Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 182
Filtrar
1.
Mol Genet Genomic Med ; 12(7): e2452, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38967264

RESUMEN

BACKGROUND: Inherited glycosylphosphatidylinositol (GPI) deficiency is an autosomal recessive disease and a set of syndromes caused by different genes involved in the biosynthesis of phosphatidylinositol characterized by severe cognitive disability, elevated serum alkaline phosphatase (ALP) levels, and distinct facial features. This report presents a patient with inherited GPI deficiency caused by a homozygous frameshift variant of PGAP3 due to uniparental isodisomy (UPiD) on chromosome 17. METHOD: Clinical characteristics of the patient were collected. Microarray analysis followed by adaptive sampling sequencing targeting chromosome 17 was used for the identification of variants. Sanger sequencing was used to confirm the variant in the target region. RESULTS: The patient was born at 38 weeks of gestation with a birthweight of 3893 g. He had a distinctive facial appearance with hypertelorism, wide nasal bridge, and cleft soft palate. Postnatal head magnetic resonance imaging revealed a Blake's pouch cyst. The serum ALP level was 940 IU/L at birth and increased to 1781 IU/L at 28 days of age. Microarray analysis revealed region of homozygosity in nearly the entire region of chromosome 17, leading to the diagnosis of UPiD. Adaptive sampling sequencing targeting chromosome 17 confirmed the homozygous variant NM_033419:c.778dupG (p.Val260Glyfs*14) in the PGAP3 gene, resulting in a diagnosis of inherited GPI deficiency. CONCLUSION: This is the first report of inherited GPI deficiency caused by UPiD. Inherited GPI deficiency must be considered in patients with unexplained hyperphosphatasemia.


Asunto(s)
Glicosilfosfatidilinositoles , Disomía Uniparental , Humanos , Masculino , Disomía Uniparental/genética , Disomía Uniparental/patología , Glicosilfosfatidilinositoles/deficiencia , Glicosilfosfatidilinositoles/genética , Errores Innatos del Metabolismo/genética , Errores Innatos del Metabolismo/patología , Homocigoto , Mutación del Sistema de Lectura , Trastornos del Metabolismo del Fósforo/genética , Trastornos del Metabolismo del Fósforo/patología , Hidrolasas de Éster Carboxílico , Receptores de Superficie Celular , Convulsiones
2.
Mol Cancer Res ; 22(6): 515-523, 2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38546397

RESUMEN

The pathogenesis of duodenal tumors in the inherited tumor syndromes familial adenomatous polyposis (FAP) and MUTYH-associated polyposis (MAP) is poorly understood. This study aimed to identify genes that are significantly mutated in these tumors and to explore the effects of these mutations. Whole exome and whole transcriptome sequencing identified recurrent somatic coding variants of phosphatidylinositol N-acetylglucosaminyltransferase subunit A (PIGA) in 19/70 (27%) FAP and MAP duodenal adenomas, and further confirmed the established driver roles for APC and KRAS. PIGA catalyzes the first step in glycosylphosphatidylinositol (GPI) anchor biosynthesis. Flow cytometry of PIGA-mutant adenoma-derived and CRISPR-edited duodenal organoids confirmed loss of GPI anchors in duodenal epithelial cells and transcriptional profiling of duodenal adenomas revealed transcriptional signatures associated with loss of PIGA. IMPLICATIONS: PIGA somatic mutation in duodenal tumors from patients with FAP and MAP and loss of membrane GPI-anchors may present new opportunities for understanding and intervention in duodenal tumorigenesis.


Asunto(s)
Poliposis Adenomatosa del Colon , Neoplasias Duodenales , Glicosilfosfatidilinositoles , Mutación , Humanos , Glicosilfosfatidilinositoles/metabolismo , Glicosilfosfatidilinositoles/genética , Neoplasias Duodenales/genética , Neoplasias Duodenales/metabolismo , Neoplasias Duodenales/patología , Poliposis Adenomatosa del Colon/genética , Poliposis Adenomatosa del Colon/metabolismo , Poliposis Adenomatosa del Colon/patología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Carcinogénesis/genética , Masculino , Femenino
3.
Environ Mol Mutagen ; 64(8-9): 480-493, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37926486

RESUMEN

The blood cell phosphatidylinositol glycan class A (PIG-A) gene mutation assay has been extensively researched in rodents for in vivo mutagenicity testing and is now being investigated in humans. The PIG-A gene is involved in glycosyl phosphatidylinositol (GPI)-anchor biosynthesis. A single mutation in this X-linked gene can lead to loss of membrane-bound GPI anchors, which can be enumerated via corresponding GPI-anchored proteins (e.g., CD55) using flow cytometry. The studies published to date by different research groups demonstrate a remarkable consistency in PIG-A mutant frequencies. Moreover, with the low background level of mutant erythrocytes in healthy subjects (2.9-5.56 × 10-6 mutants), induction of mutation post genotoxic exposure can be detected. Cigarette smoking, radiotherapy, and occupational exposures, including lead, have been shown to increase mutant levels. Future applications of this test include identifying new harmful agents and establishing new exposure limits. This mutational monitoring approach may also identify individuals at higher risk of cancer development. In addition, identifying protective agents that could mitigate these effects may reduce baseline somatic mutation levels and such behaviors can be encouraged. Further technological progress is required including establishing underlying mechanisms of GPI anchor loss, protocol standardization, and the development of cryopreservation methods to improve GPI-anchor stability over time. If successful, this assay has the potential be widely employed, for example, in rural and low-income countries. Here, we review the current literature on PIG-A mutation in humans and discuss the potential role of this assay in human biomonitoring and disease detection.


Asunto(s)
Monitoreo Biológico , Glicosilfosfatidilinositoles , Humanos , Glicosilfosfatidilinositoles/genética , Glicosilfosfatidilinositoles/metabolismo , Proteínas de la Membrana/genética , Mutación , Eritrocitos/metabolismo
4.
Medicina (Kaunas) ; 59(9)2023 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-37763731

RESUMEN

Paroxysmal nocturnal hemoglobinuria (PNH) is a nonmalignant clonal hematopoietic disorder characterized by the lack of glycosylphosphatidylinositol-anchored proteins (GPI-APs) as a consequence of somatic mutations in the phosphatidylinositol glycan anchor biosynthesis class A (PIGA) gene. Clinical manifestations of PNH are intravascular hemolysis, thrombophilia, and bone marrow failure. Treatment of PNH mainly relies on the use of complement-targeted therapy (C5 inhibitors), with the newest agents being explored against other factors involved in the complement cascade to alleviate unresolved intravascular hemolysis and extravascular hemolysis. This review summarizes the biology and current treatment strategies for PNH with the aim of reaching a general audience with an interest in hematologic disorders.


Asunto(s)
Hemoglobinuria Paroxística , Trombofilia , Humanos , Hemoglobinuria Paroxística/tratamiento farmacológico , Hemoglobinuria Paroxística/genética , Hemólisis , Proteínas del Sistema Complemento , Glicosilfosfatidilinositoles/genética , Glicosilfosfatidilinositoles/metabolismo , Biología
5.
Yi Chuan ; 45(8): 669-683, 2023 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-37609818

RESUMEN

In human cells, there are more than 146 glycosylphosphatidylinositol-anchored proteins (GPI-APs), including receptors, ligands, adhesion molecules and enzymes. The proteins are associated with membrane microdomains called lipid rafts through GPI, and plays a variety of important biological functions. At present, plenty of studies have been carried out on the biosynthesis of GPI-APs. The biosynthesis of GPI-APs requires at least 20 steps, and more than 40 GPI biosynthetic genes have been identified. However, it remains unclear how expression of GPI-AP related genes is regulated in normal and cancer tissues. In this study, we utilized gene expression data from both the TCGA database and GTEx portal to analysis the gene expression involved in GPI-AP biosynthesis and encoding GPI-APs in normal and cancer tissues. In order to perform a comprehensive analysis, we employed the GlycoMaple, a tool that is specifically designed to analyze glycosylation pathways. The results showed that compared with normal tissues, the expression of genes involved in GPI-AP biosynthesis in cancer tissues such as early glioma, glioblastoma multiforme, pancreatic cancer, testicular germ cell carcinoma, skin primary cutaneous melanoma and skin metastatic cutaneous melanoma, was changed significantly. Particularly, the expression of PIGY in these six cancers was increased. In addition, the expression of CD14, a GPI-AP gene, was increased in these six cancers. The expression of GAS1, GPC2 and GPC4 was increased only in early glioma and glioblastoma multiforme indicating that some GPI-APs such as GAS1 can be used as biomarkers of glioma. This study provides new insights into the expression of GPI-AP related genes in normal and cancer tissues, and lays a solid foundation for the development of GPI-APs as biomarkers.


Asunto(s)
Glioblastoma , Glioma , Melanoma , Neoplasias Cutáneas , Humanos , Glicosilfosfatidilinositoles/genética , Melanoma Cutáneo Maligno
6.
Front Immunol ; 14: 1329403, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38288112

RESUMEN

Introduction: Paroxysmal nocturnal hemoglobinuria (PNH) is a rare hematological disease characterized by intravascular hemolysis, thrombosis, and bone marrow (BM) failure. Although PNH is caused by excessive proliferation of hematopoietic stem cell (HSC) clones with loss of function mutations in phosphatidylinositol N-acetylglucosaminyltransferase subunit A (PIGA) genes, what drives PNH clones to expand remains elusive. Case description: We present a case of a 26-year-old female who presented with hemolytic anemia, thrombocytopenia, and leukopenia. Flow cytometry analysis of peripheral blood showed that 71.9% and 15.3% of the granulocytes and erythrocytes were glycosylphosphatidylinositol-anchored protein deficient (GPI[-]) cells. The patient was diagnosed with PNH with non-severe aplastic anemia. Deep-targeted sequencing covering 390 different genes of sorted GPI(-) granulocytes revealed three different PIGA mutations (p.I69fs, variant allele frequency (VAF) 24.2%; p.T192P, VAF 5.8%; p.V300fs, VAF 5.1%) and no other mutations. She received six cycles of eculizumab and oral cyclosporine. Although the patient's serum lactate dehydrogenase level decreased, she remained dependent on red blood cell transfusion. Six months after diagnosis, she received a syngeneic bone marrow transplant (BMT) from a genetically identical healthy twin, following an immune ablative conditioning regimen consisting of cyclophosphamide 200 mg/kg and rabbit anti-thymocyte globulin 10 mg/kg. After four years, the patient's blood count remained normal without any signs of hemolysis. However, the peripheral blood still contained 0.2% GPI (-) granulocytes, and the three PIGA mutations that had been detected before BMT persisted at similar proportions to those before transplantation (p.I69fs, VAF 36.1%; p.T192P, VAF 3.7%; p.V300fs, VAF 8.6%) in the small PNH clones that persisted after transplantation. Conclusions: The PNH clones that had increased excessively before BMT decreased, but persisted at low percentages for more than four years after the immunoablative conditioning regimen followed by syngeneic BMT. These findings indicate that as opposed to conventional theory, immune pressure on HSCs, which caused BM failure before BMT, was sufficient for PIGA-mutated HSCs to clonally expand to develop PNH.


Asunto(s)
Anemia Aplásica , Hemoglobinuria Paroxística , Femenino , Humanos , Adulto , Hemoglobinuria Paroxística/diagnóstico , Glicosilfosfatidilinositoles/genética , Células Madre Hematopoyéticas/metabolismo , Anemia Aplásica/genética , Anemia Aplásica/complicaciones , Células Clonales/metabolismo
7.
Semin Hematol ; 59(3): 143-149, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-36115691

RESUMEN

Paroxysmal nocturnal hemoglobinuria (PNH) is an acquired clonal hematopoietic stem cell disorder caused by a mutation of the X-linked PIGA gene, resulting in a deficient expression of glycosylphosphatidylinositol (GPI)-anchored proteins. While large clonal expansions of GPI(-) cells cause hemolytic symptoms, tiny GPI(-) cell populations can be found in healthy individuals and remain miniscule throughout life. The slight expansion of PNH clones often occurs in patients with acquired aplastic anemia (AA), an autoimmune bone marrow (BM) failure caused by autoreactive cytotoxic T lymphocyte attack on hematopoietic stem and progenitor cells (HSPCs). The presence of PNH clones is thought to represent the immune pathophysiology of BM failure and be derived from GPI(-) HSPCs that evaded immune attack against HSPCs. However, which mechanisms underlie the selection of GPI(-) HSPCs as well as their overwhelming clonal expansion remains unclear. Ancestral or secondary somatic mutations in GPI(-) HSPCs contribute to the clonal expansion of the aberrant HSPCs in certain patients with PNH; however, it remains unclear whether such driver mutations are responsible for clonal expansion of all patients. Increased sensitivity to TGF-ß in GPI(-) HSPCs partly explains the predominance of GPI(-) erythrocytes in immune-mediated BM failure. CD4+ T cells specific to antigens presented by HLA-DR15 on HSPCs also contribute to the immune escape of GPI(-) HSPCs. Studying the evolution of HSPCs in AA and PNH will yield further information for understanding human autoimmunity and stem cell biology.


Asunto(s)
Hemoglobinuria Paroxística , Trastornos de Fallo de la Médula Ósea , Glicosilfosfatidilinositoles/genética , Hemoglobinuria Paroxística/genética , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Mutación , Factor de Crecimiento Transformador beta
8.
Nat Commun ; 13(1): 3107, 2022 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-35661110

RESUMEN

Inherited glycosylphosphatidylinositol (GPI) deficiency (IGD) is caused by mutations in GPI biosynthesis genes. The mechanisms of its systemic, especially neurological, symptoms are not clarified and fundamental therapy has not been established. Here, we report establishment of mouse models of IGD caused by PIGO mutations as well as development of effective gene therapy. As the clinical manifestations of IGD are systemic and lifelong lasting, we treated the mice with adeno-associated virus for homology-independent knock-in as well as extra-chromosomal expression of Pigo cDNA. Significant amelioration of neuronal phenotypes and growth defect was achieved, opening a new avenue for curing IGDs.


Asunto(s)
Glicosilfosfatidilinositoles , Convulsiones , Animales , Modelos Animales de Enfermedad , Terapia Genética , Glicosilfosfatidilinositoles/deficiencia , Glicosilfosfatidilinositoles/genética , Inmunoglobulina D/genética , Ratones , Convulsiones/genética
9.
Rinsho Ketsueki ; 62(8): 944-953, 2021.
Artículo en Japonés | MEDLINE | ID: mdl-34497235

RESUMEN

Paroxysmal Nocturnal hemoglobinuria, PNH is usually caused by the somatic mutation of X-linked PIGA gene followed by the clonal expansion of the GPI (glycosylphosphatidylinositol) anchor defective hematopoietic stem cell clone. There are two hypotheses for the mechanism of clonal expansion, one is selection theory, in which GPI deficient cells escape from attacks of cytotoxic cells, and another is benign tumor theory in which GPI deficient cells get the additional mutations and acquire proliferative nature. Recently, we identified two types of PNH patients caused by the biallelic mutation of PIGT on chromosome 20 and PIGB on chromosome 15. Both PNH clones had the germ-line mutation in one allele and another allele was somatically mutated in a hematopoietic stem cell. Both somatic mutations were loss of heterozygosity (LOH), deletion in PIGT-PNH and copy neutral LOH (CN-LOH) in PIGB-PNH. These PNH patients had typical PNH symptoms, but they have in addition auto-inflammatory features. Unlike in PIGA-PNH cells, GPI is synthesized in PIGT-PNH cells and, since its attachment to proteins is blocked, free GPI is expressed on the cell surface. Similarly, in PIGB-PNH cells, GPI intermediates are accumulated and expressed on the cell surface. Those GPIs together with complement activation cause the inflammasome activation.


Asunto(s)
Hemoglobinuria Paroxística , Alelos , Glicosilfosfatidilinositoles/genética , Células Madre Hematopoyéticas , Hemoglobinuria Paroxística/genética , Humanos , Manosiltransferasas , Mutación
10.
Hum Genet ; 140(5): 791-803, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33392778

RESUMEN

PIGK gene, encoding a key component of glycosylphosphatidylinositol (GPI) transamidase, was recently reported to be associated with inherited GPI deficiency disorders (IGDs). However, little is known about the specific downstream effects of PIGK on neurodevelopment due to the rarity of the disease and the lack of in vivo study. Here, we described 2 patients in a Chinese family presented with profound global developmental delay, severe hypotonia, seizures, and postnatal progressive global brain atrophy including hemisphere, cerebellar and corpus callosum atrophy. Two novel compound heterozygous variants in PIGK were identified via genetic analysis, which was proved to cause significant decrease of PIGK protein and reduced cell surface presence of GPI-APs in the patients. To explore the role of Pigk on embryonic and neuronal development, we constructed Pigk knock-down zebrafish and knock-in mouse models. Zebrafish injected with a small dose of morpholino oligonucleotides displayed severe developmental defects including small eyes, deformed head, curly spinal cord, and unconsumed yolk sac. Primary motor neuronal dysplasia and extensive neural cell apoptosis were further observed. Meanwhile, the mouse models, carrying the two variants respectively homologous with the patients, both resulted in complete embryonic lethality of the homozygotes, which suggested the intolerable effect caused by amino acid substitution of Asp204 as well as the truncated mutation. Our findings provide the in vivo evidence for the essential role of PIGK during the embryonic and neuronal development. Based on these data, we propose a basis for further study of pathological and molecular mechanisms of PIGK-related neurodevelopmental defects.


Asunto(s)
Encefalopatías/genética , Moléculas de Adhesión Celular/genética , Glicosilfosfatidilinositoles/deficiencia , Malformaciones del Sistema Nervioso/genética , Neurogénesis/genética , Convulsiones/genética , Anomalías Múltiples/genética , Animales , Apoptosis/genética , Línea Celular , Preescolar , Modelos Animales de Enfermedad , Desarrollo Embrionario/genética , Técnicas de Sustitución del Gen , Glicosilfosfatidilinositoles/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Pez Cebra
11.
Front Immunol ; 12: 830172, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35154088

RESUMEN

Paroxysmal Nocturnal Hemoglobinuria (PNH) is a disease as simple as it is complex. PNH patients develop somatic loss-of-function mutations in phosphatidylinositol N-acetylglucosaminyltransferase subunit A gene (PIGA), required for the biosynthesis of glycosylphosphatidylinositol (GPI) anchors. Ubiquitous in eukaryotes, GPI anchors are a group of conserved glycolipid molecules responsible for attaching nearly 150 distinct proteins to the surface of cell membranes. The loss of two GPI-anchored surface proteins, CD55 and CD59, from red blood cells causes unregulated complement activation and hemolysis in classical PNH disease. In PNH patients, PIGA-mutant, GPI (-) hematopoietic cells clonally expand to make up a large portion of patients' blood production, yet mechanisms leading to clonal expansion of GPI (-) cells remain enigmatic. Historical models of PNH in mice and the more recent PNH model in rhesus macaques showed that GPI (-) cells reconstitute near-normal hematopoiesis but have no intrinsic growth advantage and do not clonally expand over time. Landmark studies identified several potential mechanisms which can promote PNH clonal expansion. However, to what extent these contribute to PNH cell selection in patients continues to be a matter of active debate. Recent advancements in disease models and immunologic technologies, together with the growing understanding of autoimmune marrow failure, offer new opportunities to evaluate the mechanisms of clonal expansion in PNH. Here, we critically review published data on PNH cell biology and clonal expansion and highlight limitations and opportunities to further our understanding of the emergence of PNH clones.


Asunto(s)
Hemoglobinuria Paroxística/etiología , Hemoglobinuria Paroxística/metabolismo , Animales , Autoinmunidad , Biomarcadores , Evolución Clonal/genética , Citocinas/metabolismo , Manejo de la Enfermedad , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Predisposición Genética a la Enfermedad , Glicosilfosfatidilinositoles/genética , Glicosilfosfatidilinositoles/metabolismo , Hematopoyesis/genética , Hemoglobinuria Paroxística/diagnóstico , Hemoglobinuria Paroxística/terapia , Humanos , Subgrupos Linfocitarios/inmunología , Subgrupos Linfocitarios/metabolismo , Subgrupos Linfocitarios/patología , Mutación
12.
J Cell Sci ; 133(3)2020 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-31932507

RESUMEN

GDE2 (also known as GDPD5) is a multispanning membrane phosphodiesterase with phospholipase D-like activity that cleaves select glycosylphosphatidylinositol (GPI)-anchored proteins and thereby promotes neuronal differentiation both in vitro and in vivo GDE2 is a prognostic marker in neuroblastoma, while loss of GDE2 leads to progressive neurodegeneration in mice; however, its regulation remains unclear. Here, we report that, in immature neuronal cells, GDE2 undergoes constitutive endocytosis and travels back along both fast and slow recycling routes. GDE2 trafficking is directed by C-terminal tail sequences that determine the ability of GDE2 to cleave GPI-anchored glypican-6 (GPC6) and induce a neuronal differentiation program. Specifically, we define a GDE2 truncation mutant that shows aberrant recycling and is dysfunctional, whereas a consecutive deletion results in cell-surface retention and gain of GDE2 function, thus uncovering distinctive regulatory sequences. Moreover, we identify a C-terminal leucine residue in a unique motif that is essential for GDE2 internalization. These findings establish a mechanistic link between GDE2 neuronal function and sequence-dependent trafficking, a crucial process gone awry in neurodegenerative diseases.This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Neuroblastoma , Fosfolipasas , Animales , Diferenciación Celular/genética , Glicosilfosfatidilinositoles/genética , Ratones , Hidrolasas Diéster Fosfóricas/genética
13.
Am J Med Genet A ; 179(7): 1270-1275, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31148362

RESUMEN

PIGQ (OMIM *605754) encodes phosphatidylinositol glycan biosynthesis class Q (PIGQ) and is required for proper functioning of an N-acetylglucosamine transferase complex in a similar manner to the more established PIGA, PIGC, and PIGH. There are two previous patients reported with homozygous and apparently deleterious PIGQ mutations. Here, we provide the first detailed clinical report of a patient with heterozygous deleterious mutations associated with glycosylphosphatidylinositol-anchored protein (GPI-AP) biosynthesis deficiency. Our patient died at 10 months of age. The rare skeletal findings in this disorder expand the differential diagnosis of long bone radiolucent lesions and sphenoid wing dysplasia. This clinical report describes a new and rare disorder-PIGQ GPI-AP biosynthesis deficiency syndrome.


Asunto(s)
Anomalías Múltiples/genética , Enfermedades del Desarrollo Óseo/genética , Glicosilfosfatidilinositoles/deficiencia , Proteínas de la Membrana/genética , Hipotonía Muscular/genética , Mutación , Convulsiones/genética , Anomalías Múltiples/diagnóstico , Anomalías Múltiples/metabolismo , Anomalías Múltiples/patología , Enfermedades del Desarrollo Óseo/diagnóstico , Enfermedades del Desarrollo Óseo/metabolismo , Enfermedades del Desarrollo Óseo/patología , Resultado Fatal , Expresión Génica , Glicosilfosfatidilinositoles/genética , Glicosilfosfatidilinositoles/metabolismo , Heterocigoto , Humanos , Lactante , Masculino , Proteínas de la Membrana/deficiencia , Hipotonía Muscular/diagnóstico , Hipotonía Muscular/metabolismo , Hipotonía Muscular/patología , Fenotipo , Convulsiones/diagnóstico , Convulsiones/metabolismo , Convulsiones/patología , Hueso Esfenoides/metabolismo , Hueso Esfenoides/patología , Síndrome , Secuenciación del Exoma
14.
Mol Ther ; 27(5): 912-921, 2019 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-30819613

RESUMEN

Efficient delivery of gene therapy vectors across the blood-brain barrier (BBB) is the holy grail of neurological disease therapies. A variant of the neurotropic vector adeno-associated virus (AAV) serotype 9, called AAV-PHP.B, was shown to very efficiently deliver transgenes across the BBB in C57BL/6J mice. Based on our recent observation that this phenotype is mouse strain dependent, we used whole-exome sequencing-based genetics to map this phenotype to a specific haplotype of lymphocyte antigen 6 complex, locus A (Ly6a) (stem cell antigen-1 [Sca-1]), which encodes a glycosylphosphatidylinositol (GPI)-anchored protein whose function had been thought to be limited to the biology of hematopoiesis. Additional biochemical and genetic studies definitively linked high BBB transport to the binding of AAV-PHP.B with LY6A (SCA-1). These studies identify, for the first time, a ligand for this GPI-anchored protein and suggest a role for it in BBB transport that could be hijacked by viruses in natural infections or by gene therapy vectors to treat neurological diseases.


Asunto(s)
Antígenos Ly/genética , Barrera Hematoencefálica/metabolismo , Técnicas de Transferencia de Gen , Terapia Genética , Proteínas de la Membrana/genética , Animales , Antígenos Ly/farmacología , Transporte Biológico/genética , Encéfalo/efectos de los fármacos , Encéfalo/patología , Dependovirus/genética , Vectores Genéticos/genética , Vectores Genéticos/uso terapéutico , Glicosilfosfatidilinositoles/genética , Hematopoyesis/genética , Humanos , Proteínas de la Membrana/farmacología , Ratones , Neuronas/efectos de los fármacos , Neuronas/patología , Secuenciación del Exoma
15.
Environ Mol Mutagen ; 60(1): 42-46, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30338550

RESUMEN

The Pig-a assay is an emerging and promising in vivo method to determine mutagenic potential of chemicals. Since its development in 2008, remarkable progress has been made in harmonizing and characterizing the test procedures, primarily using known mutagenic chemicals. The purpose of the present study was to evaluate specificity of the Pig-a assay using two nongenotoxic and well-characterized rodent liver carcinogens, phenobarbital and clofibrate, in male F344/DuCrl rats. Daily oral administration of phenobarbital or clofibrate at established hepatotoxic doses for 28 days resulted in substantial hepatic alterations, however, did not increase the frequency of Pig-a mutation markers (RETCD59- and RBCCD59- ) compared to vehicle control or pre-exposure (Day -5) mutant frequencies. These results are consistent with the existing literature on the nonmutagenic mode of action (MoA) of phenobarbital and clofibrate liver tumors. The present study contributes to the limited, but expanding evidence on the specificity of the Pig-a assay and further for the investigations of carcinogenic MoAs, i.e., mutagenic or nonmutagenic potential of chemicals. Environ. Mol. Mutagen. 60:42-46, 2019. © 2018 Wiley Periodicals, Inc.


Asunto(s)
Carcinógenos/toxicidad , Clofibrato/toxicidad , Glicosilfosfatidilinositoles/genética , Neoplasias Hepáticas/inducido químicamente , Pruebas de Mutagenicidad/métodos , Fenobarbital/toxicidad , Animales , Bioensayo , Hígado/efectos de los fármacos , Neoplasias Hepáticas/patología , Masculino , Mutación/efectos de los fármacos , Proyectos Piloto , Ratas , Ratas Endogámicas F344 , Sensibilidad y Especificidad
16.
Environ Mol Mutagen ; 60(1): 47-55, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30264522

RESUMEN

The rodent blood Pig-a assay has been undergoing international validation for use as an in vivo hematopoietic cell gene mutation assay, and given the promising results an Organization for Economic Co-operation and Development (OECD) Test Guideline is currently under development. Enthusiasm for the assay stems in part from its alignment with 3Rs principles permitting combination with other genotoxicity endpoint(s) and integration into repeat-dose toxicology studies. One logistical requirement and experimental design limitation has been that blood samples required antibody labeling and flow cytometric analysis within one week of collection. In the current report, we describe the performance of freeze-thaw reagents that enable storage and subsequent labeling and analysis of rat blood samples for at least seven months. Data generated from three laboratories are presented that demonstrate rat erythrocyte recoveries in the range of 80-90%. Despite some loss of erythrocytes, Pearson coefficients and Bland-Altman analyses based on fresh blood vs. frozen/thawed matched pairs indicate that mutant cell and reticulocyte frequencies are not significantly affected, as the measurements are highly correlated and exhibit low bias. Collectively, these data support the effectiveness and suitability of a freeze-thaw procedure that endows the assay with several new advantageous characteristics that include: flexibility in scheduling personnel/instrumentation; reliability when shipping samples from in-life facilities to analytical sites; 3Rs-friendly, as blood from positive control animals can be stored frozen to serve as analytical controls; and ability to defer a decision to generate Pig-a data until more toxicological information becomes available on a test substance. Environ. Mol. Mutagen. 60:47-55, 2019. © 2018 Wiley Periodicals, Inc.


Asunto(s)
Conservación de la Sangre/métodos , Carboplatino/toxicidad , Eritrocitos/efectos de los fármacos , Etilnitrosourea/toxicidad , Glicosilfosfatidilinositoles/genética , Pruebas de Mutagenicidad/métodos , Mutágenos/toxicidad , Reticulocitos/efectos de los fármacos , Animales , Criopreservación/métodos , Eritrocitos/citología , Femenino , Citometría de Flujo/métodos , Masculino , Ratas , Ratas Sprague-Dawley , Reproducibilidad de los Resultados , Reticulocitos/citología
17.
Am J Hematol ; 94(1): 93-102, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30370942

RESUMEN

Adult B-lymphoblastic leukemia (B-ALL) is a hematological malignancy characterized by genetic heterogeneity. Despite successful remission induction with classical chemotherapeutics and novel targeted agents, enduring remission is often hampered by disease relapse due to outgrowth of a pre-existing subclone resistant against the treatment. In this study, we show that small glycophosphatidylinositol (GPI)-anchor deficient CD52-negative B-cell populations are frequently present already at diagnosis in B-ALL patients, but not in patients suffering from other B-cell malignancies. We demonstrate that the GPI-anchor negative phenotype results from loss of mRNA expression of the PIGH gene, which is involved in the first step of GPI-anchor synthesis. Loss of PIGH mRNA expression within these B-ALL cells follows epigenetic silencing rather than gene mutation or deletion. The coinciding loss of CD52 membrane expression may contribute to the development of resistance to alemtuzumab (ALM) treatment in B-ALL patients resulting in the outgrowth of CD52-negative escape variants. Additional treatment with 5-aza-2'-deoxycytidine may restore expression of CD52 and revert ALM resistance.


Asunto(s)
Linfocitos B/metabolismo , Antígeno CD52/deficiencia , Metilación de ADN/efectos de los fármacos , Regulación Leucémica de la Expresión Génica , Silenciador del Gen , Glicosilfosfatidilinositoles/deficiencia , Proteínas de la Membrana/genética , Proteínas de Neoplasias/deficiencia , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Alemtuzumab/uso terapéutico , Antimetabolitos Antineoplásicos/farmacología , Antimetabolitos Antineoplásicos/uso terapéutico , Antineoplásicos Inmunológicos/uso terapéutico , Linfocitos B/patología , Antígeno CD52/biosíntesis , Antígeno CD52/genética , Línea Celular Tumoral , Decitabina/farmacología , Decitabina/uso terapéutico , Regulación hacia Abajo/efectos de los fármacos , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Glicosilfosfatidilinositoles/biosíntesis , Glicosilfosfatidilinositoles/genética , Humanos , Proteínas de la Membrana/biosíntesis , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Fenotipo , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Neoplásico/biosíntesis , ARN Neoplásico/genética
18.
Cytometry B Clin Cytom ; 94(1): 16-22, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29236352

RESUMEN

Paroxysmal nocturnal hemoglobinuria (PNH) arises as a consequence of the non-malignant clonal expansion of one or more hematopoietic stem cells with an acquired somatic mutation of the PIGA gene (Brodsky RA. Blood 113 (2009) 6522-6527). Progeny of affected stem cells are deficient in glycosyl phosphatidylinositol-anchored proteins (GPI-APs). This deficiency is readily detected by flow cytometry. Though this seems straightforward, the clinical utility of this testing requires that the ordering clinician understand not only the characteristics of the test, but also the biology of the underlying disease, and the clinical and laboratory manifestations in the individual patient. When interpreted correctly, the results from PNH flow cytometry testing, including presence and size of the clonal populations and the cell types involved, can allow the clinician to classify the disease appropriately; evaluate the risk of disease progression; and subsequently monitor response to therapy. In these guidelines, we discuss the evaluation of a patient with suspected PNH or other bone marrow failure disorders, with specific emphasis on the contribution of this testing to the diagnosis, classification, and monitoring of patients. For convenience we will commonly refer to these flow cytometry studies as "PNH testing" recognizing that an abnormal result is not diagnostic of PNH; rather both laboratory and clinical features are used to establish this diagnosis. © 2017 International Clinical Cytometry Society.


Asunto(s)
Citometría de Flujo/normas , Glicosilfosfatidilinositoles/genética , Hemoglobinuria Paroxística/diagnóstico , Hemoglobinuria Paroxística/genética , Adolescente , Anciano , Células Clonales , Progresión de la Enfermedad , Femenino , Células Madre Hematopoyéticas/metabolismo , Humanos , Masculino , Mutación/genética
19.
Environ Mol Mutagen ; 59(1): 18-29, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29115020

RESUMEN

Lack of cell surface glycosylphosphatidylinositol (GPI)-anchored protein(s) has been used as a reporter of Pig-a gene mutation in several model systems. As an extension of this work, our laboratory initiated development of an in vitro mutation assay based on the flow cytometric assessment of CD90.2 expression on the cell surface of the mouse lymphoma cell line L5178Y/Tk+/- . Cells were exposed to mutagenic and nonmutagenic compounds for 24 hr followed by washout and incubation for an additional 7 days. Following this mutant manifestation time, cells were labeled with fluorescent antibodies against CD90.2 and CD45 antigens. These reagents indicated the presence of GPI-anchored proteins and general cell surface membrane receptor integrity, respectively. Instrument set-up was aided by parallel processing of a GPI anchor-deficient subclone. Results show that the mutagens reproducibly caused increased frequencies of mutant phenotype cells, while the nonmutagens did not. Further modifications to the method, including application of a viability dye and an isotype control for instrument set-up, were investigated. As a means to verify that the GPI-anchored protein-negative phenotype reflects bona fide Pig-a gene mutation, sequencing was performed on 38 CD90.2-negative L5178Y/Tk+/- clones derived from cultures treated with ethyl methanesulfonate. All clones were found to have mutation(s) within the Pig-a gene. The continued investigation of L5178Y/Tk+/- cells, CD90.2 labeling, and flow cytometric analysis as the basis of an in vitro mutation assay is clearly supported by this work. These data also provide evidence of the reliability of using GPI anchor-deficiency as a valid reporter of Pig-a gene mutation. Environ. Mol. Mutagen. 59:18-29, 2018. © 2017 Wiley Periodicals, Inc.


Asunto(s)
Glicosilfosfatidilinositoles/deficiencia , Glicosilfosfatidilinositoles/genética , Proteínas de la Membrana/genética , Mutación/genética , Antígenos Thy-1/genética , Animales , Línea Celular Tumoral , Metanosulfonato de Etilo/farmacología , Citometría de Flujo/métodos , Antígenos Comunes de Leucocito/genética , Ratones , Mutagénesis/efectos de los fármacos , Mutagénesis/genética , Pruebas de Mutagenicidad/métodos , Mutágenos/farmacología , Mutación/efectos de los fármacos , Reproducibilidad de los Resultados , Convulsiones
20.
Adv Exp Med Biol ; 1018: 207-224, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29052140

RESUMEN

A wealth of evidence suggests that proteins from prion protein (PrP) family contribute to tumorigenesis in many types of cancers, including pancreatic ductal adenocarcinoma (PDAC), breast cancer, glioblastoma, colorectal cancer, gastric cancer, melanoma, etc. It is well documented that PrP is a biomarker for PDAC, breast cancer, and gastric cancer. However, the underlying mechanisms remain unclear. The major reasons for cancer cell-caused patient death are metastasis and multiple drug resistance, both of which connect to physiological functions of PrP expressing in cancer cells. PrP enhances tumorigenesis by multiple pathways. For example, PrP existed as pro-PrP in most of the PDAC cell lines, thus increasing cancer cell motility by binding to cytoskeletal protein filamin A (FLNa). Using PDAC cell lines BxPC-3 and AsPC-1 as model system, we identified that dysfunction of glycosylphosphatidylinositol (GPI) anchor synthesis machinery resulted in the biogenesis of pro-PrP. In addition, in cancer cells without FLNa expression, pro-PrP can modify cytoskeleton structure by affecting cofilin/F-actin axis, thus influencing cancer cell movement. Besides pro-PrP, we showed that GPI-anchored unglycosylated PrP can elevate cell mobility by interacting with VEGFR2, thus stimulating cell migration under serum-free condition. Besides affecting cancer cell motility, overexpressed PrP or doppel (Dpl) in cancer cells has been shown to increase cell proliferation, multiple drug resistance, and angiogenesis, thus, proteins from PrP gene family by affecting important processes via multiple pathways for cancer cell growth exacerbating tumorigenesis.


Asunto(s)
Adenocarcinoma/genética , Carcinogénesis/genética , Carcinoma Ductal Pancreático/genética , Proteínas Priónicas/genética , Adenocarcinoma/patología , Carcinoma Ductal Pancreático/patología , Filaminas/genética , Proteínas Ligadas a GPI/genética , Glicosilfosfatidilinositoles/genética , Humanos , Metástasis de la Neoplasia , Priones/genética , Transducción de Señal/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA