Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Leuk Res ; 137: 107441, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38301422

RESUMEN

Inherited bone marrow failure syndromes and germline predisposition syndromes (IBMFS/GPS) are associated with increased risk for hematologic malignancies, particularly myeloid neoplasms, such as myelodysplastic neoplasms (MDS) and acute myeloid leukemia (AML). The diagnosis of MDS in these syndromes poses difficulty due to frequent bone marrow hypocellularity and the presence of some degree of dysplastic features related to the underlying germline defect causing abnormal maturation of one or more cell lines. Yet, the diagnosis of MDS is usually associated with a worse outcome in several IBMFS/GPS. Criteria for the diagnosis of MDS in IBMFS/GPS have not been standardized with some authors suggesting a mixture of morphologic, cytogenetic, and genetic criteria. This review highlights these challenges and suggests a more standardized approach to nomenclature and diagnostic criteria.


Asunto(s)
Enfermedades de la Médula Ósea , Leucemia Mieloide Aguda , Síndromes Mielodisplásicos , Humanos , Enfermedades de la Médula Ósea/genética , Enfermedades de la Médula Ósea/complicaciones , Enfermedades de la Médula Ósea/patología , Síndromes Congénitos de Insuficiencia de la Médula Ósea/complicaciones , Síndromes Mielodisplásicos/diagnóstico , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/patología , Leucemia Mieloide Aguda/genética , Predisposición Genética a la Enfermedad , Células Germinativas/patología
2.
Eur J Paediatr Neurol ; 49: 60-65, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38377647

RESUMEN

Fatty acid oxidation (FAO) disorders are autosomal recessive genetic disorders affecting either the transport or the oxidation of fatty acids. Acute symptoms arise during prolonged fasting, intercurrent infections, or intense physical activity. Metabolic crises are characterized by alteration of consciousness, hypoglycemic coma, hepatomegaly, cardiomegaly, arrhythmias, rhabdomyolysis, and can lead to death. In this retrospective and multicentric study, the data of 54 patients with FAO disorders were collected. Overall, 35 patients (64.8%) were diagnosed after newborn screening (NBS), 17 patients on clinical presentation (31.5%), and two patients after family screening (3.7%). Deficiencies identified included medium-chain acyl-CoA dehydrogenase (MCAD) deficiency (75.9%), very long-chain acyl-CoA dehydrogenase (VLCAD) deficiency (11.1%), long-chain hydroxyacyl-CoA dehydrogenase (LCHAD) deficiency (3.7%), mitochondrial trifunctional protein (MTP) deficiency (1.8%), and carnitine palmitoyltransferase 2 (CPT 2) deficiency (7.4%). The NBS results of 25 patients were reviewed and the neurological outcome of this population was compared with that of the patients who were diagnosed on clinical presentation. This article sought to provide a comprehensive overview of how NBS implementation in Southern Belgium has dramatically improved the neurological outcome of patients with FAO disorders by preventing metabolic crises and death. Further investigations are needed to better understand the physiopathology of long-term complications in order to improve the quality of life of patients and to ensure optimal management.


Asunto(s)
Acil-CoA Deshidrogenasa/deficiencia , Cardiomiopatías , Carnitina O-Palmitoiltransferasa/deficiencia , Errores Innatos del Metabolismo Lipídico , Errores Innatos del Metabolismo , Proteína Trifuncional Mitocondrial/deficiencia , Tamizaje Neonatal , Rabdomiólisis , Humanos , Recién Nacido , Estudios Retrospectivos , Masculino , Femenino , Tamizaje Neonatal/métodos , Errores Innatos del Metabolismo Lipídico/diagnóstico , Errores Innatos del Metabolismo Lipídico/complicaciones , Bélgica/epidemiología , Lactante , Síndromes Congénitos de Insuficiencia de la Médula Ósea/complicaciones , Síndromes Congénitos de Insuficiencia de la Médula Ósea/diagnóstico , Acil-CoA Deshidrogenasa de Cadena Larga/deficiencia , Ácidos Grasos/metabolismo , Preescolar , Enfermedades Musculares/diagnóstico , Niño , Miopatías Mitocondriales/diagnóstico , Miopatías Mitocondriales/complicaciones , Enfermedades Mitocondriales/diagnóstico , Enfermedades Mitocondriales/complicaciones , Enfermedades del Sistema Nervioso/etiología , Enfermedades del Sistema Nervioso/diagnóstico
3.
Hematology Am Soc Hematol Educ Program ; 2023(1): 141-148, 2023 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-38066882

RESUMEN

Inherited bone marrow failure syndromes (IBMFS) encompass a group of rare genetic disorders characterized by bone marrow failure, non-hematologic multisystemic comorbidities, disease defining congenital anomalies, and a susceptibility to myelodysplastic syndrome, acute myeloid leukemia, and in some instances solid tumors. The most common IBMFS include Fanconi anemia, Shwachman-Diamond syndrome, Diamond-Blackfan anemia, and telomere biology disorders/ dyskeratosis congenita. Allogeneic hematopoietic stem cell transplant (HCT) is a well-established curative treatment to correct the hematological manifestations but does not halt or reverse the nonhematological complications and may hasten them. With advances in HCT and in our ability to care for patients with IBMFS, an increasing number of survivors are making it imperative to not only diagnose but also treat late effects from the pre-, peri-, and post-HCT course and complications relating to the natural history of the syndrome. As the field of HCT evolves to allow for the incorporation of alternate graft sources, for expansion of donor options to include unrelated and mismatched donors, and for use of reduced-intensity conditioning or reduced toxicity myeloablative regimens, we have yet to determine if these advances modify the disease-specific course. While long-term outcomes of these patients are often included under one umbrella, this article seeks to address disease-specific post-HCT outcomes within IBMFS.


Asunto(s)
Anemia Aplásica , Enfermedades de la Médula Ósea , Trasplante de Células Madre Hematopoyéticas , Hemoglobinuria Paroxística , Humanos , Anemia Aplásica/genética , Enfermedades de la Médula Ósea/terapia , Enfermedades de la Médula Ósea/diagnóstico , Médula Ósea/patología , Síndromes Congénitos de Insuficiencia de la Médula Ósea/complicaciones , Hemoglobinuria Paroxística/diagnóstico , Hemoglobinuria Paroxística/terapia , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Progresión de la Enfermedad
4.
Hematology Am Soc Hematol Educ Program ; 2023(1): 135-140, 2023 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-38066900

RESUMEN

Hematopoietic cell transplantation (HCT) can cure blood dyscrasias and reduce the risk of hematologic cancers in patients with inherited bone marrow failure syndromes (IBMFS). However, because of its high mortality rate, HCT is generally reserved until patients with IBMFS manifest life-threatening cytopenias or myeloid malignancy, at which point outcomes are poor. Screening tests that accurately predict transformation and enable timely intervention are lacking. These unknowns and risks limit the use of HCT in patients with IBMFS, sometimes until significant disease-related sequelae have occurred. A major goal for IBMFS is to reduce cellular therapy-related complications to the point that earlier intervention can be considered before significant transfusion exposure, occurrence of comorbidities, or malignant transformation. In recent decades, disease-specific allogeneic HCT trials have yielded significant improvements in outcomes in IBMFS conditions, including Fanconi anemia and dyskeratosis congenita. This is in large part due to marked reductions in conditioning intensity to address the increased sensitivity of these patients to cytotoxic chemotherapy and radiation. The success of these approaches may also indicate an ability to leverage intrinsic fitness defects of hematopoietic stem and progenitor cells across IBMFS disorders. Now with advances in tracking somatic genetic evolution in hematopoiesis and tailored minimal intensity conditioning regimens, this question arises: is it time for preventative HCT for IBMFS?


Asunto(s)
Anemia Aplásica , Enfermedades de la Médula Ósea , Trasplante de Células Madre Hematopoyéticas , Hemoglobinuria Paroxística , Pancitopenia , Humanos , Anemia Aplásica/genética , Enfermedades de la Médula Ósea/terapia , Enfermedades de la Médula Ósea/diagnóstico , Síndromes Congénitos de Insuficiencia de la Médula Ósea/complicaciones , Hemoglobinuria Paroxística/genética , Trastornos de Fallo de la Médula Ósea , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Acondicionamiento Pretrasplante
5.
Semin Diagn Pathol ; 40(6): 429-442, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37507252

RESUMEN

The diagnostic work up and surveillance of germline disorders of bone marrow failure and predisposition to myeloid malignancy is complex and involves correlation between clinical findings, laboratory and genetic studies, and bone marrow histopathology. The rarity of these disorders and the overlap of clinical and pathologic features between primary and secondary causes of bone marrow failure, acquired aplastic anemia, and myelodysplastic syndrome may result in diagnostic uncertainty. With an emphasis on the pathologist's perspective, we review diagnostically useful features of germline disorders including Fanconi anemia, Shwachman-Diamond syndrome, telomere biology disorders, severe congenital neutropenia, GATA2 deficiency, SAMD9/SAMD9L diseases, Diamond-Blackfan anemia, and acquired aplastic anemia. We discuss the distinction between baseline morphologic and genetic findings of these disorders and features that raise concern for the development of myelodysplastic syndrome.


Asunto(s)
Anemia Aplásica , Enfermedades de la Médula Ósea , Síndromes Mielodisplásicos , Trastornos Mieloproliferativos , Neoplasias , Humanos , Anemia Aplásica/genética , Anemia Aplásica/complicaciones , Síndromes Congénitos de Insuficiencia de la Médula Ósea/complicaciones , Enfermedades de la Médula Ósea/genética , Enfermedades de la Médula Ósea/complicaciones , Enfermedades de la Médula Ósea/diagnóstico , Patólogos , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/complicaciones , Trastornos de Fallo de la Médula Ósea/complicaciones , Células Germinativas , Neoplasias/complicaciones , Péptidos y Proteínas de Señalización Intracelular
6.
Leukemia ; 37(8): 1698-1708, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37391485

RESUMEN

Many inherited bone marrow failure syndromes (IBMFSs) present a high risk of transformation to myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML). During transformation of IBMFSs, hematopoietic stem and progenitor cells (HSPCs) with poor fitness gain ectopic, dysregulated self-renewal secondary to somatic mutations via undefined mechanisms. Here, in the context of the prototypical IBMFS Fanconi anemia (FA), we performed multiplexed gene editing of mutational hotspots in MDS-associated genes in human induced pluripotent stem cells (iPSCs) followed by hematopoietic differentiation. We observed aberrant self-renewal and impaired differentiation of HSPCs with enrichment of RUNX1 insertions and deletions (indels), generating a model of IBMFS-associated MDS. We observed that compared to the failure state, FA MDS cells show mutant RUNX1-mediated blunting of the G1/S cell cycle checkpoint that is normally activated in FA in response to DNA damage. RUNX1 indels also lead to activation of innate immune signaling, which stabilizes the homologous recombination (HR) effector BRCA1, and this pathway can be targeted to abrogate viability and restore sensitivity to genotoxins in FA MDS. Together, these studies develop a paradigm for modeling clonal evolution in IBMFSs, provide basic understanding of the pathogenesis of MDS, and uncover a therapeutic target in FA-associated MDS.


Asunto(s)
Anemia de Fanconi , Células Madre Pluripotentes Inducidas , Leucemia Mieloide Aguda , Síndromes Mielodisplásicos , Humanos , Anemia de Fanconi/genética , Anemia de Fanconi/patología , Anemia de Fanconi/terapia , Síndromes Congénitos de Insuficiencia de la Médula Ósea/complicaciones , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Células Madre Pluripotentes Inducidas/patología , Síndromes Mielodisplásicos/patología , Mutación , Leucemia Mieloide Aguda/patología
7.
J Neonatal Perinatal Med ; 16(1): 183-186, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36872795

RESUMEN

BACKGROUND: Thrombocytopenia absent radius (TAR) syndrome is a rare disease with an estimated prevalence of one in 200,000 live births. TAR is associated with cardiac and renal anomalies as well as gastrointestinal problems such as cow's milk protein allergy (CMPA). Typically neonates with CMPA present with mild intolerance, with few reports in the literature of more severe intolerance resulting in pneumatosis. We present a case of a male infant with TAR syndrome who developed gastric and colonic pneumatosis intestinalis. CASE DESCRIPTION: An eight-day-old male infant born at 36 weeks gestation with a diagnosis of TAR, presented with bright red blood in his stool. At this time he was on full formula feeds. Given continued bright red blood within his stool, an abdominal radiograph was obtained which was consistent with colonic and gastric pneumatosis. A complete blood count (CBC) was notable for worsening thrombocytopenia, anemia and eosinophilia. Once enteral feeds were held there was rapid resolution of the radiographic findings and resolution of his bloody stool. He was ultimately diagnosed with a CMPA. CONCLUSION: Though there are reports of CMPA in patients with TAR, the severity of this patient's presentation with both colonic and gastric pneumatosis is unique. Without the knowledge of the association of CMPA with TAR, this case could have been misdiagnosed and led to reintroduction of cow's milk containing formula, resulting in further complications. This case highlights the importance of a timely diagnosis and severity of CMPA in this population.


Asunto(s)
Hipersensibilidad a la Leche , Trombocitopenia , Animales , Femenino , Bovinos , Masculino , Radio (Anatomía)/diagnóstico por imagen , Hipersensibilidad a la Leche/complicaciones , Hipersensibilidad a la Leche/diagnóstico , Hipersensibilidad a la Leche/epidemiología , Síndromes Congénitos de Insuficiencia de la Médula Ósea/complicaciones , Trombocitopenia/etiología , Proteínas de la Leche
8.
Pediatr Res ; 92(6): 1671-1680, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35322185

RESUMEN

BACKGROUND AND OBJECTIVES: Shwachman Diamond syndrome (SDS) is an inherited bone marrow failure syndrome (IBMFS) associated with pancreatic insufficiency, neutropenia, and skeletal dysplasia. Biallelic pathogenic variants (PV) in SBDS account for >90% of SDS. We hypothesized that the SDS phenotype varies based on genotype and conducted a genotype-phenotype correlation study to better understand these complexities. METHODS: We reviewed records of all patients with SDS or SDS-like syndromes in the National Cancer Institute's (NCI) IBMFS study. Additional published SDS cohorts were reviewed and compared with the NCI cohort. RESULTS: PVs in SBDS were present in 32/47 (68.1%) participants. Biallelic inheritance of SBDS c.258 + 2T > C and c.183_184TA > CT was the most common genotype in our study (25/32, 78.1%) and published cohorts. Most patients had the SDS hallmark features of neutropenia (45/45, 100%), pancreatic insufficiency (41/43, 95.3%), and/or bony abnormalities (29/36, 80.6%). Developmental delay was common (20/34, 58.8%). Increased risk of hematologic malignancies at young ages and the rarity of solid malignancies was observed in both the NCI cohort and published studies. CONCLUSIONS: SDS is a complex childhood illness with a narrow genotypic spectrum. Patients may first present to primary care, gastroenterology, orthopedic, and/or hematology clinics. Coordinated multidisciplinary care is important for diagnosis and patient management. CLINICAL TRIAL REGISTRATION: ClinicalTrials.gov Identifier: NCT00027274. IMPACT: The clinical and genetic spectrum of Shwachman Diamond Syndrome was comprehensively evaluated, and the findings illustrate the importance of a multidisciplinary approach for these complex patients. Our work reveals: 1. a narrow genotypic spectrum in SDS; 2. a low risk of solid tumors in patients with SDS; 3. patients with SDS have clinical manifestations in multiple organ systems.


Asunto(s)
Enfermedades de la Médula Ósea , Insuficiencia Pancreática Exocrina , Lipomatosis , Neutropenia , Humanos , Síndrome de Shwachman-Diamond/complicaciones , Enfermedades de la Médula Ósea/genética , Síndromes Congénitos de Insuficiencia de la Médula Ósea/complicaciones , Lipomatosis/diagnóstico , Insuficiencia Pancreática Exocrina/diagnóstico , Insuficiencia Pancreática Exocrina/genética , Neutropenia/genética , Genotipo
9.
Hematology Am Soc Hematol Educ Program ; 2021(1): 514-520, 2021 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-34889405

RESUMEN

A common feature of both congenital and acquired forms of bone marrow failure is an increased risk of developing acute myeloid leukemia (AML) or myelodysplastic syndrome (MDS). Indeed, the development of MDS or AML is now the major cause of mortality in patients with congenital neutropenia. Thus, there is a pressing clinical need to develop better strategies to prevent, diagnose early, and treat MDS/AML in patients with congenital neutropenia and other bone marrow failure syndromes. Here, we discuss recent data characterizing clonal hematopoiesis and progression to myeloid malignancy in congenital neutropenia, focusing on severe congenital neutropenia (SCN) and Shwachman-Diamond syndrome. We summarize recent studies showing excellent outcomes after allogenic hematopoietic stem cell transplantation for many (but not all) patients with congenital neutropenia, including patients with SCN with active myeloid malignancy who underwent transplantation. Finally, we discuss how these new data inform the current clinical management of patients with congenital neutropenia.


