Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 168
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
EMBO Rep ; 2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38816514

RESUMEN

ATP2B1 is a known regulator of calcium (Ca2+) cellular export and homeostasis. Diminished levels of intracellular Ca2+ content have been suggested to impair SARS-CoV-2 replication. Here, we demonstrate that a nontoxic caloxin-derivative compound (PI-7) reduces intracellular Ca2+ levels and impairs SARS-CoV-2 infection. Furthermore, a rare homozygous intronic variant of ATP2B1 is shown to be associated with the severity of COVID-19. The mechanism of action during SARS-CoV-2 infection involves the PI3K/Akt signaling pathway activation, inactivation of FOXO3 transcription factor function, and subsequent transcriptional inhibition of the membrane and reticulum Ca2+ pumps ATP2B1 and ATP2A1, respectively. The pharmacological action of compound PI-7 on sustaining both ATP2B1 and ATP2A1 expression reduces the intracellular cytoplasmic Ca2+ pool and thus negatively influences SARS-CoV-2 replication and propagation. As compound PI-7 lacks toxicity in vitro, its prophylactic use as a therapeutic agent against COVID-19 is envisioned here.

2.
Am J Hematol ; 2024 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-38666530

RESUMEN

Congenital Dyserythropoietic Anemia type I (CDA I) is a rare hereditary condition characterized by macrocytic/normocytic anemia, splenomegaly, iron overload, and distinct abnormalities during late erythropoiesis, particularly internuclear bridges between erythroblasts. Diagnosis of CDA I remains challenging due to its rarity, clinical heterogeneity, and overlapping phenotype with other rare hereditary anemias. In this case series, we present 36 patients with suspected CDA I. A molecular diagnosis was successfully established in 89% of cases, identifying 16 patients with CDA I through the presence of 18 causative variants in the CDAN1 or CDIN1 genes. Transcriptomic analysis of CDIN1 variants revealed impaired erythroid differentiation and disruptions in transcription, cell proliferation, and histone regulation. Conversely, 16 individuals received a different diagnosis, primarily pyruvate kinase deficiency. Comparisons between CDA I and non-CDA I patients revealed no significant differences in erythroblast morphological features. However, hemoglobin levels and red blood cell count differed between the two groups, with non-CDA I subjects being more severely affected. Notably, most patients with severe anemia belonged to the non-CDA I group (82% non-CDA I vs. 18% CDA I), with a subsequent absolute prevalence of transfusion dependency among non-CDA I patients (100% vs. 41.7%). All patients exhibited reduced bone marrow responsiveness to anemia, with a more pronounced effect observed in non-CDA I patients. Erythropoietin levels were significantly higher in non-CDA I patients compared to CDA I patients. However, evaluations of erythroferrone, soluble transferrin receptor, and hepcidin revealed no significant differences in plasma concentration between the two groups.

3.
Acta Radiol ; 65(1): 3-13, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36744376

RESUMEN

BACKGROUND: Low-dose thoracic protocols were developed massively during the COVID-19 outbreak. PURPOSE: To study the impact on image quality (IQ) and the diagnosis reliability of COVID-19 low-dose chest computed tomography (CT) protocols. MATERIAL AND METHODS: COVID-19 low-dose protocols were implemented on third- and second-generation CT scanners considering two body mass index (BMI) subgroups (<25 kg/m2 and >25 kg/m2). Contrast-to-noise ratios (CNR) were compared with a Catphan phantom. Next, two radiologists retrospectively assessed IQ for 243 CT patients using a 5-point Linkert scale for general IQ and diagnostic criteria. Kappa score and Wilcoxon rank sum tests were used to compare IQ score and CTDIvol between radiologists, protocols, and scanner models. RESULTS: In vitro analysis of Catphan inserts showed in majority significantly decreased CNR for the low dose versus standard acquisition protocols on both CT scanners. However, in vivo, there was no impact on the diagnosis: sensitivity and specificity were ≥0.8 for all protocols and CT scanners. The third-generation scanner involved a significantly lower dose compared to the second-generation scanner (CTDIvol of 1.8 vs. 2.6 mGy for BMI <25 kg/m2 and 3.3 vs. 4.6 mGy for BMI >25 kg/m2). Still, the third-generation scanner showed a significantly higher IQ with the low-dose protocol compared to the second-generation scanner (30.9 vs. 28.1 for BMI <25 kg/m2 and 29.9 vs. 27.8 for BMI >25 kg/m2). Finally, the two radiologists had good global inter-reader agreement (kappa ≥0.6) for general IQ. CONCLUSION: Low-dose protocols provided sufficient IQ independently of BMI subgroups and CT models without any impact on diagnosis reliability.


Asunto(s)
COVID-19 , Humanos , Reproducibilidad de los Resultados , Estudios Retrospectivos , Dosis de Radiación , Tomografía Computarizada por Rayos X/métodos
4.
Blood Cells Mol Dis ; 103: 102779, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37558589

RESUMEN

BACKGROUND: Reaching a precise diagnosis in rare inherited anemia is extremely difficult and challenging, especially in areas with limited use of genetic studies, which makes undiagnosed anemia a unique clinical entity in tertiary hematology centers. In this study, we aim at plotting a stepwise diagnostic approach in children with undiagnosed anemia while identifying indications for genetic testing. PATIENTS AND METHODS: A one-year cross-sectional study involved 44 children and adolescents with undiagnosed anemia after undergoing an initial routine panel of investigations. They were classified based on mean corpuscular volume (MCV) into 3 groups: microcytic (n = 19), normocytic (n = 14) and macrocytic (n = 11). An algorithm that included four levels of investigations was devised for each category. RESULTS: After applying a systematic diagnostic approach, 33 patients (75 %) were diagnosed of whom 7 (15 %) had combined diagnoses, while 11 (25 %) patients remained undiagnosed. Based on the first, second, third and fourth levels of investigations, patients were diagnosed, respectively, as follows: of the 11 patients, 7 were microcytic, 3 normocytic and 1 macrocytic; of the 7 patients, 2 were microcytic, 2 normocytic, and 3 macrocytic; of 10 patients, 5 were microcytic, 4 normocytic and 1 macrocytic; finally, of the 16 patients, 8 were microcytic, 6 normocytic and 2 macrocytic. Numbers recorded appear higher than the actual number of the patients because some of them were diagnosed by more than one level of investigation. The diagnoses obtained in the microcytic group showed hemoglobinopathies, iron refractory iron deficiency anemia (IRIDA), membrane defects, sideroblastic anemia, hypo-transferrinemia, a combined diagnosis of sickle cell trait and pyropoikilocytosis. The diagnoses also showed a combined diagnosis of hereditary spherocytosis (HS) and alpha thalassemia minor, and a combined diagnosis of iron deficiency anemia and beta thalassemia minor, while 15 % remained undiagnosed. In the normocytic group, the diagnosis revealed autosomal recessive (AR) HS, vitamin B12 deficiency, pyruvate kinase deficiency (PKD), congenital dyserythropoietic anemia (CDA) type I, Diamond Blackfan anemia and beta thalassemia major. In addition, it showed a combined diagnosis of AR HS and CDA type II, a combined diagnosis of AR HS and PKD, and a combined diagnosis of dehydrated stomatocytosis (DHS) and G6PD carrier, meanwhile 20 % remained undiagnosed. Finally, the macrocytic group was diagnosed by vitamin B12 deficiency, sideroblastic anemia, PKD, a combined diagnosis of PKD and G6PD deficiency carrier, while 45 % remained undiagnosed. CONCLUSION: Conducting a stepwise approach with different levels of investigations may help reach the diagnosis of difficult anemia without having to resort to unnecessary investigations. Combined diagnosis is an important cause of undiagnosed anemia, especially in countries with high frequency of consanguinity. The remaining 25 % of the patients continued to be undiagnosed, requiring more sophisticated investigations.


Asunto(s)
Anemia Ferropénica , Anemia Sideroblástica , Deficiencia de Vitamina B 12 , Talasemia beta , Adolescente , Humanos , Niño , Anemia Ferropénica/diagnóstico , Estudios Transversales , Países en Desarrollo
6.
Int J Mol Sci ; 24(3)2023 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-36769026

RESUMEN

Autism spectrum disorder (ASD) is a heterogeneous collection of neurodevelopmental disorders, difficult to diagnose and currently lacking treatment options. The possibility of finding reliable biomarkers useful for early identification would offer the opportunity to intervene with treatment strategies to improve the life quality of ASD patients. To date, there are many recognized risk factors for the development of ASD, both genetic and non-genetic. Although genetic and epigenetic factors may play a critical role, the extent of their contribution to ASD risk is still under study. On the other hand, non-genetic risk factors include pollution, nutrition, infection, psychological states, and lifestyle, all together known as the exposome, which impacts the mother's and fetus's life, especially during pregnancy. Pathogenic and non-pathogenic maternal immune activation (MIA) and autoimmune diseases can cause various alterations in the fetal environment, also contributing to the etiology of ASD in offspring. Activation of monocytes, macrophages, mast cells and microglia and high production of pro-inflammatory cytokines are indeed the cause of neuroinflammation, and the latter is involved in ASD's onset and development. In this review, we focused on non-genetic risk factors, especially on the connection between inflammation, macrophage polarization and ASD syndrome, MIA, and the involvement of microglia.


Asunto(s)
Trastorno del Espectro Autista , Efectos Tardíos de la Exposición Prenatal , Embarazo , Femenino , Humanos , Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/patología , Microglía/patología , Efectos Tardíos de la Exposición Prenatal/patología , Inflamación/patología , Macrófagos/patología
7.
Genet Med ; 24(8): 1653-1663, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35511137

RESUMEN

PURPOSE: Emerging evidence suggest that infection-dependent hyperactivation of complement system (CS) may worsen COVID-19 outcome. We investigated the role of predicted high impact rare variants - referred as qualifying variants (QVs) - of CS genes in predisposing asymptomatic COVID-19 in elderly individuals, known to be more susceptible to severe disease. METHODS: Exploiting exome sequencing data and 56 CS genes, we performed a gene-based collapsing test between 164 asymptomatic subjects (aged ≥60 years) and 56,885 European individuals from the Genome Aggregation Database. We replicated this test comparing the same asymptomatic individuals with 147 hospitalized patients with COVID-19. RESULTS: We found an enrichment of QVs in 3 genes (MASP1, COLEC11, and COLEC10), which belong to the lectin pathway, in the asymptomatic cohort. Analyses of complement activity in serum showed decreased activity of lectin pathway in asymptomatic individuals with QVs. Finally, we found allelic variants associated with asymptomatic COVID-19 phenotype and with a decreased expression of MASP1, COLEC11, and COLEC10 in lung tissue. CONCLUSION: This study suggests that genetic rare variants can protect from severe COVID-19 by mitigating the activity of lectin pathway and prothrombin. The genetic data obtained through ES of 786 asymptomatic and 147 hospitalized individuals are publicly available at http://espocovid.ceinge.unina.it/.


Asunto(s)
COVID-19 , Anciano , COVID-19/genética , Colectinas/genética , Colectinas/metabolismo , Células Germinativas , Humanos , Lectinas/genética , SARS-CoV-2 , Secuenciación del Exoma
8.
Blood ; 136(11): 1274-1283, 2020 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-32702750

RESUMEN

Congenital dyserythropoietic anemias (CDAs) are a heterogeneous group of inherited anemias that affect the normal differentiation-proliferation pathways of the erythroid lineage. They belong to the wide group of ineffective erythropoiesis conditions that mainly result in monolinear cytopenia. CDAs are classified into the 3 major types (I, II, III), plus the transcription factor-related CDAs, and the CDA variants, on the basis of the distinctive morphological, clinical, and genetic features. Next-generation sequencing has revolutionized the field of diagnosis of and research into CDAs, with reduced time to diagnosis, and ameliorated differential diagnosis in terms of identification of new causative/modifier genes and polygenic conditions. The main improvements regarding CDAs have been in the study of iron metabolism in CDAII. The erythroblast-derived hormone erythroferrone specifically inhibits hepcidin production, and its role in the mediation of hepatic iron overload has been dissected out. We discuss here the most recent advances in this field regarding the molecular genetics and pathogenic mechanisms of CDAs, through an analysis of the clinical and molecular classifications, and the complications and clinical management of patients. We summarize also the main cellular and animal models developed to date and the possible future therapies.


Asunto(s)
Anemia Diseritropoyética Congénita/genética , Anemia Diseritropoyética Congénita/clasificación , Anemia Diseritropoyética Congénita/diagnóstico , Anemia Diseritropoyética Congénita/terapia , Animales , Transfusión Sanguínea , Diagnóstico Diferencial , Modelos Animales de Enfermedad , Heterogeneidad Genética , Glicoproteínas/genética , Glicoproteínas/fisiología , Trasplante de Células Madre Hematopoyéticas , Hepcidinas , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Hidropesía Fetal/diagnóstico , Hidropesía Fetal/etiología , Sobrecarga de Hierro/etiología , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/fisiología , Ratones , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/fisiología , Técnicas de Diagnóstico Molecular , Proteínas Nucleares/genética , Proteínas Nucleares/fisiología , Hormonas Peptídicas/fisiología , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/fisiología , Pez Cebra
9.
Int J Mol Sci ; 23(19)2022 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-36233227

RESUMEN

Enterococcal bloodstream infections (EBSI) caused by vancomycin-resistant enterococci (VRE) are associated with a significant rate of unfavorable outcomes. No definitive data have been reported about the association between delayed antibiotic therapy and mortality. In this prospective observational study in three large hospitals in Italy (from August 2016 to April 2021), all consecutive hospitalized patients with a confirmed diagnosis of hospital-acquired monomicrobial BSI caused by VRE­with no evidence of endocarditis­were analyzed. Cox regression analysis showed that risk factors independently associated with 30-day mortality were age (HR 2.98, CI95% 1.44−6.81, p = 0.002), chronic kidney disease (HR 5.21, CI95% 1.48−22.23, p = 0.001), oncologic disease (HR 2.81, CI95% 1.45−19.8, p = 0.005), and intensive care unit admission (HR 3.71, CI95% 2.23−7.99, p < 0.001). Conversely, early effective therapy was associated with survival (HR 0.32, CI95% 0.38−0.76, p < 0.001). The administration of early effective antibiotic therapy within 48 h from blood culture collection was associated with 30-day mortality rates lower than 33%. Time from blood culture collection to appropriate therapy was an independent predictor of 30-day mortality in patients with EBSI caused by VRE. Based on these data, clinicians should start effective antibiotic therapy as soon as possible, preferably within the first 48 h from blood culture collection. Treatment strategies allowing the early delivery of in vitro active antibiotics are urgently needed, especially in critically ill patients at risk of VRE bacteremia.


Asunto(s)
Bacteriemia , Infecciones por Bacterias Grampositivas , Enterococos Resistentes a la Vancomicina , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Bacteriemia/tratamiento farmacológico , Infecciones por Bacterias Grampositivas/tratamiento farmacológico , Humanos , Estudios Retrospectivos , Vancomicina/farmacología , Vancomicina/uso terapéutico , Resistencia a la Vancomicina
10.
Int J Mol Sci ; 23(3)2022 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-35163229

RESUMEN

Biallelic pathogenic variants in the SEC23B gene cause congenital dyserythropoietic anemia type II (CDA II), a rare hereditary disorder hallmarked by ineffective erythropoiesis, hemolysis, erythroblast morphological abnormalities, and hypo-glycosylation of some red blood cell membrane proteins. Abnormalities in SEC23B, which encodes the homonymous cytoplasmic COPII (coat protein complex II) component, disturb the endoplasmic reticulum to Golgi trafficking and affect different glycosylation pathways. The most harmful complication of CDA II is the severe iron overload. Within our case series (28 CDA II patients), approximately 36% of them exhibit severe iron overload despite mild degree of anemia and slightly increased levels of ERFE (the only erythroid regulator of hepcidin suppression). Thus, we hypothesized a direct role of SEC23B loss-of-function in the pathomechanism of hepatic iron overload. We established a hepatic cell line, HuH7, stably silenced for SEC23B. In silenced cells, we observed significant alterations of the iron status, due to both the alteration in BMP/SMADs pathway effectors and a reduced capability to sense BMP6 stimulus. We demonstrated that the loss-of-function of SEC23B is responsible of the impairment in glycosylation of the membrane proteins involved in the activation of the BMP/SMADs pathway with subsequent hepcidin suppression. Most of these data were confirmed in another hepatic cell line, HepG2, stably silenced for SEC23B. Our findings suggested that the pathogenic mechanism of iron overload in CDA II is associated to both ineffective erythropoiesis and to a specific involvement of SEC23B pathogenic variants at hepatic level. Finally, we demonstrated the ability of SEC23B paralog, i.e., SEC23A, to rescue the hepcidin suppression, highlighting the functional overlap between the two SEC23 paralogs in human hepatic cells.


Asunto(s)
Hepatocitos/metabolismo , Hepcidinas/genética , Proteínas de Transporte Vesicular/metabolismo , Línea Celular , Retículo Endoplásmico/metabolismo , Eritropoyesis/genética , Glicosilación , Aparato de Golgi/metabolismo , Hepcidinas/metabolismo , Humanos , Sobrecarga de Hierro/genética , Sobrecarga de Hierro/metabolismo , Hígado/patología , Mutación con Pérdida de Función/genética , Fenotipo , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/fisiología
11.
Am J Hematol ; 96(12): 1666-1678, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34467556

RESUMEN

Methemoglobinemia is a rare disorder associated with oxidization of divalent ferro-iron of hemoglobin (Hb) to ferri-iron of methemoglobin (MetHb). Methemoglobinemia can result from either inherited or acquired processes. Acquired forms are the most common, mainly due to the exposure to substances that cause oxidation of the Hb both directly or indirectly. Inherited forms are due either to autosomal recessive variants in the CYB5R3 gene or to autosomal dominant variants in the globin genes, collectively known as HbM disease. Our recommendations are based on a systematic literature search. A series of questions regarding the key signs and symptoms, the methods for diagnosis, the clinical management in neonatal/childhood/adulthood period, and the therapeutic approach of methemoglobinemia were formulated and the relative recommendations were produced. An agreement was obtained using a Delphi-like approach and the experts panel reached a final consensus >75% of agreement for all the questions.


Asunto(s)
Metahemoglobinemia/diagnóstico , Metahemoglobinemia/terapia , Consenso , Diagnóstico Diferencial , Manejo de la Enfermedad , Humanos , Metahemoglobinemia/fisiopatología
12.
Nucleic Acids Res ; 47(20): 10728-10743, 2019 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-31584077

RESUMEN

Friedreich's ataxia (FRDA) is an untreatable disorder with neuro- and cardio-degenerative progression. This monogenic disease is caused by the hyper-expansion of naturally occurring GAA repeats in the first intron of the FXN gene, encoding for frataxin, a protein implicated in the biogenesis of iron-sulfur clusters. As the genetic defect interferes with FXN transcription, FRDA patients express a normal frataxin protein but at insufficient levels. Thus, current therapeutic strategies are mostly aimed to restore physiological FXN expression. We have previously described SINEUPs, natural and synthetic antisense long non-coding RNAs, which promote translation of partially overlapping mRNAs through the activity of an embedded SINEB2 domain. Here, by in vitro screening, we have identified a number of SINEUPs targeting human FXN mRNA and capable to up-regulate frataxin protein to physiological amounts acting at the post-transcriptional level. Furthermore, FXN-specific SINEUPs promote the recovery of disease-associated mitochondrial aconitase defects in FRDA-derived cells. In summary, we provide evidence that SINEUPs may be the first gene-specific therapeutic approach to activate FXN translation in FRDA and, more broadly, a novel scalable platform to develop new RNA-based therapies for haploinsufficient diseases.


Asunto(s)
Ataxia de Friedreich/genética , Regulación de la Expresión Génica , Proteínas de Unión a Hierro/genética , Modelos Biológicos , ARN no Traducido/metabolismo , Aconitato Hidratasa/metabolismo , Línea Celular , Fibroblastos/metabolismo , Humanos , Linfocitos/metabolismo , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN no Traducido/genética , Frataxina
13.
Int J Mol Sci ; 22(10)2021 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-34065289

RESUMEN

Genome-wide association studies (GWAS) found locus 3p21.31 associated with severe COVID-19. CCR5 resides at the same locus and, given its known biological role in other infection diseases, we investigated if common noncoding and rare coding variants, affecting CCR5, can predispose to severe COVID-19. We combined single nucleotide polymorphisms (SNPs) that met the suggestive significance level (P ≤ 1 × 10-5) at the 3p21.31 locus in public GWAS datasets (6406 COVID-19 hospitalized patients and 902,088 controls) with gene expression data from 208 lung tissues, Hi-C, and Chip-seq data. Through whole exome sequencing (WES), we explored rare coding variants in 147 severe COVID-19 patients. We identified three SNPs (rs9845542, rs12639314, and rs35951367) associated with severe COVID-19 whose risk alleles correlated with low CCR5 expression in lung tissues. The rs35951367 resided in a CTFC binding site that interacts with CCR5 gene in lung tissues and was confirmed to be associated with severe COVID-19 in two independent datasets. We also identified a rare coding variant (rs34418657) associated with the risk of developing severe COVID-19. Our results suggest a biological role of CCR5 in the progression of COVID-19 as common and rare genetic variants can increase the risk of developing severe COVID-19 by affecting the functions of CCR5.


Asunto(s)
COVID-19/genética , COVID-19/metabolismo , Predisposición Genética a la Enfermedad , Receptores CCR5/genética , Receptores CCR5/metabolismo , Alelos , Bronquios/metabolismo , Bronquios/patología , Bronquios/virología , COVID-19/fisiopatología , Cromosomas Humanos/genética , Estudios de Cohortes , Biología Computacional , Bases de Datos Genéticas , Estudio de Asociación del Genoma Completo , Genotipo , Humanos , Pulmón/metabolismo , Pulmón/patología , Pulmón/virología , Polimorfismo de Nucleótido Simple , Secuenciación del Exoma
14.
Int J Mol Sci ; 22(17)2021 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-34502454

RESUMEN

COVID-19 is a global threat that has spread since the end of 2019, causing severe clinical sequelae and deaths, in the context of a world pandemic. The infection of the highly pathogenetic and infectious SARS-CoV-2 coronavirus has been proven to exert systemic effects impacting the metabolism. Yet, the metabolic pathways involved in the pathophysiology and progression of COVID-19 are still unclear. Here, we present the results of a mass spectrometry-based targeted metabolomic analysis on a cohort of 52 hospitalized COVID-19 patients, classified according to disease severity as mild, moderate, and severe. Our analysis defines a clear signature of COVID-19 that includes increased serum levels of lactic acid in all the forms of the disease. Pathway analysis revealed dysregulation of energy production and amino acid metabolism. Globally, the variations found in the serum metabolome of COVID-19 patients may reflect a more complex systemic perturbation induced by SARS-CoV-2, possibly affecting carbon and nitrogen liver metabolism.


Asunto(s)
Biomarcadores/sangre , Carbono/metabolismo , Hígado/metabolismo , Metaboloma , Nitrógeno/metabolismo , Aminoácidos/metabolismo , COVID-19/sangre , COVID-19/patología , COVID-19/virología , Citocinas/sangre , Análisis Discriminante , Humanos , Análisis de los Mínimos Cuadrados , Redes y Vías Metabólicas/genética , Metabolómica/métodos , SARS-CoV-2/aislamiento & purificación , Índice de Severidad de la Enfermedad
15.
Am J Hematol ; 95(2): 188-197, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31737919

RESUMEN

Dehydrated hereditary stomatocytosis (DHS), or xerocytosis, is an autosomal dominant hemolytic anemia. Most patients with DHS carry mutations in the PIEZO1 gene encoding a mechanosensitive cation channel. We here demonstrate that patients with DHS have low levels of hepcidin and only a slight increase of ERFE, the erythroid negative regulator of hepcidin. We demonstrated that at the physiological level, PIEZO1 activation induced Ca2+ influx and suppression of HAMP expression in primary hepatocytes. In two hepatic cellular models expressing PIEZO1 WT and two PIEZO1 gain-of-function mutants (R2456H and R2488Q), we highlight altered expression of a few genes/proteins involved in iron metabolism. Mutant cells showed increased intracellular Ca2+ compared to WT, which was correlated to increased phosphorylation of ERK1/2, inhibition of the BMP-SMADs pathway, and suppression of HAMP transcription. Moreover, the HuH7 cells, treated with PD0325901, a potent inhibitor of ERK1/2 phosphorylation, reduced the phosphorylation of ERK1/2 with the consequent increased phosphorylation of SMAD1/5/8, confirming the link between the two pathways. Another "proof of concept" for the mechanism that links PIEZO1 to HAMP regulation was obtained by mimicking PIEZO1 activation by cell Ca2+ overload, by the Ca2+ ionophore A23187. There was strong down-regulation of HAMP gene expression after this Ca2+ overload. Finally, the inhibition of PIEZO1 by GsMTx4 leads to phenotype rescue. This is the first demonstration of a direct link between PIEZO1 and iron metabolism, which defines the channel as a new hepatic iron metabolism regulator and as a possible therapeutic target of iron overload in DHS and other iron-loading anemias.


Asunto(s)
Anemia Hemolítica Congénita , Proteínas Morfogenéticas Óseas/metabolismo , Mutación con Ganancia de Función , Hepcidinas/biosíntesis , Hidropesía Fetal , Canales Iónicos , Hierro/metabolismo , Hígado/metabolismo , Sistema de Señalización de MAP Quinasas , Proteínas Smad/metabolismo , Sustitución de Aminoácidos , Anemia Hemolítica Congénita/genética , Anemia Hemolítica Congénita/metabolismo , Anemia Hemolítica Congénita/patología , Benzamidas/farmacología , Proteínas Morfogenéticas Óseas/genética , Difenilamina/análogos & derivados , Difenilamina/farmacología , Regulación de la Expresión Génica , Células Hep G2 , Hepcidinas/genética , Humanos , Hidropesía Fetal/genética , Hidropesía Fetal/metabolismo , Hidropesía Fetal/patología , Canales Iónicos/genética , Canales Iónicos/metabolismo , Hígado/patología , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteínas Smad/genética
16.
J Pediatr Hematol Oncol ; 42(6): e507-e510, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-31593005

RESUMEN

Currently, there is no guideline for the treatment of patients with congenital dyserythropoietic anemia (CDA) type II. One approach is to follow-up patients with transfusions, on the basis of individually determined target hemoglobin levels, and iron chelation according to the thalassemia guidelines. In some transfusion-dependent CDA II patients, splenectomy reduces the number of transfusions; however, the only known curative option for CDA II patients is hematopoietic stem cell transplantation (HSCT). Only a few published case reports of allogeneic HSCT in CDA II patients are available. Here, we review the literature and add our data of a CDA II patient who developed transfusion dependence and was cured with HSCT.


Asunto(s)
Anemia Diseritropoyética Congénita/terapia , Trasplante de Células Madre Hematopoyéticas/métodos , Anemia Diseritropoyética Congénita/patología , Preescolar , Femenino , Humanos , Lactante , Pronóstico
17.
J Infect Chemother ; 26(8): 807-812, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32273175

RESUMEN

BACKGROUND: nursing home-acquired pneumonia (NHAP), is among the main causes of hospitalization and mortality of frail elderly patients. Aim of this study was analysis of patients residing in long-term care facilities (LTCF) and developing pneumonia to reach a better knowledge of criteria for hospitalization and outcomes. MATERIALS/METHODS: this is a prospective, observational study in which patients residing in 3 LTCFs (metropolitan area of Rome, Italy) and developing pneumonia, hospitalized or treated in LTCF, were recruited and followed up from January 2017 to June 2019. Primary endpoint was 30-day mortality, secondary endpoint was analysis of risk factors associated with hospitalization. RESULTS: Overall, 146 episodes of NHAP were enrolled in the study: 57 patients were treated in LTCF, while 89 patients were hospitalized. Overall incidence rates of NHAP varied from 2.6 to 7.5 per 1000 residents. Methicillin-resistant Staphylococcus aureus was the most frequently isolated pathogen (25%), and in 28 (55%) patients was documented a MDR pathogen. For hospitalized patients was reported a higher 30-day mortality (43.8% Vs 7%, p < 0.001). Multivariate analysis showed that severe pneumonia, neoplasm, chronic hepatitis, antibiotic monotherapy, and malnutrition were independent risk factors for hospitalization from LTCF. MDR pathogen, severe pneumonia, COPD, and moderate to severe renal disease were independently associated with death at 30 days. CONCLUSION: frail elderly patients in LTCF have a high risk of MDR etiology with a higher risk to receive an inadequate antibiotic therapy and a fatal outcome. These results point to the need for increased provision of acute care and strategies in LTCF.


Asunto(s)
Infección Hospitalaria/epidemiología , Casas de Salud/estadística & datos numéricos , Neumonía Bacteriana/epidemiología , Anciano , Anciano de 80 o más Años , Antibacterianos/uso terapéutico , Infección Hospitalaria/tratamiento farmacológico , Infección Hospitalaria/microbiología , Infección Hospitalaria/mortalidad , Farmacorresistencia Bacteriana Múltiple , Femenino , Hospitalización/estadística & datos numéricos , Humanos , Italia/epidemiología , Cuidados a Largo Plazo , Masculino , Desnutrición/epidemiología , Staphylococcus aureus Resistente a Meticilina/aislamiento & purificación , Neumonía Bacteriana/tratamiento farmacológico , Neumonía Bacteriana/mortalidad , Estudios Prospectivos , Factores de Riesgo , Streptococcus pneumoniae/aislamiento & purificación , Resultado del Tratamiento
18.
Int J Mol Sci ; 21(23)2020 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-33255852

RESUMEN

Macrophages are mononuclear cells that become osteoclasts (OCs) in the presence of two cytokines, macrophage colony-stimulating factor (M-CSF), and receptor activator of NF-κB ligand (RANKL). RANKL binding to its specific receptor RANK leads to OCs differentiation mainly by nuclear factor of activated T-cells cytoplasmic 1 (NFATc1). In our previous study, the analysis of the protein network in NFATc1-knockdown cells, using the Ingenuity Pathway Analysis (IPA), showed a link between NFATc1 and Mitogen-activated protein kinase kinase (MEK)-extracellular receptor kinase (ERK) signaling pathway. Therefore, this study aimed to extend our knowledge of the relationship between NFATc1 and the ERK. Here, we demonstrate that delayed ERK1/2 phosphorylation in pre-OC RANKL-induced depends on NFATc1. Indeed, the knockdown of NFATc1 reduced the phosphorylation of ERK1/2 (60%) and the pharmacological inhibition of the ERK1/2 kinase activity impairs the expression of NFATc1 without preventing its translocation into the nucleus. Furthermore, silencing of NFATc1 significantly reduced RANKL-induced migration (p < 0.01), and most pre-OCs are still mononuclear after 48 h (80 ± 5%), despite the presence of actin rings. On the other hand, the inhibitors FR180204 and PD98059 significantly reduced RANKL-induced cell migration (p < 0.01), leading to a reduction in the number of multinucleated cells. Finally, we suggest that long-lasting ERK activity depends on NFATc1 induction and is likely linked to cell migration, fusion, and OC differentiation.


Asunto(s)
Resorción Ósea/genética , Movimiento Celular/efectos de los fármacos , Factores de Transcripción NFATC/genética , Ligando RANK/genética , Animales , Resorción Ósea/patología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Movimiento Celular/genética , Flavonoides/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Factor Estimulante de Colonias de Macrófagos/genética , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Factores de Transcripción NFATC/antagonistas & inhibidores , Osteoclastos/efectos de los fármacos , Fosforilación/efectos de los fármacos , Pirazoles/farmacología , Piridazinas/farmacología , Células RAW 264.7
19.
Int J Mol Sci ; 21(15)2020 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-32759740

RESUMEN

Congenital dyserythropoietic anemia type II (CDA II) is a hypo-productive anemia defined by ineffective erythropoiesis through maturation arrest of erythroid precursors. CDA II is an autosomal recessive disorder due to loss-of-function mutations in SEC23B. Currently, management of patients with CDA II is based on transfusions, splenectomy, or hematopoietic stem-cell transplantation. Several studies have highlighted benefits of ACE-011 (sotatercept) treatment of ineffective erythropoiesis, which acts as a ligand trap against growth differentiation factor (GDF)11. Herein, we show that GDF11 levels are increased in CDA II, which suggests sotatercept as a targeted therapy for treatment of these patients. Treatment of stable clones of SEC23B-silenced erythroleukemia K562 cells with the iron-containing porphyrin hemin plus GDF11 increased expression of pSMAD2 and reduced nuclear localization of the transcription factor GATA1, with subsequent reduced gene expression of erythroid differentiation markers. We demonstrate that treatment of these SEC23B-silenced K562 cells with RAP-011, a "murinized" ortholog of sotatercept, rescues the disease phenotype by restoring gene expression of erythroid markers through inhibition of the phosphorylated SMAD2 pathway. Our data also demonstrate the effect of RAP-011 treatment in reducing the expression of erythroferrone in vitro, thus suggesting a possible beneficial role of the use of sotatercept in the management of iron overload in patients with CDA II.


Asunto(s)
Anemia Diseritropoyética Congénita/tratamiento farmacológico , Proteínas Morfogenéticas Óseas/genética , Factor de Transcripción GATA1/genética , Factores de Diferenciación de Crecimiento/genética , Proteínas Recombinantes de Fusión/farmacología , Proteínas de Transporte Vesicular/genética , Anemia Diseritropoyética Congénita/genética , Anemia Diseritropoyética Congénita/patología , Células Precursoras Eritroides/metabolismo , Eritropoyesis/genética , Femenino , Humanos , Células K562 , Mutación/genética , Fenotipo , Proteínas Recombinantes de Fusión/genética , Proteína Smad2/genética , Proteína smad3/genética
20.
Br J Haematol ; 187(1): 13-24, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31364155

RESUMEN

Hereditary erythrocyte membrane disorders are caused by mutations in genes encoding various transmembrane or cytoskeletal proteins of red blood cells. The main consequences of these genetic alterations are decreased cell deformability and shortened erythrocyte survival. Red blood cell membrane defects encompass a heterogeneous group of haemolytic anaemias caused by either (i) altered membrane structural organisation (hereditary spherocytosis, hereditary elliptocytosis, hereditary pyropoikilocytosis and Southeast Asian ovalocytosis) or (ii) altered membrane transport function (overhydrated hereditary stomatocytosis, dehydrated hereditary stomatocytosis or xerocytosis, familial pseudohyperkalaemia and cryohydrocytosis). Herein we provide a comprehensive review of the recent literature on the molecular genetics of erythrocyte membrane defects and their reported clinical consequences. We also describe the effect of low-expression genetic variants on the high inter- and intra-familial phenotype variability of erythrocyte structural defects.


Asunto(s)
Anemia Hemolítica Congénita/genética , Membrana Eritrocítica/química , Alelos , Anemia Hemolítica Congénita/sangre , Anemia Hemolítica Congénita/diagnóstico , Eliptocitosis Hereditaria/sangre , Eliptocitosis Hereditaria/diagnóstico , Eliptocitosis Hereditaria/genética , Eritrocitos Anormales/metabolismo , Humanos , Proteínas de la Membrana/sangre , Esferocitosis Hereditaria/sangre , Esferocitosis Hereditaria/diagnóstico , Esferocitosis Hereditaria/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA