Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
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
2.
Blood ; 143(15): 1476-1487, 2024 04 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
3.
Nucleic Acids Res ; 52(17): 10194-10219, 2024 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-39142653

RESUMEN

The chromatin-remodeling enzyme helicase lymphoid-specific (HELLS) interacts with cell division cycle-associated 7 (CDCA7) on nucleosomes and is involved in the regulation of DNA methylation in higher organisms. Mutations in these genes cause immunodeficiency, centromeric instability, and facial anomalies (ICF) syndrome, which also results in DNA hypomethylation of satellite repeat regions. We investigated the functional domains of human CDCA7 in HELLS using several mutant CDCA7 proteins. The central region is critical for binding to HELLS, activation of ATPase, and nucleosome sliding activities of HELLS-CDCA7. The N-terminal region tends to inhibit ATPase activity. The C-terminal 4CXXC-type zinc finger domain contributes to CpG and hemimethylated CpG DNA preference for DNA-dependent HELLS-CDCA7 ATPase activity. Furthermore, CDCA7 showed a binding preference to DNA containing hemimethylated CpG, and replication-dependent pericentromeric heterochromatin foci formation of CDCA7 with HELLS was observed in mouse embryonic stem cells; however, all these phenotypes were lost in the case of an ICF syndrome mutant of CDCA7 mutated in the zinc finger domain. Thus, CDCA7 most likely plays a role in the recruitment of HELLS, activates its chromatin remodeling function, and efficiently induces DNA methylation, especially at hemimethylated replication sites.


Asunto(s)
Ensamble y Desensamble de Cromatina , ADN Helicasas , Metilación de ADN , Dedos de Zinc , Humanos , Animales , Ratones , ADN Helicasas/metabolismo , ADN Helicasas/genética , ADN Helicasas/química , Enfermedades de Inmunodeficiencia Primaria/genética , Enfermedades de Inmunodeficiencia Primaria/metabolismo , Islas de CpG/genética , ADN/metabolismo , ADN/genética , Adenosina Trifosfatasas/metabolismo , Adenosina Trifosfatasas/genética , Mutación , Unión Proteica , Nucleosomas/metabolismo , Nucleosomas/genética , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/metabolismo , Dominios Proteicos , Células Madre Embrionarias de Ratones/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética , Heterocromatina/metabolismo , Heterocromatina/genética , Cara/anomalías , Proteínas Nucleares
4.
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
5.
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
6.
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
7.
Immunol Rev ; 291(1): 154-173, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31402502

RESUMEN

Phosphatidylinositol 3 kinases (PI3K) are a family of lipid kinases that are activated by a variety of cell-surface receptors, and regulate a wide range of downstream readouts affecting cellular metabolism, growth, survival, differentiation, adhesion, and migration. The importance of these lipid kinases in lymphocyte signaling has recently been highlighted by genetic analyses, including the recognition that both activating and inactivating mutations of the catalytic subunit of PI3Kδ, p110δ, lead to human primary immunodeficiencies. In this article, we discuss how studies on the human genetic disorder "Activated PI3K-delta syndrome" and mouse models of this disease (Pik3cdE1020K/+ mice) have provided fundamental insight into pathways regulated by PI3Kδ in T and B cells and their contribution to lymphocyte function and disease, including responses to commensal bacteria and the development of autoimmunity and tumors. We highlight critical roles of PI3Kδ in T follicular helper cells and the orchestration of the germinal center reaction, as well as in CD8+ T-cell function. We further  present data demonstrating the ability of the AKT-resistant FOXO1AAA mutant to rescue IgG1 class switching defects in Pik3cdE1020K/+ B cells, as well as data supporting a role for PI3Kδ in promoting multiple T-helper effector cell lineages.


Asunto(s)
Linfocitos B/metabolismo , Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Transducción de Señal , Linfocitos T/metabolismo , Animales , Autoinmunidad , Linfocitos B/inmunología , Biomarcadores , Susceptibilidad a Enfermedades , Metabolismo Energético , Humanos , Inmunoterapia , Neoplasias/etiología , Neoplasias/metabolismo , Neoplasias/terapia , Enfermedades de Inmunodeficiencia Primaria/etiología , Enfermedades de Inmunodeficiencia Primaria/metabolismo , Linfocitos T/inmunología
8.
Blood ; 135(9): 638-643, 2020 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-31942637

RESUMEN

Activated phosphatidylinositol 3-kinase-δ (PI3K-δ) syndrome (APDS) is a rare primary combined immunodeficiency caused by either dominant gain-of-function mutations in the PIK3CD gene encoding the catalytic subunit p110δ of PI3K-δ (referred to as type 1 APDS) or dominant loss-of-function mutations in the PIK3R1 gene encoding the p85α, p55α, and p50α regulatory subunits (type 2 APDS). In types 1 and 2 APDS, the PI3K-δ hyperactivity resulting from the gene mutations leads to similar clinical presentations, characterized by increased susceptibility to bacterial and viral infections and (to a lesser extent) autoimmune manifestations. A hallmark of this disease is lymphoproliferation, which may even be life threatening and require repeated surgical treatment. A major complication of APDS is malignancy (especially B-cell lymphomas), which greatly worsens the prognosis. Here, we review the different neoplastic conditions observed in patients with APDS and discuss the uncontrolled PI3K-δ activity in B and T cells that leads to malignant transformation.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Linfoma de Células B/etiología , Fosfatidilinositol 3-Quinasas/metabolismo , Enfermedades de Inmunodeficiencia Primaria/complicaciones , Enfermedades de Inmunodeficiencia Primaria/metabolismo , Linfocitos B/inmunología , Linfocitos B/metabolismo , Transformación Celular Neoplásica/inmunología , Humanos , Enfermedades de Inmunodeficiencia Primaria/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo
9.
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
10.
Nucleic Acids Res ; 47(16): 8388-8398, 2019 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-31226215

RESUMEN

ZBTB24, encoding a protein of the ZBTB family of transcriptional regulators, is one of four known genes-the other three being DNMT3B, CDCA7 and HELLS-that are mutated in immunodeficiency, centromeric instability and facial anomalies (ICF) syndrome, a genetic disorder characterized by DNA hypomethylation and antibody deficiency. The molecular mechanisms by which ZBTB24 regulates gene expression and the biological functions of ZBTB24 are poorly understood. Here, we identified a 12-bp consensus sequence [CT(G/T)CCAGGACCT] occupied by ZBTB24 in the mouse genome. The sequence is present at multiple loci, including the Cdca7 promoter region, and ZBTB24 binding is mostly associated with gene activation. Crystallography and DNA-binding data revealed that the last four of the eight zinc fingers (ZFs) (i.e. ZF5-8) in ZBTB24 confer specificity of DNA binding. Two ICF missense mutations have been identified in the ZBTB24 ZF domain, which alter zinc-binding cysteine residues. We demonstrated that the corresponding C382Y and C407G mutations in mouse ZBTB24 abolish specific DNA binding and fail to induce Cdca7 expression. Our analyses indicate and suggest a structural basis for the sequence specific recognition by a transcription factor centrally important for the pathogenesis of ICF syndrome.


Asunto(s)
Proteínas de Ciclo Celular/genética , Cara/anomalías , Genoma , Mutación Missense , Proteínas Nucleares/genética , Enfermedades de Inmunodeficiencia Primaria/genética , Proteínas Represoras/química , Factores de Transcripción/química , Dedos de Zinc/genética , Animales , Sitios de Unión , Proteínas de Ciclo Celular/metabolismo , Clonación Molecular , Escherichia coli/genética , Escherichia coli/metabolismo , Cara/patología , Expresión Génica , Sitios Genéticos , Vectores Genéticos , Humanos , Ratones , Modelos Moleculares , Proteínas Nucleares/metabolismo , Motivos de Nucleótidos , Enfermedades de Inmunodeficiencia Primaria/metabolismo , Enfermedades de Inmunodeficiencia Primaria/patología , Regiones Promotoras Genéticas , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
11.
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
12.
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
13.
J Clin Immunol ; 39(2): 216-224, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30911954

RESUMEN

PURPOSE: Primary ciliary dyskinesia (PCD) is a rare disorder of the mucociliary clearance leading to recurrent upper and lower respiratory tract infections. PCD is difficult to clinically distinguish from other entities leading to recurrent oto-sino-pulmonary infections, including primary immunodeficiency (PID). Nasal nitric oxide (nNO) is a sensitive and specific diagnostic test for PCD, but it has not been thoroughly examined in PID. Past publications have suggested an overlap in nNO levels among subjects with PCD and PID. We sought to determine if nNO measurements among patients diagnosed with PID would fall significantly above the established PCD diagnostic cutoff value of 77 nL/min. METHODS: Children > 5 years old and adults with definitive PID or PCD diagnoses were recruited from outpatient subspecialty clinics. Participants underwent nNO testing by standardized protocol using a chemiluminescence analyzer and completed a questionnaire concerning their chronic oto-sino-pulmonary symptoms, including key clinical criteria specific to diagnosed PCD (neonatal respiratory distress at term birth, year-round cough or nasal congestion starting before 6 months of age, any organ laterality defect). RESULTS: Participants included 32 patients with PID, 27 patients with PCD, and 19 healthy controls. Median nNO was 228.9.1 nL/min in the PID group, 19.7 nL/min in the PCD group, and 269.4 in the healthy controls (p < 0.0001). Subjects with PCD were significantly more likely to report key clinical criteria specific to PCD, but approximately 25% of PID subjects also reported at least 1 of these key clinical criteria (mainly year-round cough or nasal congestion). CONCLUSIONS: While key clinical criteria associated with PCD often overlap with the symptoms reported in PID, nNO measurement by chemiluminescence technology allows for effective discrimination between PID and PCD.


Asunto(s)
Trastornos de la Motilidad Ciliar/diagnóstico , Óxido Nítrico/metabolismo , Enfermedades de Inmunodeficiencia Primaria/diagnóstico , Adolescente , Adulto , Niño , Trastornos de la Motilidad Ciliar/metabolismo , Diagnóstico Diferencial , Femenino , Humanos , Masculino , Persona de Mediana Edad , Nariz , Enfermedades de Inmunodeficiencia Primaria/metabolismo , Adulto Joven
14.
J Autoimmun ; 99: 52-72, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30795880

RESUMEN

The primary immunodeficiency diseases (PIDs) include many genetic disorders that affect different components of the innate and adaptive responses. The number of distinct genetic PIDs has increased exponentially with improved methods of detection and advanced laboratory methodology. Patients with PIDs have an increased susceptibility to infectious diseases and non-infectious complications including allergies, malignancies and autoimmune diseases (ADs), the latter being the first manifestation of PIDs in several cases. There are two types of PIDS. Monogenic immunodeficiencies due to mutations in genes involved in immunological tolerance that increase the predisposition to develop autoimmunity including polyautoimmunity, and polygenic immunodeficiencies characterized by a heterogeneous clinical presentation that can be explained by a complex pathophysiology and which may have a multifactorial etiology. The high prevalence of ADs in PIDs demonstrates the intricate relationships between the mechanisms of these two conditions. Defects in central and peripheral tolerance, including mutations in AIRE and T regulatory cells respectively, are thought to be crucial in the development of ADs in these patients. In fact, pathology that leads to PID often also impacts the Treg/Th17 balance that may ease the appearance of a proinflammatory environment, increasing the odds for the development of autoimmunity. Furthermore, the influence of chronic and recurrent infections through molecular mimicry, bystander activation and super antigens activation are supposed to be pivotal for the development of autoimmunity. These multiple mechanisms are associated with diverse clinical subphenotypes that hinders an accurate diagnosis in clinical settings, and in some cases, may delay the selection of suitable pharmacological therapies. Herein, a comprehensively appraisal of the common mechanisms among these conditions, together with clinical pearls for treatment and diagnosis is presented.


Asunto(s)
Autoinmunidad , Enfermedades de Inmunodeficiencia Primaria/etiología , Enfermedades Autoinmunes/diagnóstico , Enfermedades Autoinmunes/etiología , Enfermedades Autoinmunes/metabolismo , Enfermedades Autoinmunes/terapia , Manejo de la Enfermedad , Susceptibilidad a Enfermedades/inmunología , Epítopos/inmunología , Humanos , Tolerancia Inmunológica , Síndromes de Inmunodeficiencia/diagnóstico , Síndromes de Inmunodeficiencia/etiología , Síndromes de Inmunodeficiencia/metabolismo , Infecciones/complicaciones , Imitación Molecular , Enfermedades de Inmunodeficiencia Primaria/diagnóstico , Enfermedades de Inmunodeficiencia Primaria/metabolismo , Enfermedades de Inmunodeficiencia Primaria/terapia , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
15.
Curr Allergy Asthma Rep ; 19(11): 52, 2019 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-31741098

RESUMEN

PURPOSE OF REVIEW: Hematopoietic cell transplantation (HCT) is an established curative treatment for children with primary immunodeficiencies. This article reviews the latest developments in conditioning regimens for primary immunodeficiency (PID). It focuses on data regarding transplant outcomes according to newer reduced toxicity conditioning regimens used in HCT for PID. RECENT FINDINGS: Conventional myeloablative conditioning regimens are associated with significant acute toxicities, transplant-related mortality, and late effects such as infertility. Reduced toxicity conditioning regimens have had significant positive impacts on HCT outcome, and there are now well-established strategies in children with PID. Treosulfan has emerged as a promising preparative agent. Use of a peripheral stem cell source has been shown to be associated with better donor chimerism in patients receiving reduced toxicity conditioning. Minimal conditioning regimens using monoclonal antibodies are in clinical trials with promising results thus far. Reduced toxicity conditioning has emerged as standard of care for PID and has resulted in improved transplant survival for patients with significant comorbidities.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas/métodos , Enfermedades de Inmunodeficiencia Primaria/terapia , Acondicionamiento Pretrasplante/métodos , Busulfano/análogos & derivados , Busulfano/farmacocinética , Busulfano/uso terapéutico , Humanos , Inmunosupresores/farmacocinética , Inmunosupresores/uso terapéutico , Enfermedades de Inmunodeficiencia Primaria/metabolismo , Vidarabina/análogos & derivados , Vidarabina/farmacocinética , Vidarabina/uso terapéutico
17.
Sci Adv ; 10(34): eadr0036, 2024 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-39178265

RESUMEN

CDCA7, encoding a protein with a carboxyl-terminal cysteine-rich domain (CRD), is mutated in immunodeficiency, centromeric instability, and facial anomalies (ICF) syndrome, a disease related to hypomethylation of juxtacentromeric satellite DNA. How CDCA7 directs DNA methylation to juxtacentromeric regions is unknown. Here, we show that the CDCA7 CRD adopts a unique zinc-binding structure that recognizes a CpG dyad in a non-B DNA formed by two sequence motifs. CDCA7, but not ICF mutants, preferentially binds the non-B DNA with strand-specific CpG hemi-methylation. The unmethylated sequence motif is highly enriched at centromeres of human chromosomes, whereas the methylated motif is distributed throughout the genome. At S phase, CDCA7, but not ICF mutants, is concentrated in constitutive heterochromatin foci, and the formation of such foci can be inhibited by exogenous hemi-methylated non-B DNA bound by the CRD. Binding of the non-B DNA formed in juxtacentromeric regions during DNA replication provides a mechanism by which CDCA7 controls the specificity of DNA methylation.


Asunto(s)
Centrómero , Islas de CpG , Metilación de ADN , Síndromes de Inmunodeficiencia , Enfermedades de Inmunodeficiencia Primaria , Unión Proteica , Humanos , Enfermedades de Inmunodeficiencia Primaria/metabolismo , Enfermedades de Inmunodeficiencia Primaria/genética , Síndromes de Inmunodeficiencia/metabolismo , Síndromes de Inmunodeficiencia/genética , Centrómero/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/química , Dominios Proteicos , ADN/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/química , Mutación , Heterocromatina/metabolismo , Heterocromatina/genética , Cara/anomalías , Proteínas Nucleares
18.
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
19.
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
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA