from donors with predictable morphology and biology as well as minimal risk of human disease transmission. Clinical series involving xenograft bone transplantation, most commonly from bovine sources, have reported poor results with frequent graft rejection and failure to integrate with host tissue. Failures have been attributed to residual alpha-Gal epitope in the xenograft which humans produce natural antibody against. To the authors' knowledge, there is currently no xenograft-derived bone graft substitute that has been adopted by orthopedic surgeons for routine clinical use. METHODS: In the current study, a bone scaffold intended to serve as a bone graft substitute was derived from porcine cancellous bone using a tissue decellularization and chemical oxidation protocol. In vitro cytocompatibility, pathogen clearance, and alpha-Gal quantification tests were used to assess the safety of the bone scaffold intended for human use. RESULTS: In vitro studies showed the scaffold was free of processing chemicals and biocompatible with mouse and human cell lines. When bacterial and viral pathogens were purposefully added to porcine donor tissue, processing successfully removed these pathogens to comply with sterility assurance levels established by allograft tissue providers. Critically, 98.5% of the alpha-Gal epitope was removed from donor tissue after decellularization as shown by ELISA inhibition assay and immunohistochemical staining. CONCLUSIONS: The current investigation supports the biologic safety of bone scaffolds derived from porcine donors using a decellularization protocol that meets current sterility assurance standards. The majority of the highly immunogenic xenograft carbohydrate was removed from donor tissue, and these findings support further in vivo investigation of xenograft-derived bone tissue for orthopedic clinical application.


Subject(s)
Bone Substitutes/metabolism , Heterografts/immunology , Tissue Scaffolds , Transplantation, Heterologous , alpha-Galactosidase/metabolism , Animals , Biomarkers/metabolism , Enzyme-Linked Immunosorbent Assay , Heterografts/metabolism , Heterografts/microbiology , Humans , Immunohistochemistry , Swine , Tissue Scaffolds/microbiology , alpha-Galactosidase/immunology
19.
J Am Soc Nephrol ; 29(12): 2879-2889, 2018 12.
Article in English | MEDLINE | ID: mdl-30385651

ABSTRACT

BACKGROUND: Use of enzyme replacement therapy (ERT) to treat Fabry disease, caused by deficient lysosomal α-galactosidase A activity, can lead to formation of neutralizing antidrug antibodies (ADAs). These antibodies are associated with increased accumulation of plasma globotriaosylceramide (Gb3) and disease progression. Because agalsidase ERT can saturate ADA-binding sites during infusions (achieving agalsidase/antibody equilibrium), we investigated in this open cohort study whether saturated patients (who have excess agalsidase after infusions) experience better clinical outcomes compared with not saturated patients (who have excess ADAs after infusions). METHODS: We isolated ADAs from sera of 26 men with Fabry disease receiving ERT (for a median of 94 months) and determined the amount of agalsidase necessary for antibody saturation. Clinical and biochemical outcomes included measurements of eGFR, interventricular septum thickness, and lyso-Gb3. RESULTS: ADA titers decreased significantly in all patients during infusion. Agalsidase-α and agalsidase-ß had similar ADA-binding capacity and comparable ADA saturation frequency. Fourteen patients with saturated ADAs presented with mild (but significant) loss of eGFR, stable septum thickness, and significantly decreased lyso-Gb3 levels. The 12 not saturated patients had a more pronounced and significant loss of eGFR, increased septum thickness, and a smaller, nonsignificant reduction in lyso-Gb3, over time. In three patients, dose escalation resulted in partially elevated ADA titers, but importantly, also in reduced lyso-Gb3 levels. CONCLUSIONS: A not saturated ADA status during infusion is associated with progressive loss of eGFR and ongoing cardiac hypertrophy. Dose escalation can result in saturation of ADAs and decreasing lyso-Gb3 levels, but may lead to increased ADA titers.


Subject(s)
Antibodies, Neutralizing/blood , Enzyme Replacement Therapy/adverse effects , Fabry Disease/drug therapy , Fabry Disease/immunology , Isoenzymes/administration & dosage , Isoenzymes/adverse effects , alpha-Galactosidase/administration & dosage , alpha-Galactosidase/adverse effects , Adult , Aged , Antigen-Antibody Reactions , Cohort Studies , Dose-Response Relationship, Drug , Humans , Isoenzymes/immunology , Male , Middle Aged , Models, Immunological , Young Adult , alpha-Galactosidase/immunology
20.
Bioconjug Chem ; 29(3): 649-656, 2018 03 21.
Article in English | MEDLINE | ID: mdl-29285931

ABSTRACT

Enzymes are attractive as immunotherapeutics because they can catalyze shifts in the local availability of immunostimulatory and immunosuppressive signals. Clinical success of enzyme immunotherapeutics frequently hinges upon achieving sustained biocatalysis over relevant time scales. The time scale and location of biocatalysis are often dictated by the location of the substrate. For example, therapeutic enzymes that convert substrates distributed systemically are typically designed to have a long half-life in circulation, whereas enzymes that convert substrates localized to a specific tissue or cell population can be more effective when designed to accumulate at the target site. This Topical Review surveys approaches to improve enzyme immunotherapeutic efficacy via chemical modification, encapsulation, and immobilization that increases enzyme accumulation at target sites or extends enzyme half-life in circulation. Examples provided illustrate "replacement therapies" to restore deficient enzyme function, as well as "enhancement therapies" that augment native enzyme function via supraphysiologic doses. Existing FDA-approved enzyme immunotherapies are highlighted, followed by discussion of emerging experimental strategies such as those designed to enhance antitumor immunity or resolve inflammation.


Subject(s)
Enzyme Therapy/methods , Immunotherapy/methods , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/immunology , Anti-Inflammatory Agents/therapeutic use , Antineoplastic Agents/chemistry , Antineoplastic Agents/immunology , Antineoplastic Agents/therapeutic use , Asparaginase/chemistry , Asparaginase/immunology , Asparaginase/therapeutic use , Biocatalysis , Enzymes, Immobilized/chemistry , Enzymes, Immobilized/immunology , Enzymes, Immobilized/therapeutic use , Fabry Disease/immunology , Fabry Disease/therapy , Gaucher Disease/immunology , Gaucher Disease/therapy , Glucosylceramidase/chemistry , Glucosylceramidase/immunology , Glucosylceramidase/therapeutic use , Glycosylation , Humans , Immunoconjugates/chemistry , Immunoconjugates/immunology , Immunoconjugates/therapeutic use , Inflammation/immunology , Inflammation/therapy , Lysosomal Storage Diseases/immunology , Lysosomal Storage Diseases/therapy , Neoplasms/immunology , Neoplasms/therapy , alpha-Galactosidase/chemistry , alpha-Galactosidase/immunology , alpha-Galactosidase/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL