Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Blood ; 143(15): 1476-1487, 2024 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-38194689

RESUMEN

ABSTRACT: Mutations in the small Rho-family guanosine triphosphate hydrolase RAC2, critical for actin cytoskeleton remodeling and intracellular signal transduction, are associated with neonatal severe combined immunodeficiency (SCID), infantile neutrophilic disorder resembling leukocyte adhesion deficiency (LAD), and later-onset combined immune deficiency (CID). We investigated 54 patients (23 previously reported) from 37 families yielding 15 novel RAC2 missense mutations, including one present only in homozygosity. Data were collected from referring physicians and literature reports with updated clinical information. Patients were grouped by presentation: neonatal SCID (n = 5), infantile LAD-like disease (n = 5), or CID (n = 44). Disease correlated to RAC2 activity: constitutively active RAS-like mutations caused neonatal SCID, dominant-negative mutations caused LAD-like disease, whereas dominant-activating mutations caused CID. Significant T- and B-lymphopenia with low immunoglobulins were seen in most patients; myeloid abnormalities included neutropenia, altered oxidative burst, impaired neutrophil migration, and visible neutrophil macropinosomes. Among 42 patients with CID with clinical data, upper and lower respiratory infections and viral infections were common. Twenty-three distinct RAC2 mutations, including 15 novel variants, were identified. Using heterologous expression systems, we assessed downstream effector functions including superoxide production, p21-activated kinase 1 binding, AKT activation, and protein stability. Confocal microscopy showed altered actin assembly evidenced by membrane ruffling and macropinosomes. Altered protein localization and aggregation were observed. All tested RAC2 mutant proteins exhibited aberrant function; no single assay was sufficient to determine functional consequence. Most mutants produced elevated superoxide; mutations unable to support superoxide formation were associated with bacterial infections. RAC2 mutations cause a spectrum of immune dysfunction, ranging from early onset SCID to later-onset combined immunodeficiencies depending on RAC2 activity. This trial was registered at www.clinicaltrials.gov as #NCT00001355 and #NCT00001467.


Asunto(s)
Síndromes de Inmunodeficiencia , Síndrome de Deficiencia de Adhesión del Leucocito , Enfermedades de Inmunodeficiencia Primaria , Inmunodeficiencia Combinada Grave , Humanos , Recién Nacido , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/metabolismo , Neutrófilos/metabolismo , Enfermedades de Inmunodeficiencia Primaria/genética , Enfermedades de Inmunodeficiencia Primaria/metabolismo , Proteínas de Unión al GTP rac/genética , Proteínas de Unión al GTP rac/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Proteína RCA2 de Unión a GTP , Inmunodeficiencia Combinada Grave/genética , Inmunodeficiencia Combinada Grave/metabolismo , Superóxidos/metabolismo
2.
Hum Mol Genet ; 32(9): 1439-1456, 2023 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-36458887

RESUMEN

Immunodeficiency, centromeric instability and facial anomalies (ICF) syndrome is in most cases caused by mutations in either DNA methyltransferase (DNMT)3B, zinc finger and BTB domain containing 24, cell division cycle associated 7 or helicase lymphoid-specific. However, the causative genes of a few ICF patients remain unknown. We, herein, identified ubiquitin-like with plant homeodomain and really interesting new gene finger domains 1 (UHRF1) as a novel causative gene of one such patient with atypical symptoms. This patient is a compound heterozygote for two previously unreported mutations in UHRF1: c.886C > T (p.R296W) and c.1852C > T (p.R618X). The R618X mutation plausibly caused nonsense-mediated decay, while the R296W mutation changed the higher order structure of UHRF1, which is indispensable for the maintenance of CG methylation along with DNMT1. Genome-wide methylation analysis revealed that the patient had a centromeric/pericentromeric hypomethylation, which is the main ICF signature, but also had a distinctive hypomethylation pattern compared to patients with the other ICF syndrome subtypes. Structural and biochemical analyses revealed that the R296W mutation disrupted the protein conformation and strengthened the binding affinity of UHRF1 with its partner LIG1 and reduced ubiquitylation activity of UHRF1 towards its ubiquitylation substrates, histone H3 and proliferating cell nuclear antigen -associated factor 15 (PAF15). We confirmed that the R296W mutation causes hypomethylation at pericentromeric repeats by generating the HEK293 cell lines that mimic the patient's UHRF1 molecular context. Since proper interactions of the UHRF1 with LIG1, PAF15 and histone H3 are essential for the maintenance of CG methylation, the mutation could disturb the maintenance process. Evidence for the importance of the UHRF1 conformation for CG methylation in humans is, herein, provided for the first time and deepens our understanding of its role in regulation of CG methylation.


Asunto(s)
Histonas , Enfermedades de Inmunodeficiencia Primaria , Humanos , Proteínas Potenciadoras de Unión a CCAAT/genética , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , ADN/genética , ADN/metabolismo , Metilación de ADN/genética , Metilación de ADN/fisiología , Células HEK293 , Histonas/genética , Histonas/metabolismo , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/metabolismo , Mutación , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Inestabilidad Cromosómica/genética , Inestabilidad Cromosómica/fisiología , Centrómero/genética , Centrómero/metabolismo , Enfermedades de Inmunodeficiencia Primaria/genética , Enfermedades de Inmunodeficiencia Primaria/metabolismo , Cara/anomalías , Genoma Humano/genética , Genoma Humano/fisiología
3.
Blood Adv ; 7(8): 1531-1535, 2023 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-36206192

RESUMEN

Most hereditary forms of hemophagocytic lymphohistiocytosis (HLH) are caused by defects of cytotoxicity, including the vesicle trafficking disorder Griscelli syndrome type 2 (GS2, RAB27A deficiency). Deficiency of the mitogen-activated protein kinase activating death domain protein (MADD) results in a protean syndrome with neurological and endocrinological involvement. MADD acts as a guanine nucleotide exchange factor for small guanosine triphosphatases, including RAB27A. A homozygous splice site mutation in MADD was identified in a female infant with syndromic features, secretory diarrhea, and features of HLH. Aberrant splicing caused by this mutation leads to an in-frame deletion of 30 base pairs and favors other aberrant variants. Patient natural killer (NK) cells and cytotoxic T cells showed a severe degranulation defect leading to absent perforin-mediated cytotoxicity. Platelets displayed defective adenosine triphosphate secretion, similar to that in GS2. To prove causality, we introduced a CRISPR/Cas9-based MADD knockout in the NK cell line NK-92mi. MADD-deficient NK-92mi cells showed a degranulation defect and impaired cytotoxicity similar to that of the patient. The defect of cytotoxicity was confirmed in another patient with MADD deficiency. In conclusion, RAB27A-interacting MADD is involved in vesicle release by cytotoxic cells and platelets. MADD deficiency causes a degranulation defect and represents a novel disease predisposing to an HLH phenotype.


Asunto(s)
Citotoxicidad Inmunológica , Enfermedades de Inmunodeficiencia Primaria , Femenino , Humanos , Dominio de Muerte , Células Asesinas Naturales/metabolismo , Linfocitos T Citotóxicos/metabolismo , Enfermedades de Inmunodeficiencia Primaria/metabolismo
4.
Proc Natl Acad Sci U S A ; 119(21): e2119483119, 2022 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-35588454

RESUMEN

Chemokine receptor nanoscale organization at the cell membrane is orchestrated by the actin cytoskeleton and influences cell responses. Using single-particle tracking analysis we show that CXCR4R334X, a truncated mutant chemokine receptor linked to WHIM syndrome (warts, hypogammaglobulinemia, infections, myelokathexis), fails to nanoclusterize after CXCL12 stimulation, and alters the lateral mobility and spatial organization of CXCR4 when coexpressed. These findings correlate with multiple phalloidin-positive protrusions in cells expressing CXCR4R334X, and their inability to correctly sense chemokine gradients. The underlying mechanisms involve inappropriate actin cytoskeleton remodeling due to the inadequate ß-arrestin1 activation by CXCR4R334X, which disrupts the equilibrium between activated and deactivated cofilin. Overall, we provide insights into the molecular mechanisms governing CXCR4 nanoclustering, signaling and cell function, and highlight the essential scaffold role of ß-arrestin1 to support CXCL12-mediated actin reorganization and receptor clustering. These defects associated with CXCR4R334X expression might contribute to the severe immunological symptoms associated with WHIM syndrome.


Asunto(s)
Enfermedades de Inmunodeficiencia Primaria , Receptores CXCR4 , Verrugas , Factores Despolimerizantes de la Actina/metabolismo , Membrana Celular/metabolismo , Movimiento Celular , Humanos , Mutación , Enfermedades de Inmunodeficiencia Primaria/genética , Enfermedades de Inmunodeficiencia Primaria/metabolismo , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Imagen Individual de Molécula , Verrugas/genética , Verrugas/metabolismo
5.
Nat Commun ; 13(1): 649, 2022 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-35115551

RESUMEN

RMRP encodes a non-coding RNA forming the core of the RNase MRP ribonucleoprotein complex. Mutations cause Cartilage Hair Hypoplasia (CHH), characterized by skeletal abnormalities and impaired T cell activation. Yeast RNase MRP cleaves a specific site in the pre-ribosomal RNA (pre-rRNA) during ribosome synthesis. CRISPR-mediated disruption of RMRP in human cells lines caused growth arrest, with pre-rRNA accumulation. Here, we analyzed disease-relevant primary cells, showing that mutations in RMRP impair mouse T cell activation and delay pre-rRNA processing. Patient-derived human fibroblasts with CHH-linked mutations showed similar pre-rRNA processing delay. Human cells engineered with the most common CHH mutation (70AG in RMRP) show specifically impaired pre-rRNA processing, resulting in reduced mature rRNA and a reduced ratio of cytosolic to mitochondrial ribosomes. Moreover, the 70AG mutation caused a reduction in intact RNase MRP complexes. Together, these results indicate that CHH is a ribosomopathy.


Asunto(s)
Endorribonucleasas/genética , Mutación , ARN Largo no Codificante/genética , ARN Ribosómico/genética , Ribosomas/genética , Animales , Secuencia de Bases , Proliferación Celular/genética , Células Cultivadas , Endorribonucleasas/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Cabello/anomalías , Cabello/metabolismo , Enfermedad de Hirschsprung/genética , Enfermedad de Hirschsprung/metabolismo , Humanos , Células K562 , Ratones Endogámicos C57BL , Ratones Noqueados , Osteocondrodisplasias/congénito , Osteocondrodisplasias/genética , Osteocondrodisplasias/metabolismo , Enfermedades de Inmunodeficiencia Primaria/genética , Enfermedades de Inmunodeficiencia Primaria/metabolismo , Pliegue del ARN , Precursores del ARN/química , Precursores del ARN/genética , Precursores del ARN/metabolismo , ARN Ribosómico/química , ARN Ribosómico/metabolismo , Ribosomas/metabolismo , Linfocitos T/citología , Linfocitos T/metabolismo
6.
Int Immunopharmacol ; 104: 108472, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35008008

RESUMEN

A population pharmacokinetic (PK) model for comparing the PK of subcutaneously administered immunoglobulin G (IgG) replacement therapy (SCIG) with Gamunex-C 10% or SCIG 20% formulations in patients with primary immunodeficiency diseases was developed using data from 3 clinical trials (N = 95, 69.5% adults, 30.5% <18 years) of intravenous IG (IVIG) 10% and SCIG 10% or SCIG 20%. Serum IgG exposure following switches from IVIG 10% every 3 or 4 weeks to biweekly SCIG 20% (dose adjustment factor 1.0 or 1.37) and from weekly SCIG 20% to biweekly SCIG 20% or SCIG 20% 2-7 times/week was simulated. The PK of IVIG 10% and SCIG 20% were adequately described by a 2-compartment model with first-order absorption rate constant of exogenous IgG from an SC depot compartment into the central compartment and first-order elimination from the central compartment. Switching from IVIG 10% every 4 weeks to biweekly SCIG 20% produced similar serum IgG exposure, with lower peak and higher trough serum IgG concentrations. Switching from IVIG 10% every 3 or 4 weeks to weekly and biweekly SCIG 20% yielded comparable IgG exposure and clinically effective trough IgG concentrations.


Asunto(s)
Inmunoglobulina G/administración & dosificación , Modelos Biológicos , Enfermedades de Inmunodeficiencia Primaria/metabolismo , Administración Intravenosa , Adolescente , Adulto , Anciano , Niño , Preescolar , Simulación por Computador , Estudios Cruzados , Femenino , Humanos , Inmunoglobulina G/sangre , Inyecciones Subcutáneas , Masculino , Persona de Mediana Edad , Enfermedades de Inmunodeficiencia Primaria/sangre , Adulto Joven
7.
J Biol Chem ; 298(2): 101551, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34973340

RESUMEN

WHIM syndrome is a rare immunodeficiency disorder that is characterized by warts, hypogammaglobulinemia, infections, and myelokathexis. While several gain-of-function mutations that lead to C-terminal truncations, frame shifts and point mutations in the chemokine receptor CXCR4 have been identified in WHIM syndrome patients, the functional effect of these mutations are not fully understood. Here, we report on a new WHIM syndrome mutation that results in a frame shift within the codon for Ser339 (S339fs5) and compare the properties of S339fs5 with wild-type CXCR4 and a previously identified WHIM syndrome mutant, R334X. The S339fs5 and R334X mutants exhibited significantly increased signaling compared to wild-type CXCR4 including agonist-promoted calcium flux and extracellular-signal-regulated kinase activation. This increase is at least partially due to a significant decrease in agonist-promoted phosphorylation, ß-arrestin binding, and endocytosis of S339fs5 and R334X compared with wild-type CXCR4. Interestingly, there were also significant differences in receptor degradation, with S339fs5 having a very high basal level of degradation compared with that of R334X and wild-type CXCR4. In contrast to wild-type CXCR4, both R334X and S339fs5 were largely insensitive to CXCL12-promoted degradation. Moreover, while basal and agonist-promoted degradation of wild-type CXCR4 was effectively inhibited by the CXCR4 antagonist TE-14016, this had no effect on the degradation of the WHIM mutants. Taken together, these studies identify a new WHIM syndrome mutant, CXCR4-S339fs5, which promotes enhanced signaling, reduced phosphorylation, ß-arrestin binding and endocytosis, and a very high basal rate of degradation that is not protected by antagonist treatment.


Asunto(s)
Enfermedades de Inmunodeficiencia Primaria , Receptores CXCR4 , Verrugas , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Humanos , Enfermedades de Inmunodeficiencia Primaria/genética , Enfermedades de Inmunodeficiencia Primaria/metabolismo , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Verrugas/genética , Verrugas/metabolismo , beta-Arrestina 1/metabolismo , beta-Arrestinas/metabolismo
8.
Front Immunol ; 12: 788278, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34887873

RESUMEN

B-cell lymphoma/leukemia 11B (BCL11B) is a C2H2 zinc finger transcription factor that is critically important for regulating the development and function of a variety of systems including the central nervous system, the skin, and the immune system. Germline heterozygous variants are associated with a spectrum of clinical disorders, including severe combined immunodeficiency as well as neurological, craniofacial, and dermal defects. Of these individuals, ~50% present with severe allergic disease. Here, we report the detailed clinical and laboratory workup of one of the most severe BCL11B-dependent atopic cases to date. Leveraging a zebrafish model, we were able to confirm a strong T-cell defect in the patient. Based on these data, we classify germline BCL11B-dependent atopic disease as a novel primary atopic disorder.


Asunto(s)
Mutación de Línea Germinal , Hipersensibilidad/genética , Enfermedades de Inmunodeficiencia Primaria/genética , Proteínas Represoras/genética , Linfocitos T/inmunología , Proteínas Supresoras de Tumor/genética , Adolescente , Animales , Análisis Mutacional de ADN , Femenino , Predisposición Genética a la Enfermedad , Heterocigoto , Humanos , Hipersensibilidad/diagnóstico , Hipersensibilidad/inmunología , Fenotipo , Enfermedades de Inmunodeficiencia Primaria/diagnóstico , Enfermedades de Inmunodeficiencia Primaria/inmunología , Enfermedades de Inmunodeficiencia Primaria/metabolismo , Proteínas Represoras/metabolismo , Índice de Severidad de la Enfermedad , Linfocitos T/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
9.
Nat Immunol ; 22(7): 893-903, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34155405

RESUMEN

In the present study, we report a human-inherited, impaired, adaptive immunity disorder, which predominantly manifested as a B cell differentiation defect, caused by a heterozygous IKZF3 missense variant, resulting in a glycine-to-arginine replacement within the DNA-binding domain of the encoded AIOLOS protein. Using mice that bear the corresponding variant and recapitulate the B and T cell phenotypes, we show that the mutant AIOLOS homodimers and AIOLOS-IKAROS heterodimers did not bind the canonical AIOLOS-IKAROS DNA sequence. In addition, homodimers and heterodimers containing one mutant AIOLOS bound to genomic regions lacking both canonical motifs. However, the removal of the dimerization capacity from mutant AIOLOS restored B cell development. Hence, the adaptive immunity defect is caused by the AIOLOS variant hijacking IKAROS function. Heterodimeric interference is a new mechanism of autosomal dominance that causes inborn errors of immunity by impairing protein function via the mutation of its heterodimeric partner.


Asunto(s)
Inmunidad Adaptativa , Linfocitos B/metabolismo , Diferenciación Celular , Factor de Transcripción Ikaros/metabolismo , Enfermedades de Inmunodeficiencia Primaria/metabolismo , Linfocitos T/metabolismo , Animales , Linfocitos B/inmunología , Células COS , Chlorocebus aethiops , Modelos Animales de Enfermedad , Femenino , Células HEK293 , Humanos , Factor de Transcripción Ikaros/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación Missense , Células 3T3 NIH , Enfermedades de Inmunodeficiencia Primaria/genética , Enfermedades de Inmunodeficiencia Primaria/inmunología , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Multimerización de Proteína , Transducción de Señal , Linfocitos T/inmunología
10.
Front Immunol ; 12: 655354, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33815417

RESUMEN

Inborn errors of thymic stromal cell development and function lead to impaired T-cell development resulting in a susceptibility to opportunistic infections and autoimmunity. In their most severe form, congenital athymia, these disorders are life-threatening if left untreated. Athymia is rare and is typically associated with complete DiGeorge syndrome, which has multiple genetic and environmental etiologies. It is also found in rare cases of T-cell lymphopenia due to Nude SCID and Otofaciocervical Syndrome type 2, or in the context of genetically undefined defects. This group of disorders cannot be corrected by hematopoietic stem cell transplantation, but upon timely recognition as thymic defects, can successfully be treated by thymus transplantation using cultured postnatal thymic tissue with the generation of naïve T-cells showing a diverse repertoire. Mortality after this treatment usually occurs before immune reconstitution and is mainly associated with infections most often acquired pre-transplantation. In this review, we will discuss the current approaches to the diagnosis and management of thymic stromal cell defects, in particular those resulting in athymia. We will discuss the impact of the expanding implementation of newborn screening for T-cell lymphopenia, in combination with next generation sequencing, as well as the role of novel diagnostic tools distinguishing between hematopoietic and thymic stromal cell defects in facilitating the early consideration for thymus transplantation of an increasing number of patients and disorders. Immune reconstitution after the current treatment is usually incomplete with relatively common inflammatory and autoimmune complications, emphasizing the importance for improving strategies for thymus replacement therapy by optimizing the current use of postnatal thymus tissue and developing new approaches using engineered thymus tissue.


Asunto(s)
Enfermedades de Inmunodeficiencia Primaria/etiología , Enfermedades de Inmunodeficiencia Primaria/metabolismo , Células del Estroma/metabolismo , Timo/anomalías , Timo/metabolismo , Alelos , Animales , Terapia Combinada , Susceptibilidad a Enfermedades , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Humanos , Trasplante de Órganos/efectos adversos , Trasplante de Órganos/métodos , Fenotipo , Enfermedades de Inmunodeficiencia Primaria/diagnóstico , Enfermedades de Inmunodeficiencia Primaria/terapia , Células del Estroma/patología , Timo/patología , Resultado del Tratamiento
11.
Front Immunol ; 12: 615477, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33692789

RESUMEN

Megakaryoblastic leukemia 1 (MKL1) deficiency is one of the most recently discovered primary immunodeficiencies (PIDs) caused by cytoskeletal abnormalities. These immunological "actinopathies" primarily affect hematopoietic cells, resulting in defects in both the innate immune system (phagocyte defects) and adaptive immune system (T-cell and B-cell defects). MKL1 is a transcriptional coactivator that operates together with serum response factor (SRF) to regulate gene transcription. The MKL/SRF pathway has been originally described to have important functions in actin regulation in cells. Recent results indicate that MKL1 also has very important roles in immune cells, and that MKL1 deficiency results in an immunodeficiency affecting the migration and function of primarily myeloid cells such as neutrophils. Interestingly, several actinopathies are caused by mutations in genes which are recognized MKL(1/2)-dependent SRF-target genes, namely ACTB, WIPF1, WDR1, and MSN. Here we summarize these and related (ARPC1B) actinopathies and their effects on immune cell function, especially focusing on their effects on leukocyte adhesion and migration. Furthermore, we summarize recent therapeutic efforts targeting the MKL/SRF pathway in disease.


Asunto(s)
Movimiento Celular , Leucocitos/metabolismo , Enfermedades de Inmunodeficiencia Primaria/etiología , Enfermedades de Inmunodeficiencia Primaria/metabolismo , Factor de Respuesta Sérica/metabolismo , Transactivadores/metabolismo , Animales , Biomarcadores , Adhesión Celular , Movimiento Celular/genética , Movimiento Celular/inmunología , Susceptibilidad a Enfermedades/inmunología , Humanos , Leucocitos/inmunología , Enfermedades de Inmunodeficiencia Primaria/diagnóstico , Factor de Respuesta Sérica/genética , Transducción de Señal , Transactivadores/genética
12.
Turk J Haematol ; 38(1): 1-14, 2021 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-33442967

RESUMEN

Primary immune regulatory disorders (PIRDs) are a group of diseases belonging to inborn errors of immunity. They usually exhibit lymphoproliferation, autoimmunities, and malignancies, with less susceptibility to recurrent infections. Unlike classical primary immune deficiencies, in autoimmune manifestations, such as cytopenias, enteropathy can be the first symptom of diseases, and they are typically resistant to treatment. Increasing awareness of PIRDs among specialists and a multidisciplinary team approach would provide early diagnosis and treatment that could prevent end-organ damage related to the diseases. In recent years, many PIRDs have been described, and understanding the immunological pathways linked to these disorders provides us an opportunity to use directed therapies for specific molecules, which usually offer better disease control than known classical immunosuppressants. In this review, in light of the most recent literature, we will discuss the common PIRDs and explain their clinical symptoms and recent treatment modalities.


Asunto(s)
Predisposición Genética a la Enfermedad , Terapia Genética , Terapia Molecular Dirigida , Enfermedades de Inmunodeficiencia Primaria/etiología , Enfermedades de Inmunodeficiencia Primaria/terapia , Alelos , Animales , Autoinmunidad/genética , Biomarcadores , Citofagocitosis , Citotoxicidad Inmunológica , Mutación con Ganancia de Función , Regulación de la Expresión Génica , Terapia Genética/métodos , Genotipo , Humanos , Inmunomodulación/genética , Linfocitos/inmunología , Linfocitos/metabolismo , Terapia Molecular Dirigida/métodos , Especificidad de Órganos/genética , Especificidad de Órganos/inmunología , Fenotipo , Enfermedades de Inmunodeficiencia Primaria/diagnóstico , Enfermedades de Inmunodeficiencia Primaria/metabolismo , Transducción de Señal
13.
J Immunol Res ; 2020: 8317671, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33274244

RESUMEN

Primary antibody deficiencies (PAD) represent a heterogeneous group of disorders, with common variable immunodeficiency being the most common with clinical significance. The main phenotypic defect resides in the inability of B cells to produce antibodies, and the cornerstone of therapy is immunoglobulin replacement treatment in order to fight infections. However, the management of the other inflammatory manifestations is inadequate, reinforcing the hypothesis that a complex genetic background affecting additional cell populations, such as polymorphonuclear cells (PMN) and monocytes, influences the expression of the clinical phenotype of the disease. In this study, we investigated by flow cytometry in different conditions (resting state, and after isolation and incubation, with and without stimuli) the expression pattern of several markers on PMN and monocytes, indicative of their maturation, capacity for chemotaxis, adhesion, opsonization, migration, and phagocytosis in 25 PAD patients, 12 healthy blood donors, and 4 septic patients. In this context, we also analyzed patients before and after the initiation of replacement treatment, as well as an untreated patient in different clinical conditions. Interestingly, we observed that PAD patients exhibit a chronic activation status of the innate immunity compartment, along with several differences in the expression of activation, maturation, and adhesion markers, with respect to different clinical conditions. Moreover, immunoglobulin replacement treatment had a favorable effect on PMN, as it was expressed by a more mature and less activated phenotype on basal state cells, and an enhanced activation capacity after LPS exposure. Thus, we conclude that PAD patients display a persistent innate immune cell activation, which is probably associated with the chronic inflammatory stress, usually observed in these disorders.


Asunto(s)
Inmunidad Innata , Activación de Linfocitos/inmunología , Linfocitos/inmunología , Enfermedades de Inmunodeficiencia Primaria/etiología , Adolescente , Adulto , Anciano , Biomarcadores , Inmunodeficiencia Variable Común/diagnóstico , Inmunodeficiencia Variable Común/etiología , Inmunodeficiencia Variable Común/metabolismo , Susceptibilidad a Enfermedades , Femenino , Humanos , Inmunofenotipificación , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Linfocitos/metabolismo , Masculino , Persona de Mediana Edad , Monocitos/inmunología , Monocitos/metabolismo , Fenotipo , Enfermedades de Inmunodeficiencia Primaria/diagnóstico , Enfermedades de Inmunodeficiencia Primaria/metabolismo , Adulto Joven
14.
Nat Commun ; 11(1): 5647, 2020 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-33159050

RESUMEN

The human Immunodeficiency Centromeric Instability Facial Anomalies (ICF) 4 syndrome is a severe disease with increased mortality caused by mutation in the LSH gene. Although LSH belongs to a family of chromatin remodeling proteins, it remains unknown how LSH mediates its function on chromatin in vivo. Here, we use chemical-induced proximity to rapidly recruit LSH to an engineered locus and find that LSH specifically induces macroH2A1.2 and macroH2A2 deposition in an ATP-dependent manner. Tethering of LSH induces transcriptional repression and silencing is dependent on macroH2A deposition. Loss of LSH decreases macroH2A enrichment at repeat sequences and results in transcriptional reactivation. Likewise, reduction of macroH2A by siRNA interference mimicks transcriptional reactivation. ChIP-seq analysis confirmed that LSH is a major regulator of genome-wide macroH2A distribution. Tethering of ICF4 mutations fails to induce macroH2A deposition and ICF4 patient cells display reduced macroH2A deposition and transcriptional reactivation supporting a pathogenic role for altered marcoH2A deposition. We propose that LSH is a major chromatin modulator of the histone variant macroH2A and that its ability to insert marcoH2A into chromatin and transcriptionally silence is disturbed in the ICF4 syndrome.


Asunto(s)
ADN Helicasas/metabolismo , Histonas/metabolismo , Enfermedades de Inmunodeficiencia Primaria/metabolismo , Animales , Cromatina/genética , Cromatina/metabolismo , ADN Helicasas/genética , Regulación hacia Abajo , Femenino , Histonas/genética , Humanos , Masculino , Ratones , Enfermedades de Inmunodeficiencia Primaria/enzimología , Enfermedades de Inmunodeficiencia Primaria/genética , Transcripción Genética
15.
Int J Mol Sci ; 21(16)2020 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-32784523

RESUMEN

Given its pleiotropic functions, including its prominent role in inflammation, immune responses and cancer, the C-X-C chemokine receptor type 4 (CXCR4) has gained significant attention in recent years and has become a relevant target in drug development. Although the signaling properties of CXCR4 have been extensively studied, several aspects deserve deeper investigations. Mutations in the C-term tail of the CXCR4 gene cause WHIM syndrome, a rare congenital immunodeficiency associated by chronic leukopenia. Similar mutations have also been recently identified in 30% of patients affected by Waldenstrom's macroglobulinaemia, a B-cell neoplasia with bone marrow accumulation of malignant cells. An ample body of work has been generated to define the impact of WHIM mutations on CXCR4 signaling properties and evaluate their role on pathogenesis, diagnosis, and response to therapy, although the identity of disease-causing signaling pathways and their relevance for disease development in different genetic variants are still open questions. This review discusses the current knowledge on biochemical properties of CXCR4 mutations to identify their prototypic signaling profile potentially useful to highlighting novel opportunities for therapeutic intervention.


Asunto(s)
Enfermedades de Inmunodeficiencia Primaria/metabolismo , Receptores CXCR4/metabolismo , Transducción de Señal , Macroglobulinemia de Waldenström/metabolismo , Verrugas/metabolismo , Humanos , Mutación/genética , Enfermedades de Inmunodeficiencia Primaria/genética , Multimerización de Proteína , Macroglobulinemia de Waldenström/genética , Verrugas/genética
16.
Immunol Allergy Clin North Am ; 40(3): 461-470, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32654692

RESUMEN

Ikaros zinc finger 1 (IKZF1 or Ikaros) is a hematopoietic zinc finger DNA-binding transcription factor that acts as a critical regulator of lymphocyte and myeloid differentiation. Loss-of-function germline heterozygous mutations in IKZF1 affecting DNA-binding were described as causative of 2 distinct primary immunodeficiency (PID)/inborn error of immunity diseases. Mutations acting by haploinsufficiency present with a common variable immune deficiency-like phenotype mainly characterized by increased susceptibility to infections. Mutations acting in a dominant negative fashion present with a combined immunodeficiency phenotype with high prevalence of Pneumocystis jirovecii pneumonia. Pathophysiology and manifestations of IKAROS-associated diseases in patients with PID are reviewed here.


Asunto(s)
Factor de Transcripción Ikaros/genética , Enfermedades de Inmunodeficiencia Primaria/etiología , Enfermedades de Inmunodeficiencia Primaria/metabolismo , Dedos de Zinc/genética , Alelos , Terapia Combinada , Manejo de la Enfermedad , Susceptibilidad a Enfermedades , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Pruebas Genéticas , Variación Genética , Haploinsuficiencia , Humanos , Factor de Transcripción Ikaros/metabolismo , Patrón de Herencia , Fenotipo , Enfermedades de Inmunodeficiencia Primaria/diagnóstico , Enfermedades de Inmunodeficiencia Primaria/terapia
17.
Front Immunol ; 11: 1257, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32695102

RESUMEN

Introduction: Complete or near complete absence of the purine nucleoside phosphorylase (PNP) enzyme causes a profound T cell immunodeficiency and neurological abnormalities that are often lethal in infancy and early childhood. We hypothesized that patients with partial PNP deficiency, characterized by a late and mild phenotype due to residual PNP enzyme, would provide important information about the minimal PNP activity needed for normal development. Methods: Three siblings with a homozygous PNP gene mutation (c.769C>G, p.His257Asp) resulting in partial PNP deficiency were investigated. PNP activity was semi-quantitively assayed by the conversion of [14C]inosine in hemolysates, mononuclear cells, and lymphoblastoid B cells. PNP protein expression was determined by Western Blotting in lymphoblastoid B cells. DNA repair was quantified by measuring viability of lymphoblastoid B cells following ionizing irradiation. Results: A 21-year-old female was referred for recurrent sino-pulmonary infections while her older male siblings, aged 25- and 28- years, did not suffer from significant infections. Two of the siblings had moderately reduced numbers of T, B, and NK cells, while the other had near normal lymphocyte subset numbers. T cell proliferations were normal in the two siblings tested. Hypogammaglobulinemia was noted in two siblings, including one that required immunoglobulin replacement. All siblings had typical (normal) neurological development. PNP activity in various cells from two patients were 8-11% of the normal level. All siblings had normal blood uric acid and increased PNP substrates in the urine. PNP protein expression in cells from the two patients examined was similar to that observed in cells from healthy controls. The survival of lymphoblastoid B cells from 2 partial PNP-deficient patients after irradiation was similar to that of PNP-proficient cells and markedly higher than the survival of cells from a patient with absent PNP activity or a patient with ataxia telangiectasia. Conclusions: Patients with partial PNP deficiency can present in the third decade of life with mild-moderate immune abnormalities and typical development. Near-normal immunity might be achieved with relatively low PNP activity.


Asunto(s)
Neurogénesis , Enfermedades de Inmunodeficiencia Primaria/inmunología , Enfermedades de Inmunodeficiencia Primaria/metabolismo , Purina-Nucleósido Fosforilasa/deficiencia , Purina-Nucleósido Fosforilasa/metabolismo , Errores Innatos del Metabolismo de la Purina-Pirimidina/inmunología , Errores Innatos del Metabolismo de la Purina-Pirimidina/metabolismo , Adulto , Alelos , Análisis Mutacional de ADN , Activación Enzimática , Femenino , Genotipo , Humanos , Inmunofenotipificación , Linfocitos/inmunología , Linfocitos/metabolismo , Linfocitos/efectos de la radiación , Masculino , Mutación , Neurogénesis/genética , Neurogénesis/inmunología , Linaje , Enfermedades de Inmunodeficiencia Primaria/genética , Enfermedades de Inmunodeficiencia Primaria/terapia , Purina-Nucleósido Fosforilasa/genética , Purina-Nucleósido Fosforilasa/inmunología , Errores Innatos del Metabolismo de la Purina-Pirimidina/genética , Errores Innatos del Metabolismo de la Purina-Pirimidina/terapia , Purinas/química , Tolerancia a Radiación , Adulto Joven
18.
Pharmacol Res ; 159: 105010, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32544428

RESUMEN

AMD3100 (plerixafor), a CXCR4 antagonist, has opened a variety of avenues for potential therapeutic approaches in different refractory diseases. The CXCL12/CXCR4 axis and its signaling pathways are involved in diverse disorders including HIV-1 infection, tumor development, non-Hodgkin lymphoma, multiple myeloma, WHIM Syndrome, and so on. The mechanisms of action of AMD3100 may relate to mobilizing hematopoietic stem cells, blocking infection of X4 HIV-1, increasing circulating neutrophils, lymphocytes and monocytes, reducing myeloid-derived suppressor cells, and enhancing cytotoxic T-cell infiltration in tumors. Here, we first revisit the pharmacological discovery of AMD3100. We then review monotherapy of AMD3100 and combination use of AMD3100 with other agents in various diseases. Among those, we highlight the perspective of AMD3100 as an immunomodulator to regulate immune responses particularly in the tumor microenvironment and synergize with other therapeutics. All the pre-clinical studies support the clinical testing of the monotherapy and combination therapies with AMD3100 and further development for use in humans.


Asunto(s)
Fármacos Anti-VIH/uso terapéutico , Antineoplásicos/uso terapéutico , Bencilaminas/uso terapéutico , Ciclamas/uso terapéutico , Infecciones por VIH/tratamiento farmacológico , Neoplasias/tratamiento farmacológico , Receptores CXCR4/antagonistas & inhibidores , Animales , Fármacos Anti-VIH/efectos adversos , Antineoplásicos/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica , Enfermedades Autoinmunes/tratamiento farmacológico , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/metabolismo , Bencilaminas/efectos adversos , Ciclamas/efectos adversos , Contaminación de Medicamentos , Infecciones por VIH/inmunología , Infecciones por VIH/metabolismo , Infecciones por VIH/virología , Humanos , Neoplasias/inmunología , Neoplasias/metabolismo , Enfermedades de Inmunodeficiencia Primaria/tratamiento farmacológico , Enfermedades de Inmunodeficiencia Primaria/inmunología , Enfermedades de Inmunodeficiencia Primaria/metabolismo , Receptores CXCR4/metabolismo , Transducción de Señal , Microambiente Tumoral , Verrugas/tratamiento farmacológico , Verrugas/inmunología , Verrugas/metabolismo
19.
J Leukoc Biol ; 108(2): 687-696, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32542921

RESUMEN

RAC2 is a GTPase that is exclusively expressed in hematopoietic cells. Animal models have suggested important roles for RAC2 in the biology of different cell types, such as neutrophils and lymphocytes. Primary immunodeficiencies represent "experimentum naturae" and offer priceless insight on the function of the human immune system. Mutations in RAC2 have been identified in a small number of patients giving rise to different forms of primary immunodeficiencies ranging from granulocyte defects caused by dominant negative mutations to combined immunodeficiency due to dominant activating mutations. This review will focus on the clinical and immunologic phenotype of patients with germline mutations in RAC2.


Asunto(s)
Susceptibilidad a Enfermedades , Enfermedades de Inmunodeficiencia Primaria/etiología , Enfermedades de Inmunodeficiencia Primaria/metabolismo , Proteínas de Unión al GTP rac/genética , Proteínas de Unión al GTP rac/metabolismo , Animales , Biomarcadores , Susceptibilidad a Enfermedades/inmunología , Predisposición Genética a la Enfermedad , Mutación de Línea Germinal , Humanos , Familia de Multigenes , Mutación , Proteína RCA2 de Unión a GTP
20.
Blood ; 135(24): 2171-2181, 2020 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-32128589

RESUMEN

Megakaryoblastic leukemia 1 (MKL1) promotes the regulation of essential cell processes, including actin cytoskeletal dynamics, by coactivating serum response factor. Recently, the first human with MKL1 deficiency, leading to a novel primary immunodeficiency, was identified. We report a second family with 2 siblings with a homozygous frameshift mutation in MKL1. The index case died as an infant from progressive and severe pneumonia caused by Pseudomonas aeruginosa and poor wound healing. The younger sibling was preemptively transplanted shortly after birth. The immunodeficiency was marked by a pronounced actin polymerization defect and a strongly reduced motility and chemotactic response by MKL1-deficient neutrophils. In addition to the lack of MKL1, subsequent proteomic and transcriptomic analyses of patient neutrophils revealed actin and several actin-related proteins to be downregulated, confirming a role for MKL1 as a transcriptional coregulator. Degranulation was enhanced upon suboptimal neutrophil activation, whereas production of reactive oxygen species was normal. Neutrophil adhesion was intact but without proper spreading. The latter could explain the observed failure in firm adherence and transendothelial migration under flow conditions. No apparent defect in phagocytosis or bacterial killing was found. Also, monocyte-derived macrophages showed intact phagocytosis, and lymphocyte counts and proliferative capacity were normal. Nonhematopoietic primary fibroblasts demonstrated defective differentiation into myofibroblasts but normal migration and F-actin content, most likely as a result of compensatory mechanisms of MKL2, which is not expressed in neutrophils. Our findings extend current insight into the severe immune dysfunction in MKL1 deficiency, with cytoskeletal dysfunction and defective extravasation of neutrophils as the most prominent features.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Mutación del Sistema de Lectura , Neutrófilos/fisiología , Enfermedades de Inmunodeficiencia Primaria/genética , Enfermedades de Inmunodeficiencia Primaria/metabolismo , Transactivadores/deficiencia , Transactivadores/genética , Citoesqueleto de Actina/química , Movimiento Celular/genética , Movimiento Celular/fisiología , Consanguinidad , Femenino , Fibroblastos/metabolismo , Perfilación de la Expresión Génica , Trasplante de Células Madre Hematopoyéticas , Humanos , Lactante , Masculino , Linaje , Polimerizacion , Enfermedades de Inmunodeficiencia Primaria/terapia , Proteómica , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...