Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 32
1.
Paediatr Drugs ; 26(3): 287-308, 2024 May.
Article En | MEDLINE | ID: mdl-38664313

Inborn errors of metabolism (IEMs) are a group of genetic diseases that occur due to the either deficiency of an enzyme involved in a metabolic/biochemical pathway or other disturbances in the metabolic pathway including transport protein or activator protein deficiencies, cofactor deficiencies, organelle biogenesis, maturation or trafficking problems. These disorders are collectively significant due to their substantial impact on both the well-being and survival of affected individuals. In the quest for effective treatments, enzyme replacement therapy (ERT) has emerged as a viable strategy for patients with many of the lysosomal storage disorders (LSD) and enzyme substitution therapy in the rare form of the other inborn errors of metabolism including phenylketonuria and hypophosphatasia. However, a major challenge associated with enzyme infusion in patients with these disorders, mainly LSD, is the development of high antibody titres. Strategies focusing on immunomodulation have shown promise in inducing immune tolerance to ERT, leading to improved overall survival rates. The implementation of immunomodulation concurrent with ERT administration has also resulted in a decreased occurrence of IgG antibody development compared with cases treated solely with ERT. By incorporating the knowledge gained from current approaches and analysing the outcomes of immune tolerance induction (ITI) modalities from clinical and preclinical trials have demonstrated significant improvement in the efficacy of ERT. In this comprehensive review, the progress in ITI modalities is assessed, drawing insights from both clinical and preclinical trials. The focus is on evaluating the advancements in ITI within the context of IEM, specifically addressing LSDs managed through ERT.


Enzyme Replacement Therapy , Immune Tolerance , Humans , Lysosomal Storage Diseases/drug therapy , Lysosomal Storage Diseases/immunology , Lysosomal Storage Diseases/therapy , Metabolism, Inborn Errors/immunology , Metabolism, Inborn Errors/drug therapy , Metabolism, Inborn Errors/therapy , Animals
2.
Int J Mol Sci ; 23(4)2022 Feb 11.
Article En | MEDLINE | ID: mdl-35216110

Mucopolysaccharidoses are rare paediatric lysosomal storage disorders, characterised by accumulation of glycosaminoglycans within lysosomes. This is caused by deficiencies in lysosomal enzymes involved in degradation of these molecules. Dependent on disease, progressive build-up of sugars may lead to musculoskeletal abnormalities and multi-organ failure, and in others, to cognitive decline, which is still a challenge for current therapies. The worsening of neuropathology, observed in patients following recovery from flu-like infections, suggests that inflammation is highly implicated in disease progression. This review provides an overview of the pathological features associated with the mucopolysaccharidoses and summarises current knowledge regarding the inflammatory responses observed in the central nervous system and periphery. We propose a model whereby progressive accumulation of glycosaminoglycans elicits an innate immune response, initiated by the Toll-like receptor 4 pathway, but also precipitated by secondary storage components. Its activation induces cells of the immune system to release pro-inflammatory cytokines, such as TNF-α and IL-1, which induce progression through chronic neuroinflammation. While TNF-α is mostly associated with bone and joint disease in mucopolysaccharidoses, increasing evidence implicates IL-1 as a main effector of innate immunity in the central nervous system. The (NOD)-like receptor protein 3 inflammasome is therefore implicated in chronic neuroinflammation and should be investigated further to identify novel anti-inflammatory treatments.


Immunity, Innate/immunology , Lysosomal Storage Diseases/immunology , Mucopolysaccharidoses/immunology , Animals , Cytokines/immunology , Humans , Inflammation/immunology , Joint Diseases/immunology , Lysosomes/immunology
3.
Int J Mol Sci ; 22(23)2021 Nov 28.
Article En | MEDLINE | ID: mdl-34884674

Acid sphingomyelinase deficiency (ASMD) is a lysosomal storage disease caused by deficient activity of acid sphingomyelinase (ASM) enzyme, leading to the accumulation of varying degrees of sphingomyelin. Lipid storage leads to foam cell infiltration in tissues, and clinical features including hepatosplenomegaly, pulmonary insufficiency and in some cases central nervous system involvement. ASM enzyme replacement therapy is currently in clinical trial being the first treatment addressing the underlying pathology of the disease. Therefore, presently, it is critical to better comprehend ASMD to improve its diagnose and monitoring. Lung disease, including recurrent pulmonary infections, are common in ASMD patients. Along with lung disease, several immune system alterations have been described both in patients and in ASMD animal models, thus highlighting the role of ASM enzyme in the immune system. In this review, we summarized the pivotal roles of ASM in several immune system cells namely on macrophages, Natural Killer (NK) cells, NKT cells, B cells and T cells. In addition, an overview of diagnose, monitoring and treatment of ASMD is provided highlighting the new enzyme replacement therapy available.


Lysosomal Storage Diseases/immunology , Sphingomyelin Phosphodiesterase/deficiency , Animals , Enzyme Replacement Therapy , Humans , Lung Diseases/enzymology , Lysosomal Storage Diseases/diagnosis , Lysosomal Storage Diseases/genetics , Lysosomal Storage Diseases/therapy , Sphingomyelin Phosphodiesterase/genetics , Sphingomyelin Phosphodiesterase/immunology
4.
Essays Biochem ; 64(3): 565-578, 2020 09 23.
Article En | MEDLINE | ID: mdl-32808655

Lyso-glycosphingolipids are generated in excess in glycosphingolipid storage disorders. In the course of these pathologies glycosylated sphingolipid species accumulate within lysosomes due to flaws in the respective lipid degrading machinery. Deacylation of accumulating glycosphingolipids drives the formation of lyso-glycosphingolipids. In lysosomal storage diseases such as Gaucher Disease, Fabry Disease, Krabbe disease, GM1 -and GM2 gangliosidosis, Niemann Pick type C and Metachromatic leukodystrophy massive intra-lysosomal glycosphingolipid accumulation occurs. The lysosomal enzyme acid ceramidase generates the deacylated lyso-glycosphingolipid species. This review discusses how the various lyso-glycosphingolipids are synthesized, how they may contribute to abnormal immunity in glycosphingolipid storing lysosomal diseases and what therapeutic opportunities exist.


Enzyme Replacement Therapy/methods , Genetic Therapy/methods , Glycosphingolipids/biosynthesis , Lysosomal Storage Diseases/drug therapy , Lysosomal Storage Diseases/metabolism , Lysosomes/metabolism , Molecular Targeted Therapy/methods , Acid Ceramidase/metabolism , Animals , Humans , Immunity , Lysosomal Storage Diseases/immunology
5.
Neuropathology ; 40(5): 492-495, 2020 Oct.
Article En | MEDLINE | ID: mdl-32424839

Scleromyxedema (SME) is characterized by widespread waxy papules on the skin, with mucin deposits in the upper dermis. Twenty-one SME cases of myopathy have been reported; of the cases, six showed vacuolar formation, and two showed mucin deposition. We report the first case of SME with mucin-associated vacuolated fibers. A 45-year-old woman with SME developed progressive proximal muscle weakness. Muscle biopsy revealed myopathic changes with numerous vacuoles linked to mucin in the affected muscle fibers, which were heavily immunostained for fibroblast growth factor 2 (FGF2). Despite repeated high dose oral prednisolone and intravenous immunoglobulin administrations, muscle weakness recurred continuingly, culminating in death due to congestive heart failure. Immunotherapy was partly effective in our case, although it was refractory. Treatment responsiveness in patients with SME myopathy varied; however, due to its rarity, the mechanism remains to be elucidated. To address this issue, we investigated muscle specimens immunohistochemically and detected marked upregulation of FGF2 in the affected muscle fibers of our patient. FGF2, a strong myogenesis inhibitor, may exert a suppressive effect on muscle fiber regeneration, which may have conferred refractoriness to our patient's SME myopathy.


Fibroblast Growth Factor 2/metabolism , Immunotherapy , Lysosomal Storage Diseases/metabolism , Lysosomal Storage Diseases/therapy , Mucins/metabolism , Muscular Diseases/metabolism , Muscular Diseases/therapy , Scleromyxedema/metabolism , Scleromyxedema/therapy , Female , Humans , Lysosomal Storage Diseases/immunology , Lysosomal Storage Diseases/pathology , Middle Aged , Muscular Diseases/immunology , Muscular Diseases/pathology , Scleromyxedema/immunology , Scleromyxedema/pathology
6.
Orphanet J Rare Dis ; 13(1): 127, 2018 07 31.
Article En | MEDLINE | ID: mdl-30064518

BACKGROUND: Fabry disease (OMIM #301500) is an X-linked disorder caused by alpha-galactosidase A deficiency with two major clinical phenotypes: classic and non-classic of different prognosis. From 2001, enzyme replacement therapies (ERT) have been available. We aimed to determine the epidemiology and the functional characteristics of anti-drug antibodies. Patients from the French multicenter cohort FFABRY (n = 103 patients, 53 males) were prospectively screened for total anti-agalsidase IgG and IgG subclasses with a home-made enzyme-linked immunosorbent assay (ELISA), enzyme-inhibition assessed with neutralization assays and lysoGb3 plasma levels, and compared for clinical outcomes. RESULTS: Among the patients exposed to agalsidase, 40% of men (n = 18/45) and 8% of women (n = 2/25) had antibodies with a complete cross-reactivity towards both ERTs. Antibodies developed preferentially in men with non-missense GLA mutations (relative risk 2.88, p = 0.006) and classic phenotype (58.6% (17/29) vs 6.7% (1/16), p = 0.0005). Specific anti-agalsidase IgG1 were the most frequently observed (16/18 men), but the highest concentrations were observed for IgG4 (median 1.89 µg/ml, interquartile range (IQR) [0.41-12.24]). In the men exposed to agalsidase, inhibition was correlated with the total IgG titer (r = 0.67, p < 0.0001), especially IgG4 (r = 0.75, p = 0.0005) and IgG2 (r = 0.72, p = 0.001). Inhibition was confirmed intracellularly in Fabry patient leucocytes cultured with IgG-positive versus negative serum (median: 42.0 vs 75.6%, p = 0.04), which was correlated with IgG2 (r = 0.67, p = 0.017, n = 12) and IgG4 levels (r = 0.59, p = 0.041, n = 12). Plasma LysoGb3 levels were correlated with total IgG (r = 0.66, p = 0.001), IgG2 (r = 0.72, p = 0.004), IgG4 (r = 0.58, p = 0.03) and IgG1 (r = 0.55, p = 0.04) titers. Within the classic group, no clinical difference was observed but lysoGb3 levels were higher in antibody-positive patients (median 33.2 ng/ml [IQR 20.6-55.6] vs 12.5 [10.1-24.0], p = 0.005). CONCLUSION: Anti-agalsidase antibodies preferentially develop in the severe classic Fabry phenotype. They are frequently associated with enzyme inhibition and higher lysoGb3 levels. As such, they could be considered as a hallmark of severity associated with the classic phenotype. The distinction of the clinical phenotypes should now be mandatory in studies dealing with Fabry disease and its current and future therapies.


Antibodies/blood , Fabry Disease/immunology , Fabry Disease/pathology , alpha-Galactosidase/antagonists & inhibitors , Adult , Enzyme Replacement Therapy , Enzyme-Linked Immunosorbent Assay , Fabry Disease/blood , Fabry Disease/drug therapy , Female , Glycolipids/blood , Humans , Immunoglobulin G/blood , Lysosomal Storage Diseases/blood , Lysosomal Storage Diseases/drug therapy , Lysosomal Storage Diseases/immunology , Lysosomal Storage Diseases/pathology , Male , Middle Aged , Prospective Studies , Sphingolipids/blood , alpha-Galactosidase/immunology
7.
Semin Immunol ; 37: 30-42, 2018 06.
Article En | MEDLINE | ID: mdl-29478824

The complement system is well appreciated for its role as an important effector of innate immunity that is activated by the classical, lectin or alternative pathway. C5a is one important mediator of the system that is generated in response to canonical and non-canonical C5 cleavage by circulating or cell-derived proteases. In addition to its function as a chemoattractant for neutrophils and other myeloid effectors, C5a and its sister molecule C3a have concerted roles in cell homeostasis and surveillance. Through activation of their cognate G protein coupled receptors, C3a and C5a regulate multiple intracellular pathways within the mitochondria and the lysosomal compartments that harbor multiple enzymes critical for protein, carbohydrate and lipid metabolism. Genetic mutations of such lysosomal enzymes or their receptors can result in the compartmental accumulation of specific classes of substrates in this organelle summarized as lysosomal storage diseases (LSD). A frequent LSD is Gaucher disease (GD), caused by autosomal recessively inherited mutations in GBA1, resulting in functional defects of the encoded enzyme, acid ß-glucosidase (glucocerebrosidase, GCase). Such mutations promote excessive accumulation of ß-glucosylceramide (GC or GL1) in innate and adaptive immune cells frequently associated with chronic inflammation. Recently, we uncovered an unexpected link between the C5a and C5a receptor 1 (C5aR1) axis and the accumulation of GL1 in experimental and clinical GD. Here, we will review the pathways of complement activation in GD, its role as a mediator of the inflammatory response, and its impact on glucosphingolipid metabolism. Further, we will discuss the potential role of the C5a/C5aR1 axis in GL1-specific autoantibody formation and as a novel therapeutic target in GD.


Complement C5a/metabolism , Gaucher Disease/immunology , Glucosylceramidase/genetics , Inflammation/immunology , Lysosomal Storage Diseases/immunology , Animals , Autoantibodies/metabolism , Gaucher Disease/genetics , Glucosylceramides/metabolism , Humans , Receptor, Anaphylatoxin C5a/metabolism
8.
Bioconjug Chem ; 29(3): 649-656, 2018 03 21.
Article En | MEDLINE | ID: mdl-29285931

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.


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
9.
J Inherit Metab Dis ; 39(4): 499-512, 2016 07.
Article En | MEDLINE | ID: mdl-26883220

In the light of clinical experience in infantile onset Pompe patients, the immunological impact on the tolerability and long-term efficacy of enzyme replacement therapy (ERT) for lysosomal storage disorders has come under renewed scrutiny. This article details the currently proposed immunological mechanisms involved in the development of anti-drug antibodies and the current therapies used in their treatment. Given the current understanding of the adaptive immune response, it focuses particularly on T cell dependent mechanisms and the paradigm of using lymphocytic negative selection as a predictor of antibody formation. This concept originally postulated in the 1970s, stipulated that the genotypically determined lack of production or production of a variant protein determines an individual's lymphocytic repertoire. This in turn is the key factor in determining the potential severity of an individual's immunological response to ERT. It also highlights the need for immunological assay standardization particularly those looking at describing the degree of functional impact, robust biochemical or clinical endpoints and detailed patient subgroup identification if the true evaluations of impact are to be realised.


Enzyme Replacement Therapy , Immune System/physiology , Lysosomal Storage Diseases/drug therapy , Lysosomal Storage Diseases/immunology , Antibody Formation , Enzyme Replacement Therapy/adverse effects , Enzyme Therapy , Enzymes/immunology , Humans , Treatment Outcome , alpha-Glucosidases/immunology , alpha-Glucosidases/therapeutic use
10.
Mol Genet Metab ; 117(2): 66-83, 2016 Feb.
Article En | MEDLINE | ID: mdl-26597321

The US Food and Drug Administration (FDA) and National Organization for Rare Disease (NORD) convened a public workshop titled "Immune Responses to Enzyme Replacement Therapies: Role of Immune Tolerance Induction" to discuss the impact of anti-drug antibodies (ADAs) on efficacy and safety of enzyme replacement therapies (ERTs) intended to treat patients with lysosomal storage diseases (LSDs). Participants in the workshop included FDA staff, clinicians, scientists, patients, industry, and advocacy group representatives. The risks and benefits of implementing prophylactic immune tolerance induction (ITI) to reduce the potential clinical impact of antibody development were considered. Complications due to immune responses to ERT are being recognized with increasing experience and lengths of exposure to ERTs to treat several LSDs. Strategies to mitigate immune responses and to optimize therapies are needed. Discussions during the workshop resulted in the identification of knowledge gaps and future areas of research, as well as the following proposals from the participants: (1) systematic collection of longitudinal data on immunogenicity to better understand the impact of ADAs on long-term clinical outcomes; (2) development of disease-specific biomarkers and outcome measures to assess the effect of ADAs and ITI on efficacy and safety; (3) development of consistent approaches to ADA assays to allow comparisons of immunogenicity data across different products and disease groups, and to expedite reporting of results; (4) establishment of a system to widely share data on antibody titers following treatment with ERTs; (5) identification of components of the protein that are immunogenic so that triggers and components of the immune responses can be targeted in ITI; and (6) consideration of early ITI in patients who are at risk of developing clinically relevant ADA that have been demonstrated to worsen treatment outcomes.


Hydrolases/therapeutic use , Lysosomal Storage Diseases/drug therapy , Animals , Enzyme Replacement Therapy , Humans , Hydrolases/immunology , Immune Tolerance , Lysosomal Storage Diseases/immunology , Recombinant Proteins/immunology , Recombinant Proteins/therapeutic use
11.
Cell Metab ; 22(3): 485-98, 2015 Sep 01.
Article En | MEDLINE | ID: mdl-26299452

The endolysosomal system is critical for the maintenance of cellular homeostasis. However, how endolysosomal compartment is regulated by mitochondrial function is largely unknown. We have generated a mouse model with defective mitochondrial function in CD4(+) T lymphocytes by genetic deletion of the mitochondrial transcription factor A (Tfam). Mitochondrial respiration deficiency impairs lysosome function, promotes p62 and sphingomyelin accumulation, and disrupts endolysosomal trafficking pathways and autophagy, thus linking a primary mitochondrial dysfunction to a lysosomal storage disorder. The impaired lysosome function in Tfam-deficient cells subverts T cell differentiation toward proinflammatory subsets and exacerbates the in vivo inflammatory response. Restoration of NAD(+) levels improves lysosome function and corrects the inflammatory defects in Tfam-deficient T cells. Our results uncover a mechanism by which mitochondria regulate lysosome function to preserve T cell differentiation and effector functions, and identify strategies for intervention in mitochondrial-related diseases.


DNA-Binding Proteins/immunology , Lysosomal Storage Diseases/immunology , Lysosomes/immunology , Mitochondria/immunology , Mitochondrial Proteins/immunology , Sphingolipidoses/immunology , T-Lymphocytes/immunology , Transcription Factors/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology , Cell Respiration , DNA-Binding Proteins/genetics , Gene Deletion , Immunity, Cellular , Lysosomal Storage Diseases/genetics , Lysosomal Storage Diseases/pathology , Lysosomes/genetics , Lysosomes/pathology , Mice , Mitochondria/genetics , Mitochondria/pathology , Mitochondrial Proteins/genetics , Sphingolipidoses/genetics , Sphingolipidoses/pathology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Transcription Factors/genetics
12.
Best Pract Res Clin Endocrinol Metab ; 29(2): 183-94, 2015 Mar.
Article En | MEDLINE | ID: mdl-25987172

Substitution of the defective lysosomal enzyme in lysosomal storage disorders (LSDs) often elicits antibody formation towards the infused protein. Aside from Gaucher disease, antibodies often lead to infusion associated reactions and a reduced biochemical response. In Pompe disease, antibody titer is predictive of clinical outcome, but this is less apparent in other LSDs and warrants further study. Few laboratories are capable of enzyme-antibody determination: often physicians need to rely on the enzyme manufacturer for analysis. Currently, laboratories employ different antibody assays which hamper comparisons between cohorts or treatment regimens. Assay standardisation, including measurement of antibody-related enzyme inhibition, is therefore urgently needed. Successful immunomodulation has been reported in Pompe and in Gaucher disease, with variable success. Immunomodulation regimens that contain temporary depletion of B-cells (anti-CD20) are most used. Bone marrow transplantation in MPS-I results in disappearance of antibodies. No other clinical studies have been conducted in humans with immunomodulation in other LSDs.


Enzyme Replacement Therapy , Enzyme Therapy , Isoantibodies/immunology , Lysosomal Storage Diseases/drug therapy , Antibodies/immunology , Enzymes/immunology , Fabry Disease/drug therapy , Fabry Disease/immunology , Gaucher Disease/drug therapy , Gaucher Disease/immunology , Glucan 1,4-alpha-Glucosidase/immunology , Glucan 1,4-alpha-Glucosidase/therapeutic use , Glucosylceramidase/immunology , Glucosylceramidase/therapeutic use , Glycogen Storage Disease Type II/drug therapy , Glycogen Storage Disease Type II/immunology , Humans , Lysosomal Storage Diseases/immunology , alpha-Galactosidase/immunology , alpha-Galactosidase/therapeutic use
13.
Biol Chem ; 396(6-7): 659-67, 2015 Jun.
Article En | MEDLINE | ID: mdl-25720063

Lysosomal storage diseases (LSDs) are mainly caused by the defective activity of lysosomal hydrolases. A sub-class of LSDs are the sphingolipidoses, in which sphingolipids accumulate intra-cellularly. We here discuss the role of innate immunity in the sphingolipidoses, and compare the pathways of activation in two classical sphingolipidoses, namely Gaucher disease and Sandhoff disease, and in Niemann-Pick C disease, in which the main storage material is cholesterol but sphingolipids also accumulate. We discuss the mechanisms leading to neuroinflammation, and the different pathways of neuroinflammation in the different diseases, and suggest that intervention in these pathways may be a useful therapeutic approach to address these devastating human diseases.


Brain/immunology , Immunity, Innate/immunology , Lysosomal Storage Diseases/immunology , Sphingolipidoses/immunology , Animals , Gaucher Disease/immunology , Humans , Niemann-Pick Disease, Type C/immunology
14.
An Pediatr (Barc) ; 76(2): 92-7, 2012 Feb.
Article Es | MEDLINE | ID: mdl-22032885

INTRODUCTION: Haemophagocytic syndrome (HS) is a common manifestation of several congenital disorders characterised by a disruption of lysosomal secretion, interrupting the cytolytic pathway and triggering a dysfunction in the immune synapse. In this situation, the recognition of certain extra-immunological manifestations may help in the diagnostic process. PATIENTS AND METHODS: We describe the clinical and biological features present in two brothers with familial haemophagocytic lymphohistiocytosis type 3 (FHL-3), two patients with Griscelli syndrome type 2 (GS-2) and one patient with Chédiak-Higashi syndrome (CHS). RESULTS: Mutational assays at UNC13D were carried out on two brothers after diagnosing an early onset HS in the first one, yielding a positive result in both cases with a consequent diagnosis of FHL-3. The diagnosis of GS-2 was supported by positive results of mutational Rab27A studies in one patient with HS and abnormal pigmentation, and in her cousin who was affected by a similar abnormal pigmentation. The diagnosis of CHS was established in one patient with HS, abnormal pigmentation and atypical granules on cytological examination of a bone marrow smear. Diagnosis was confirmed in this patient by the finding of a homozygous LYST mutation. CONCLUSIONS: We point out the importance of recognising the presence of typical extra-immunological manifestations of certain congenital disorders of lysosome secretion in patients diagnosed with HS. The association of albinism and immunodeficiency has played a critical role in the recent identification of the molecular mechanism involved in these disorders.


Chediak-Higashi Syndrome , Immunologic Deficiency Syndromes , Lymphohistiocytosis, Hemophagocytic , Lysosomal Storage Diseases , Piebaldism , Antigen-Presenting Cells , Chediak-Higashi Syndrome/diagnosis , Chediak-Higashi Syndrome/genetics , Chediak-Higashi Syndrome/immunology , Child , Child, Preschool , Humans , Immunologic Deficiency Syndromes/diagnosis , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/immunology , Infant , Lymphocytes , Lymphohistiocytosis, Hemophagocytic/diagnosis , Lymphohistiocytosis, Hemophagocytic/genetics , Lymphohistiocytosis, Hemophagocytic/immunology , Lysosomal Storage Diseases/diagnosis , Lysosomal Storage Diseases/genetics , Lysosomal Storage Diseases/immunology , Piebaldism/diagnosis , Piebaldism/genetics , Piebaldism/immunology , Primary Immunodeficiency Diseases
15.
Antioxid Redox Signal ; 14(11): 2233-43, 2011 Jun.
Article En | MEDLINE | ID: mdl-20812858

Autophagy is a basic cell biological process ongoing under physiologic circumstances in almost all cell types of the human organism and upregulated by various stress conditions including those leading to inflammation. Since autophagy affects the effector cells of innate and adaptive immunity mediating the inflammatory response, its activity in these cells influences the antimicrobial response, the development of an effective cognate immune defense, and the course of the normal sterile inflammatory reactions. The level of autophagic activity may determine whether tissue cells die by apoptosis, necrosis, or through autophagy, and, as a consequence, whether the clearance of these dying cells is a silent process or results in an inflammatory response. Loss or decreased autophagy may lead to necrotic death that can initiate an inflammatory reaction in phagocytes through their surface and cytosolic receptors. Engulfment of certain cells dying through autophagy can activate the inflammasome. The intertwining regulatory connections between inflammation and immunity extend to pathologic conditions including chronic inflammatory diseases, autoimmunity and cancer.


Autophagy , Inflammation/metabolism , Adaptive Immunity , Animals , Apoptosis , Crohn Disease/immunology , Crohn Disease/metabolism , Crohn Disease/pathology , Humans , Immunity, Innate , Inflammation/immunology , Inflammation/pathology , Lysosomal Storage Diseases/immunology , Lysosomal Storage Diseases/metabolism , Lysosomal Storage Diseases/pathology , Necrosis , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/pathology , Neurodegenerative Diseases/immunology , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology
16.
Mol Ther ; 18(11): 1983-94, 2010 Nov.
Article En | MEDLINE | ID: mdl-20736932

Liver-directed gene therapy with adeno-associated virus (AAV) vectors effectively treats mouse models of lysosomal storage diseases (LSDs). We asked whether these results were likely to translate to patients. To understand to what extent preexisting anti-AAV8 antibodies could impede AAV8-mediated liver transduction in primates, commonly preexposed to AAV, we quantified the effects of preexisting antibodies on liver transduction and subsequent transgene expression in mouse and nonhuman primate (NHP) models. Using the highest viral dose previously reported in a clinical trial, passive transfer of NHP sera containing relatively low anti-AAV8 titers into mice blocked liver transduction, which could be partially overcome by increasing vector dose tenfold. Based on this and a survey of anti-AAV8 titers in 112 humans, we predict that high-dose systemic gene therapy would successfully transduce liver in >50% of human patients. However, although high-dose AAV8 administration to mice and monkeys with equivalent anti-AAV8 titers led to comparable liver vector copy numbers, the resulting transgene expression in primates was ~1.5-logs lower than mice. This suggests vector fate differs in these species and that strategies focused solely on overcoming preexisting vector-specific antibodies may be insufficient to achieve clinically meaningful expression levels of LSD genes using a liver-directed gene therapy approach in patients.


Dependovirus/genetics , Genetic Therapy , Hepatocytes/immunology , Lysosomal Storage Diseases/therapy , Transgenes/physiology , alpha-Galactosidase/blood , Animals , Antibodies, Neutralizing/immunology , Blotting, Western , Genetic Vectors/administration & dosage , HeLa Cells , Hepatocytes/metabolism , Humans , Lysosomal Storage Diseases/genetics , Lysosomal Storage Diseases/immunology , Macaca fascicularis , Macaca mulatta , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Plasmapheresis , Protein Biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , alpha-Galactosidase/genetics
18.
Int J Clin Pharmacol Ther ; 47 Suppl 1: S86-99, 2009.
Article En | MEDLINE | ID: mdl-20040318

In this article, the formation of antibodies during enzyme replacement therapy (ERT) for lysosomal storage diseases (LSDs) is reviewed in the light of present-day immunological concepts of immunogenicity and tolerance. Except in Gaucher disease, anti-enzyme antibodies frequently form (mainly immunoglobulin G) in patients receiving ERT, though they tend to wane as treatment continues. If the therapeutic enzyme is inhibited by antibodies, no significant modification to treatment is normally warranted, in clear contrast to therapy of hemophilia with clotting factors. The main adverse consequences of ERT, observed in only some patients, are sporadic hypersensitivity reactions, which are likely to be humorally mediated. Some infusion-related reactions are probably due to antibodies. In order to minimize immunogenicity, infused enzymes should be deaggregated and administered at low doses. In addition, inadvertent exposure to co-stimuli that might activate antigen-specific T or B lymphocytes should be avoided. The presence of cross-reacting immunological material, such as in patients with low levels or missense mutations of a gene coding for a lysosomal enzyme, tends to correlate with immune tolerance to the administered enzyme. There is a need for reliable biomarkers for therapeutic efficacy: some directions for further exploration are suggested. In animal models of LSDs, gene therapy delivered via viral vectors can rectify the lysosomal defect, and regulatory T cells that suppress antibody formation can be induced. This is a promising strategy that warrants further investigation in patients with LSDs.


B-Lymphocytes/immunology , Enzyme Replacement Therapy/adverse effects , Immune System/immunology , Lysosomal Storage Diseases/immunology , Animals , Antibodies/adverse effects , Antigen-Antibody Reactions , Biomarkers/metabolism , Enzyme Replacement Therapy/methods , Genetic Therapy/methods , Humans , Hypersensitivity, Immediate/immunology , Immune Tolerance/genetics , Lysosomal Storage Diseases/drug therapy , Models, Molecular
20.
Curr Gene Ther ; 9(6): 503-10, 2009 Dec.
Article En | MEDLINE | ID: mdl-19807648

Significant advances in therapy for lysosomal storage disorders have occurred with an accelerating pace over the past decade. Although enzyme replacement therapy has improved the outcome of lysosomal storage disorders, antibody responses have occurred and sometimes prevented efficacy, especially in cross-reacting immune material negative patients with Pompe disease. Preclinical gene therapy experiments have revealed the relevance of immune responses to long-term efficacy. The choice of regulatory cassette played a critical role in evading humoral and cellular immune responses to gene therapy in knockout mouse models, at least in adult animals. Liver-specific regulatory cassettes prevented antibody formation and enhanced the efficacy of gene therapy. Regulatory T cells prevented transgene directed immune responses, as shown by adoptive transfer of antigen-specific immune tolerance to enzyme therapy. Immunomodulatory gene therapy with a very low vector dose could enhance the efficacy of enzyme therapy in Pompe disease and other lysosomal storage disorders.


Genetic Therapy , Lysosomal Storage Diseases/therapy , Antigens, CD/immunology , Humans , Lysosomal Storage Diseases/genetics , Lysosomal Storage Diseases/immunology , T-Lymphocytes, Regulatory/immunology , Transgenes
...