Asunto(s)
Síndromes Congénitos de Insuficiencia de la Médula Ósea/complicaciones , Leucemia Mieloide Aguda/etiología , Síndromes Mielodisplásicos/etiología , Mielopoyesis , Neutropenia/congénito , Preescolar , Hematopoyesis Clonal , Síndromes Congénitos de Insuficiencia de la Médula Ósea/fisiopatología , Síndromes Congénitos de Insuficiencia de la Médula Ósea/terapia , Trasplante de Células Madre Hematopoyéticas , Humanos , Lactante , Leucemia Mieloide Aguda/fisiopatología , Leucemia Mieloide Aguda/terapia , Persona de Mediana Edad , Síndromes Mielodisplásicos/fisiopatología , Síndromes Mielodisplásicos/terapia , Neutropenia/complicaciones , Neutropenia/fisiopatología , Neutropenia/terapia , Síndrome de Shwachman-Diamond/complicaciones , Síndrome de Shwachman-Diamond/fisiopatología , Síndrome de Shwachman-Diamond/terapia
10.
Mol Genet Metab ; 134(4): 301-308, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34862134

RESUMEN

There is a limited understanding of system-level clinical outcomes and interventions associated with single large-scale mitochondrial DNA deletion syndromes (SLSMDS). Additionally, no research exists that describes patient reported outcomes (PROs) of children with SLSMDS. A global and observational registry was established to understand the multi-systemic course of SLSMDS and track clinical outcomes. The development and design of the registry is described. Demographic characteristics, history and diagnoses, and system level prevalence of problems and interventions are reported for 42 children. System level problems and interventions include information on the following body systems: audiology, cardiac, endocrine, gastrointestinal (including pancreatic and hepatobiliary system), hematological, metabolic, neurological (including autonomic, mobility, & learning), ophthalmic, psychiatric, renal, and respiratory. Results emphasize the need of patient registries and suggest that the diagnostic odyssey and burden of disease for children with SLSMDS is significant. System-level findings may help families and clinical providers with diagnosis, prognostication, and treatment. A multidisciplinary team of clinical experts with a central coordinating specialist for children with SLSMDS is recommended.


Asunto(s)
Síndromes Congénitos de Insuficiencia de la Médula Ósea/complicaciones , Síndrome de Kearns-Sayre/complicaciones , Errores Innatos del Metabolismo Lipídico/complicaciones , Enfermedades Mitocondriales/complicaciones , Enfermedades Musculares/complicaciones , Medición de Resultados Informados por el Paciente , Adolescente , Niño , Preescolar , Síndromes Congénitos de Insuficiencia de la Médula Ósea/diagnóstico , Síndromes Congénitos de Insuficiencia de la Médula Ósea/terapia , Femenino , Humanos , Lactante , Recién Nacido , Síndrome de Kearns-Sayre/diagnóstico , Síndrome de Kearns-Sayre/terapia , Errores Innatos del Metabolismo Lipídico/diagnóstico , Errores Innatos del Metabolismo Lipídico/terapia , Masculino , Enfermedades Mitocondriales/diagnóstico , Enfermedades Mitocondriales/terapia , Enfermedades Musculares/diagnóstico , Enfermedades Musculares/terapia
13.
J Pediatr Hematol Oncol ; 43(6): 232-235, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32815886

RESUMEN

Thrombocytopenia-absent radius (TAR) syndrome is a rare inherited bone marrow failure syndrome not generally associated with acute leukemia. The authors report a case of T-cell acute lymphoblastic leukemia in an adult female individual newly diagnosed with TAR syndrome. A 347-kb microdeletion of chromosome 1q21.1 involving the RBM8A gene was detected within a gain of whole chromosome 1. Next-generation sequencing on fibroblasts confirmed germline heterozygous deletion of RBM8A but on the other allele, noncoding low-frequency regulatory single-nucleotide polymorphism of RBM8A (rs139428292; rs201779890) were not found. The tolerance of the treatment was unusual and mostly marked by a slow hematopoietic recovery leading to a 6-month delay at the beginning of the maintenance phase. Only 5 cases of acute leukemia were reported in patients with TAR syndrome in the literature: 4 acute myeloid leukemia and one B-cell acute lymphoblastic leukemia. This is the first report of T-cell acute lymphoid leukemia occurring in the context of TAR syndrome.


Asunto(s)
Síndromes Congénitos de Insuficiencia de la Médula Ósea/complicaciones , Leucemia-Linfoma Linfoblástico de Células T Precursoras/complicaciones , Trombocitopenia/complicaciones , Deformidades Congénitas de las Extremidades Superiores/complicaciones , Adulto , Deleción Cromosómica , Síndromes Congénitos de Insuficiencia de la Médula Ósea/genética , Síndromes Congénitos de Insuficiencia de la Médula Ósea/terapia , Femenino , Humanos , Polimorfismo de Nucleótido Simple , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/terapia , Proteínas de Unión al ARN/genética , Radio (Anatomía) , Trombocitopenia/genética , Trombocitopenia/terapia , Deformidades Congénitas de las Extremidades Superiores/genética , Deformidades Congénitas de las Extremidades Superiores/terapia , Adulto Joven
14.
Int J Mol Sci ; 21(7)2020 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-32276429

RESUMEN

Patients with very long-chain acyl-CoA dehydrogenase deficiency (VLCADD) can present with life-threatening cardiac arrhythmias. The pathophysiological mechanism is unknown. We reprogrammed fibroblasts from one mildly and one severely affected VLCADD patient, into human induced pluripotent stem cells (hiPSCs) and differentiated these into cardiomyocytes (VLCADD-CMs). VLCADD-CMs displayed shorter action potentials (APs), more delayed afterdepolarizations (DADs) and higher systolic and diastolic intracellular Ca2+ concentration ([Ca2+]i) than control CMs. The mitochondrial booster resveratrol mitigated the biochemical, electrophysiological and [Ca2+]i changes in the mild but not in the severe VLCADD-CMs. Accumulation of potentially toxic intermediates of fatty acid oxidation was blocked by substrate reduction with etomoxir. Incubation with etomoxir led to marked prolongation of AP duration and reduced DADs and [Ca2+]i in both VLCADD-CMs. These results provide compelling evidence that reduced accumulation of fatty acid oxidation intermediates, either by enhanced fatty acid oxidation flux through increased mitochondria biogenesis (resveratrol) or by inhibition of fatty acid transport into the mitochondria (etomoxir), rescues pro-arrhythmia defects in VLCADD-CMs and open doors for new treatments.


Asunto(s)
Acil-CoA Deshidrogenasa de Cadena Larga/deficiencia , Arritmias Cardíacas/prevención & control , Síndromes Congénitos de Insuficiencia de la Médula Ósea/fisiopatología , Compuestos Epoxi/farmacología , Ácidos Grasos/química , Errores Innatos del Metabolismo Lipídico/fisiopatología , Mitocondrias/fisiología , Enfermedades Mitocondriales/fisiopatología , Enfermedades Musculares/fisiopatología , Miocitos Cardíacos/fisiología , Resveratrol/farmacología , Potenciales de Acción , Arritmias Cardíacas/etiología , Electrofisiología Cardíaca , Síndromes Congénitos de Insuficiencia de la Médula Ósea/complicaciones , Ácidos Grasos/metabolismo , Humanos , Células Madre Pluripotentes Inducidas , Errores Innatos del Metabolismo Lipídico/complicaciones , Enfermedades Mitocondriales/complicaciones , Enfermedades Musculares/complicaciones , Miocitos Cardíacos/efectos de los fármacos , Oxidación-Reducción
16.
Proteomics Clin Appl ; 14(3): e1900058, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32026584

RESUMEN

PURPOSE: To decipher the underlying immunological mechanisms in predisposition to oral microbial dysbiosis in severe congenital neutropenia (SCN) patients. EXPERIMENTAL DESIGN: Ten SCN patients (5-23 years old) and 12 healthy controls (5-22 years old) are periodontally examined and provided saliva, subgingival plaque, and gingival crevicular fluid (GCF) samples. The SCN patients received oral hygiene therapy and are re-evaluated after 6 months. Antimicrobial peptides HPN1-3 and LL-37 are assessed in saliva by ELISA. Concentration of 30 cytokines is measured in saliva and GCF by human 30-plex panel, while bacterial profiles of saliva and subgingival plaque are assessed using 16S rDNA amplicon sequencing. RESULTS: There is no significant difference in salivary HPN1-3 and LL-37 concentration between the SCN patients and controls. At baseline, clinical, immunological, and microbiological parameters of the patients are indicative of oral ecological dysbiosis. The SCN patients have significantly higher bleeding on probing (BOP)%, GCF volume, and cytokine levels, high bacterial load with low bacterial diversity in saliva. The associations between the microbiome and immunological parameters in the SCN patients differ from those in the healthy individuals. CONCLUSIONS AND CLINICAL RELEVANCE: SCN patients have a dysregulated immune response toward commensal oral microbiota, which could be responsible for the observed clinical and microbiological signs of dysbiosis.


Asunto(s)
Síndromes Congénitos de Insuficiencia de la Médula Ósea/complicaciones , Disbiosis/complicaciones , Neutropenia/congénito , Adolescente , Estudios de Casos y Controles , Niño , Citocinas/metabolismo , Disbiosis/inmunología , Disbiosis/metabolismo , Disbiosis/microbiología , Femenino , Humanos , Masculino , Microbiota , Boca/microbiología , Neutropenia/complicaciones , Proteómica , Saliva/metabolismo , Saliva/microbiología , Adulto Joven
17.
BMC Med Genet ; 21(1): 35, 2020 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-32066420

RESUMEN

BACKGROUND: We report a large family with four successive generations, presenting with a complex phenotype of severe congenital neutropenia (SCN), partially penetrant monocytosis, and hearing loss of varying severity. METHODS: We performed whole exome sequencing to identify the causative variants. Sanger sequencing was used to perform segregation analyses on remaining family members. RESULTS: We identified and classified a pathogenic GFI1 variant and a likely pathogenic variant in MYO6 which together explain the complex phenotypes seen in this family. CONCLUSIONS: We present a case illustrating the benefits of a broad screening approach that allows identification of oligogenic determinants of complex human phenotypes which may have been missed if the screening was limited to a targeted gene panel with the assumption of a syndromic disorder. This is important for correct genetic diagnosis of families and disentangling the range and severity of phenotypes associated with high impact variants.


Asunto(s)
Síndromes Congénitos de Insuficiencia de la Médula Ósea/genética , Proteínas de Unión al ADN/genética , Pérdida Auditiva Sensorineural/genética , Cadenas Pesadas de Miosina/genética , Neutropenia/congénito , Factores de Transcripción/genética , Adulto , Anciano , Síndromes Congénitos de Insuficiencia de la Médula Ósea/complicaciones , Síndromes Congénitos de Insuficiencia de la Médula Ósea/diagnóstico , Síndromes Congénitos de Insuficiencia de la Médula Ósea/fisiopatología , Exoma/genética , Femenino , Enfermedades Genéticas Congénitas/complicaciones , Enfermedades Genéticas Congénitas/diagnóstico , Enfermedades Genéticas Congénitas/genética , Enfermedades Genéticas Congénitas/fisiopatología , Pérdida Auditiva Sensorineural/complicaciones , Pérdida Auditiva Sensorineural/diagnóstico , Pérdida Auditiva Sensorineural/patología , Humanos , Masculino , Persona de Mediana Edad , Mutación/genética , Neutropenia/complicaciones , Neutropenia/diagnóstico , Neutropenia/genética , Neutropenia/fisiopatología , Linaje , Fenotipo , Secuenciación del Exoma
18.
Mutagenesis ; 35(5): 381-389, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33511998

RESUMEN

Severe congenital neutropenia (SCN) is a rare blood disorder characterised by abnormally low levels of circulating neutrophils. The most common recurrent mutations that cause SCN involve neutrophil elastase (ELANE). The treatment of choice for SCN is the administration of granulocyte-colony stimulating factor (G-CSF), which increases the neutrophil number and improves the survival and quality of life. Long-term survival is however linked to the development of myelodysplastic syndrome/acute myeloid leukemia (MDS/AML). About 70% of MDS/AML patients acquire nonsense mutations affecting the cytoplasmic domain of CSF3R (the G-CSF receptor). About 70% of SCN patients with AML harbour additional mutations in RUNX1. We hypothesised that this coding region of CSF3R constitutes a hotspot vulnerable to mutations resulting from excessive oxidative stress or endoplasmic reticulum (ER) stress. We used the murine Ba/F3 cell line to measure the effect of induced oxidative or ER stress on the mutation rate in our hypothesised hotspot of the exogenous human CSF3R, the corresponding region in the endogenous Csf3r, and Runx1. Ba/F3 cells transduced with the cDNA for partial C-terminal of CSF3R fused in-frame with a green fluorescent protein (GFP) tag were subjected to stress-inducing treatment for 30 days (~51 doubling times). The amplicon-based targeted deep sequencing data for days 15 and 30 samples show that although there was increased mutagenesis observed in all the three genes of interest (partial CSF3R, Csf3r and Runx1), there were more mutations in the GFP region compared with the partial CSF3R region. Our findings also indicate that there is no correlation between the stress-inducing chemical treatments and mutagenesis in Ba/F3 cells. Our data suggest that oxidative or ER stress induction does not promote genomic instability, affecting partial C-terminal of the transduced CSF3R, the endogenous Csf3R and the endogenous Runx1 in Ba/F3 cells that could account for these targets to being mutational hotspots. We conclude that other mechanisms to acquire mutations of CSF3R that help drive the evolution of SCN to MDS/AML.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Deficiencia GATA2/tratamiento farmacológico , Receptores del Factor Estimulante de Colonias/genética , Animales , Síndromes Congénitos de Insuficiencia de la Médula Ósea/complicaciones , Síndromes Congénitos de Insuficiencia de la Médula Ósea/tratamiento farmacológico , Síndromes Congénitos de Insuficiencia de la Médula Ósea/genética , Síndromes Congénitos de Insuficiencia de la Médula Ósea/patología , Subunidad alfa 2 del Factor de Unión al Sitio Principal/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Deficiencia GATA2/complicaciones , Deficiencia GATA2/genética , Deficiencia GATA2/patología , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Factor Estimulante de Colonias de Granulocitos/farmacología , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Ratones , Mutagénesis/efectos de los fármacos , Mutagénesis/genética , Mutación/efectos de los fármacos , Tasa de Mutación , Neutropenia/complicaciones , Neutropenia/congénito , Neutropenia/tratamiento farmacológico , Neutropenia/genética , Neutropenia/patología , Neutrófilos/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Receptores del Factor Estimulante de Colonias/administración & dosificación , Transducción de Señal , Respuesta de Proteína Desplegada/efectos de los fármacos
19.
Ann Noninvasive Electrocardiol ; 25(1): e12681, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31475425

RESUMEN

Pearson marrow-pancreas syndrome (PS), an exceedingly rare mitochondrial disorder, involves multiple systems including hematologic system and pancreas. Other mitochondrial disorders have been associated with progressive infrahisian block but this has not yet been described as a major feature of PS. We report a 7-year-old girl with classical features of PS and cardiac conduction defect. Her electrocardiogram revealed QRS prolongation with right bundle and left anterior fascicular blocks. Follow-up Holter revealed bifascicular block, alternating left and right bundle branch blocks, supraventricular tachycardia (with alternating bundles), and suspicion for nonsustained ventricular tachycardia. She underwent successful transvenous single-chamber ventricular pacemaker.


Asunto(s)
Trastorno del Sistema de Conducción Cardíaco/complicaciones , Trastorno del Sistema de Conducción Cardíaco/diagnóstico , Trastorno del Sistema de Conducción Cardíaco/terapia , Síndromes Congénitos de Insuficiencia de la Médula Ósea/complicaciones , Electrocardiografía/métodos , Sistema de Conducción Cardíaco/fisiopatología , Errores Innatos del Metabolismo Lipídico/complicaciones , Enfermedades Mitocondriales/complicaciones , Enfermedades Musculares/complicaciones , Marcapaso Artificial , Niño , Síndromes Congénitos de Insuficiencia de la Médula Ósea/fisiopatología , Femenino , Humanos , Errores Innatos del Metabolismo Lipídico/fisiopatología , Enfermedades Mitocondriales/fisiopatología , Enfermedades Musculares/fisiopatología
20.
J Crohns Colitis ; 14(1): 142-147, 2020 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-31157858

RESUMEN

Mendelian disorders in glucose-6-phosphate metabolism can present with inflammatory bowel disease [IBD]. Using whole genome sequencing we identified a homozygous variant in the glucose-6-phosphatase G6PC3 gene [c.911dupC; p.Q305fs*82] in an adult patient with congenital neutropenia, lymphopenia and childhood-onset, therapy-refractory Crohn's disease. Because G6PC3 is expressed in several haematopoietic and non-haematopoietic cells it was unclear whether allogeneic stem cell transplantation [HSCT] would benefit this patient with intestinal inflammation. We show that HSCT resolves G6PC3-associated immunodeficiency and the Crohn's disease phenotype. It illustrates how even in adulthood, next-generation sequencing can have a significant impact on clinical practice and healthcare utilization in patients with immunodeficiency and monogenic IBD.


Asunto(s)
Síndromes Congénitos de Insuficiencia de la Médula Ósea/complicaciones , Enfermedad de Crohn/terapia , Enfermedad del Almacenamiento de Glucógeno Tipo I/complicaciones , Trasplante de Células Madre Hematopoyéticas , Neutropenia/congénito , Enfermedad de Crohn/diagnóstico , Enfermedad de Crohn/etiología , Humanos , Masculino , Neutropenia/complicaciones , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA