Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
1.
Cancer ; 128(8): 1595-1604, 2022 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-35157306

RESUMEN

BACKGROUND: Ofatumumab is a humanized type 1 anti-CD20 monoclonal antibody. Preclinical studies show improved complement-mediated cytotoxicity (CMC) compared to rituximab in mantle cell lymphoma (MCL). This study evaluates the safety and efficacy of combining ofatumumab with HyperCVAD/MA (O-HyperCVAD) in newly diagnosed MCL. METHODS: In this single-arm phase 2 study, 37 patients were treated with the combination of O-HyperCVAD for 4 or 6 cycles, followed by high dose chemotherapy and autologous stem cell transplant. Primary objectives were overall response rate (ORR) and complete response (CR) rate at the end of therapy. Secondary objectives included minimal residual disease (MRD) negativity, progression-free survival (PFS), and overall survival (OS). RESULTS: Median age was 60 years; ORR was 86% and 73% achieved a CR by modified Cheson criteria. The MRD negativity rate was 78% after 2 cycles of therapy, increasing to 96% at the end of induction; median PFS and OS were 45.5 months and 56 months, respectively. Achieving a post-induction CR by both imaging and flow cytometry was associated with improved PFS and OS. Early MRD negativity (post-2 cycles) was also associated with an improved PFS but not OS. There were 3 deaths while on therapy, and grades 3 and 4 adverse events (AEs) were observed in 22% and 68% of the patients. CONCLUSION: The addition of ofatumumab to HyperCVAD/HD-MA led to high rates of MRD negativity by flow cytometry in patients with newly diagnosed MCL. Achieving a CR post-induction by both imaging and flow cytometry is associated with improved overall survival.


Asunto(s)
Anticuerpos Monoclonales Humanizados , Linfoma de Células del Manto , Adulto , Anticuerpos Monoclonales Humanizados/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Humanos , Linfoma de Células del Manto/terapia , Persona de Mediana Edad , Neoplasia Residual/diagnóstico , Rituximab
2.
Cell Mol Biol (Noisy-le-grand) ; 66(3): 221-229, 2020 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-32538775

RESUMEN

It can be misleading to think that the new severe acute respiratory syndrome coronavirus (SARS-CoV2) which has a very strong mutation and adaptation capabilities, uses only the angiotensin-converting enzyme II (ACE2) pathway to reach target cells. Despite all the precautions taken, the pandemic attack continues and the rapid increase in the number of deaths suggest that this virus has entered the cell through different pathways and caused damage through different mechanisms. The main reason why the ACE2 pathway comes to the fore in all scientific studies is that this receptor is located at the entry point of basic mechanisms that provide alveolo-capillary homeostasis. SARS-CoV-2 has to use nuclear factor-κB (NF-kB), caveloae, clathrin, lipoxin, serine protease and proteasome pathways in addition to ACE2 to enter the target cell and initiate damage. For this reason, while new drug development studies are continuing, in order to be beneficial to patients in their acute period, it is imperative that we are able to come up with drugs that activate or inhibit these pathways and are currently in clinical use. It is also critical that we adopt these new pathways to the treatment of pregnant women affected by SARS-CoV-2, based on the scientific data we use to treat the general population.


Asunto(s)
Betacoronavirus/metabolismo , Caveolina 1/metabolismo , Infecciones por Coronavirus/metabolismo , Lipoxinas/metabolismo , FN-kappa B/metabolismo , Neumonía Viral/metabolismo , Complicaciones Infecciosas del Embarazo/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Bloqueadores del Receptor Tipo 1 de Angiotensina II/uso terapéutico , Enzima Convertidora de Angiotensina 2 , Anticolesterolemiantes/uso terapéutico , Sitios de Unión , COVID-19 , Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/transmisión , Infecciones por Coronavirus/virología , Descubrimiento de Drogas/métodos , Reposicionamiento de Medicamentos/métodos , Femenino , Humanos , Transmisión Vertical de Enfermedad Infecciosa/prevención & control , FN-kappa B/antagonistas & inhibidores , Uso Fuera de lo Indicado , Pandemias , Peptidil-Dipeptidasa A/metabolismo , Neumonía Viral/tratamiento farmacológico , Neumonía Viral/transmisión , Neumonía Viral/virología , Embarazo , Complicaciones Infecciosas del Embarazo/virología , Inhibidores de Proteasoma/uso terapéutico , SARS-CoV-2 , Serina Endopeptidasas/metabolismo , Inhibidores de Serina Proteinasa/uso terapéutico , Internalización del Virus
3.
Hum Mol Genet ; 26(7): 1328-1339, 2017 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-28204537

RESUMEN

Mutations in mitochondrial complex II (succinate dehydrogenase; SDH) genes predispose to paraganglioma tumors that show constitutive activation of hypoxia responses. We recently showed that SDHB mRNAs in hypoxic monocytes gain a stop codon mutation by APOBEC3A-mediated C-to-U RNA editing. Here, we test the hypothesis that inhibition of complex II facilitates hypoxic gene expression in monocytes using an integrative experimental approach. By RNA sequencing, we show that specific inhibition of complex II by atpenin A5 in normoxic conditions mimics hypoxia and induces hypoxic transcripts as well as APOBEC3A-mediated RNA editing in human monocytes. Myxothiazol, a complex III inhibitor, has similar effects in normoxic monocytes. Atpenin A5 partially inhibits oxygen consumption, and neither hypoxia nor atpenin A5 in normoxia robustly stabilizes hypoxia-inducible factor (HIF)-1α in primary monocytes. Several earlier studies in transformed cell lines suggested that normoxic stabilization of HIF-1α explains the persistent expression of hypoxic genes upon complex II inactivation. On the contrary, we find that atpenin A5 antagonizes the stabilization of HIF-1α and reduces hypoxic gene expression in transformed cell lines. Accordingly, compound germline heterozygosity of mouse Sdhb/Sdhc/Sdhd null alleles blunts chronic hypoxia-induced increases in hemoglobin levels, an adaptive response mainly regulated by HIF-2α. In contrast, atpenin A5 or myxothiazol does not reduce hypoxia-induced gene expression or RNA editing in monocytes. These results reveal a novel role for mitochondrial respiratory inhibition in induction of the hypoxic transcriptome in monocytes and suggest that inhibition of complex II activates a distinct hypoxia signaling pathway in a cell-type specific manner.


Asunto(s)
Citidina Desaminasa/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/biosíntesis , Proteínas/genética , Succinato Deshidrogenasa/genética , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/biosíntesis , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Hipoxia de la Célula/genética , Citidina Desaminasa/metabolismo , Complejo II de Transporte de Electrones/genética , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Proteínas de la Membrana/genética , Metacrilatos/administración & dosificación , Ratones , Monocitos/metabolismo , Monocitos/patología , Consumo de Oxígeno/efectos de los fármacos , Proteínas/metabolismo , Piridonas/administración & dosificación , Edición de ARN/genética , Transducción de Señal/efectos de los fármacos , Tiazoles/administración & dosificación
4.
RNA Biol ; 14(5): 603-610, 2017 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-27149507

RESUMEN

APOBEC3A cytidine deaminase induces site-specific C-to-U RNA editing of hundreds of genes in monocytes exposed to hypoxia and/or interferons and in pro-inflammatory macrophages. To examine the impact of APOBEC3A overexpression, we transiently expressed APOBEC3A in HEK293T cell line and performed RNA sequencing. APOBEC3A overexpression induces C-to-U editing at more than 4,200 sites in transcripts of 3,078 genes resulting in protein recoding of 1,110 genes. We validate recoding RNA editing of genes associated with breast cancer, hematologic neoplasms, amyotrophic lateral sclerosis, Alzheimer disease and primary pulmonary hypertension. These results highlight the fundamental impact of APOBEC3A overexpression on human transcriptome by widespread RNA editing.


Asunto(s)
Citidina Desaminasa/metabolismo , Proteínas/metabolismo , Edición de ARN , ARN/metabolismo , Transcriptoma , Secuencia de Bases , Citidina Desaminasa/genética , Enfermedad/genética , Células HEK293 , Humanos , Hipoxia/metabolismo , Interferones/metabolismo , Macrófagos/metabolismo , Monocitos/metabolismo , Proteínas/genética , ARN/genética
5.
Arch Gynecol Obstet ; 296(4): 721-730, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28707059

RESUMEN

PURPOSE: To assess the predictive power of the cerebro-placental ratio (CPR) and the venous-arterial index (VAI) for the development of intrapartum fetal distress (FD) and neonatal intensive care unit (NICU) admission. METHODS: Fetal umbilical artery, middle cerebral artery and umbilical vein Doppler measurements were obtained before the active phase of labor in 311 singleton pregnancies at ≥37 weeks. A continuous electronic fetal monitorization was applied, and an umbilical cord blood sample was obtained for each participant. FD and NICU admission were the primary outcomes. RESULTS: Labor was concluded as uncomplicated spontaneous vaginal delivery (SVD) in 261 (83.9%) cases. The 22 (7.1%) FD cases were subdivided into FD with NICU admission (n: 7; 2.3%) and without NICU admission (n: 15; 4.8%). Six out of 7 (85.8%) FD with NICU admission cases were from nulliparous pregnancies. The combinatory indices (VAI and CPR) reached the highest sensitivity (31.8%) and negative predictive value (94.7%). None of the fetuses, distressed or non-distressed, with CPR ≤ 10th percentile was born with a cord pH < 7.20. CONCLUSION: FD frequency was increased in fetuses with a low CPR or low VAI. However, the Doppler patterns were heterogeneous in both subgroups: FD with and without NICU admission. FD seems to be a common endpoint of different circulatory-metabolic disturbances. Parity affects the FD frequency in a manner related but not limited to fetal arterial and venous circulation. Low CPR could be a part of the adaptive mechanisms providing metabolic preparedness for hypoxic episodes.


Asunto(s)
Sufrimiento Fetal/diagnóstico por imagen , Arteria Cerebral Media/diagnóstico por imagen , Ultrasonografía Doppler/métodos , Ultrasonografía Prenatal/métodos , Adulto , Parto Obstétrico/efectos adversos , Femenino , Sufrimiento Fetal/etiología , Feto/irrigación sanguínea , Feto/fisiopatología , Humanos , Recién Nacido , Recién Nacido Pequeño para la Edad Gestacional , Placenta/diagnóstico por imagen , Embarazo , Resultado del Embarazo , Estudios Prospectivos , Nacimiento a Término , Arterias Umbilicales/diagnóstico por imagen
6.
Biochim Biophys Acta ; 1827(5): 573-7, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23291190

RESUMEN

Germ line heterozygous mutations in the structural subunit genes of mitochondrial complex II (succinate dehydrogenase; SDH) and the regulatory gene SDHAF2 predispose to paraganglioma tumors which show constitutive activation of hypoxia inducible pathways. Mutations in SDHD and SDHAF2 cause highly penetrant multifocal tumor development after a paternal transmission, whereas maternal transmission rarely, if ever, leads to tumor development. This transmission pattern is consistent with genomic imprinting. Recent molecular evidence supports a model for tissue-specific imprinted regulation of the SDHD gene by a long range epigenetic mechanism. In addition, there is evidence of SDHB mRNA editing in peripheral blood mononuclear cells and long-term balancing selection operating on the SDHA gene. Regulation of SDH subunit expression by diverse epigenetic mechanisms implicates a crucial dosage-dependent role for SDH in oxygen homeostasis. This article is part of a Special Issue entitled: Respiratory complex II: Role in cellular physiology and disease.


Asunto(s)
Complejo II de Transporte de Electrones/genética , Impresión Genómica , Mutación , Paraganglioma/genética , Complejo II de Transporte de Electrones/metabolismo , Humanos , Mitocondrias/genética , Mitocondrias/metabolismo , Modelos Genéticos , Paraganglioma/metabolismo , Paraganglioma/patología , Succinato Deshidrogenasa/genética , Succinato Deshidrogenasa/metabolismo
7.
Commun Biol ; 7(1): 529, 2024 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-38704509

RESUMEN

Intra-organism biodiversity is thought to arise from epigenetic modification of constituent genes and post-translational modifications of translated proteins. Here, we show that post-transcriptional modifications, like RNA editing, may also contribute. RNA editing enzymes APOBEC3A and APOBEC3G catalyze the deamination of cytosine to uracil. RNAsee (RNA site editing evaluation) is a computational tool developed to predict the cytosines edited by these enzymes. We find that 4.5% of non-synonymous DNA single nucleotide polymorphisms that result in cytosine to uracil changes in RNA are probable sites for APOBEC3A/G RNA editing; the variant proteins created by such polymorphisms may also result from transient RNA editing. These polymorphisms are associated with over 20% of Medical Subject Headings across ten categories of disease, including nutritional and metabolic, neoplastic, cardiovascular, and nervous system diseases. Because RNA editing is transient and not organism-wide, future work is necessary to confirm the extent and effects of such editing in humans.


Asunto(s)
Desaminasas APOBEC , Citidina Desaminasa , Edición de ARN , Humanos , Citidina Desaminasa/metabolismo , Citidina Desaminasa/genética , Polimorfismo de Nucleótido Simple , Citosina/metabolismo , Desaminasa APOBEC-3G/metabolismo , Desaminasa APOBEC-3G/genética , Uracilo/metabolismo , Proteínas/genética , Proteínas/metabolismo , Citosina Desaminasa/genética , Citosina Desaminasa/metabolismo
8.
Hum Mol Genet ; 20(22): 4452-61, 2011 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-21862453

RESUMEN

Germline mutations in SDHD, a mitochondrial complex II (succinate dehydrogenase) subunit gene at chromosome band 11q23, cause highly penetrant paraganglioma (PGL) tumors when transmitted through fathers. In contrast, maternal transmission rarely, if ever, leads to tumor development. The mechanism underlying this unusual monogenic tumor predisposition pattern is poorly understood. Here, we describe identification of imprinted methylation within an alternative promoter for a large intergenic non-coding RNA located at a distant gene desert boundary flanking SDHD. Methylation at this site primarily occurs within two consecutive HpaII restriction enzyme sites in a tissue-specific manner, most commonly in the adrenal gland. Informative fetal tissues and PGL tumors demonstrate maternal allelic hypermethylation. While a strong binding site for the enhancer-blocking protein CTCF within the alternative promoter shows no evidence of methylation, hyper-methylated adrenal tissues show increased binding of the chromatin-looping factor cohesin relative to the hypo-methylated tissues. These results suggest that the differential allelic methylation we observe at this locus is associated with altered chromatin architectures. These results provide molecular evidence for imprinting at a boundary element flanking the SDHD locus and suggest that epigenetic suppression of the maternal allele is the underlying mechanism of the imprinted penetrance of SDHD mutations.


Asunto(s)
Impresión Genómica/genética , Elementos Aisladores/genética , Succinato Deshidrogenasa/genética , Inmunoprecipitación de Cromatina , Biología Computacional , Metilación de ADN/genética , Humanos , Técnicas In Vitro , Mutación , Polimorfismo Genético/genética
9.
Nutrients ; 15(8)2023 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-37111213

RESUMEN

The effects of maternal diet on the neuroimmune responses of the offspring remain to be elucidated. We investigated the impact of maternal ketogenic diet (KD) on the NLRP3 inflammasome response in the offspring's brain. C57BL/6 female mice were randomly allocated into standard diet (SD) and ketogenic diet (KD) groups for 30 days. After mating, the presence of sperm in the vaginal smear was considered day 0 of pregnancy, and female mice continued their respective diets during pregnancy and the lactation period. Following birth, pups were further allocated into two groups and given either LPS or intraperitoneal saline on postnatal (PN) days 4, 5 and 6; they were sacrificed on PN11 or PN21. Neuronal densities were significantly lower globally in the KD group when compared to the SD group at PN11. Neuronal density in the prefrontal cortex (PFC) and dentate gyrus (DG) regions were also significantly lower in the KD group when compared to the SD group at PN21. Following administration of LPS, the decrease in the neuronal count was more prominent in the SD group when compared to the KD group in the PFC and DG regions at PN11 and PN21. NLRP3 and IL-1ß were higher in the KD group than in the SD group at PN21 in the PFC, CA1 and DG regions, and were significantly lower in the DG region of the KD group especially when compared to the SD group following LPS. Results of our study reveal that maternal KD negatively affects the offspring's brain in the mouse model. The effects of KD exhibited regional variations. On the other hand, in the presence of KD exposure, NLRP3 expression after LPS injection was lower in the DG and CA1 areas but not in the PFC when compared to SD group. Further experimental and clinical studies are warranted to elucidate the molecular mechanisms underlying the impact of antenatal KD exposure and regional discrepancies on the developing brain.


Asunto(s)
Dieta Cetogénica , Inflamasomas , Femenino , Masculino , Ratones , Animales , Embarazo , Inflamasomas/metabolismo , Dieta Cetogénica/efectos adversos , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Lipopolisacáridos , Ratones Endogámicos C57BL , Semen , Encéfalo/metabolismo
10.
bioRxiv ; 2023 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-37577456

RESUMEN

Intra-organism biodiversity is thought to arise from epigenetic modification of our constituent genes and post-translational modifications after mRNA is translated into proteins. We have found that post-transcriptional modification, also known as RNA editing, is also responsible for a significant amount of our biodiversity, substantively expanding this story. The APOBEC (apolipoprotein B mRNA editing catalytic polypeptide-like) family RNA editing enzymes APOBEC3A and APOBEC3G catalyze the deamination of cytosines to uracils (C>U) in specific stem-loop structures.1,2 We used RNAsee (RNA site editing evaluation), a tool developed to predict the locations of APOBEC3A/G RNA editing sites, to determine whether known single nucleotide polymorphisms (SNPs) in DNA could be replicated in RNA via RNA editing. About 4.5% of non-synonymous SNPs which result in C>U changes in RNA, and about 5.4% of such SNPs labelled as pathogenic, were identified as probable sites for APOBEC3A/G editing. This suggests that the variant proteins created by these DNA mutations may also be created by transient RNA editing, with the potential to affect human health. Those SNPs identified as potential APOBEC3A/G-mediated RNA editing sites were disproportionately associated with cardiovascular diseases, digestive system diseases, and musculoskeletal diseases. Future work should focus on common sites of RNA editing, any variant proteins created by these RNA editing sites, and the effects of these variants on protein diversity and human health. Classically, our biodiversity is thought to come from our constitutive genetics, epigenetic phenomenon, transcriptional differences, and post-translational modification of proteins. Here, we have shown evidence that RNA editing, often stimulated by environmental factors, could account for a significant degree of the protein biodiversity leading to human disease. In an era where worries about our changing environment are ever increasing, from the warming of our climate to the emergence of new diseases to the infiltration of microplastics and pollutants into our bodies, understanding how environmentally sensitive mechanisms like RNA editing affect our own cells is essential.

11.
Case Rep Hematol ; 2022: 9785588, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35795544

RESUMEN

Chronic myeloid leukemia (CML) is a clonal hematopoietic stem cell disorder, characterized by reciprocal translocation t(9,22) (q34; q11), leading to increased myeloid proliferation. Most cases are diagnosed in the chronic phase (CP). However, a minority of cases can be present in the blastic phase (BP). In most patients with CML-BP, the blasts have a myeloid phenotype, however, in 20-30% of cases, the blasts have a lymphoid phenotype, mostly a B-cell phenotype. It is challenging to differentiate CML B-lymphoblastic phase (CML-BLP) from Ph + primary B-acute lymphoblastic leukemia (B-ALL) especially when the CML-BLP is the initial presentation of the disease, which is uncommon. We report here an unusual case of CML-BLP as an initial presentation of the disease without typical CML morphological findings. This case demonstrates diagnostic challenges and emphasizes the importance of an integrated approach using morphology, multiparametric flow cytometry, cytogenetic studies, and molecular studies to render an accurate diagnosis.

12.
JCI Insight ; 7(17)2022 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-35881479

RESUMEN

Increased red cell distribution width (RDW), which measures erythrocyte mean corpuscular volume (MCV) variability (anisocytosis), has been linked to early mortality in many diseases and in older adults through unknown mechanisms. Hypoxic stress has been proposed as a potential mechanism. However, experimental models to investigate the link between increased RDW and reduced survival are lacking. Here, we show that lifelong hypobaric hypoxia (~10% O2) increased erythrocyte numbers, hemoglobin, and RDW, while reducing longevity in male mice. Compound heterozygous knockout (hKO) mutations in succinate dehydrogenase (Sdh; mitochondrial complex II) genes Sdhb, Sdhc, and Sdhd reduced Sdh subunit protein levels, reduced RDW, and increased healthy life span compared with WT mice in chronic hypoxia. RDW-SD, a direct measure of MCV variability, and the SD of MCV showed the most statistically significant reductions in Sdh hKO mice. Tissue metabolomic profiling of 147 common metabolites showed the largest increase in succinate with elevated succinate/fumarate and succinate/oxoglutarate (2-ketoglutarate) ratios in Sdh hKO mice. These results demonstrate that mitochondrial complex II level is an underlying determinant of both RDW and healthy life span in hypoxia and suggest that therapeutic targeting of Sdh might reduce high RDW-associated clinical mortality in hypoxic diseases.


Asunto(s)
Índices de Eritrocitos , Succinato Deshidrogenasa , Animales , Hipoxia , Longevidad , Masculino , Ratones , Succinato Deshidrogenasa/genética , Succinato Deshidrogenasa/metabolismo , Succinatos
13.
Commun Biol ; 4(1): 102, 2021 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-33483601

RESUMEN

Pro-inflammatory M1 macrophage polarization is associated with microbicidal and antitumor responses. We recently described APOBEC3A-mediated cytosine-to-uracil (C > U) RNA editing during M1 polarization. However, the functional significance of this editing is unknown. Here we find that APOBEC3A-mediated cellular RNA editing can also be induced by influenza or Maraba virus infections in normal human macrophages, and by interferons in tumor-associated macrophages. Gene knockdown and RNA_Seq analyses show that APOBEC3A mediates C>U RNA editing of 209 exonic/UTR sites in 203 genes during M1 polarization. The highest level of nonsynonymous RNA editing alters a highly-conserved amino acid in THOC5, which encodes a nuclear mRNA export protein implicated in M-CSF-driven macrophage differentiation. Knockdown of APOBEC3A reduces IL6, IL23A and IL12B gene expression, CD86 surface protein expression, and TNF-α, IL-1ß and IL-6 cytokine secretion, and increases glycolysis. These results show a key role of APOBEC3A cytidine deaminase in transcriptomic and functional polarization of M1 macrophages.


Asunto(s)
Citidina Desaminasa/metabolismo , Macrófagos/metabolismo , Proteínas Nucleares/metabolismo , Proteínas/metabolismo , Edición de ARN , Humanos , Cultivo Primario de Células
14.
J Hum Genet ; 55(3): 182-5, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20111059

RESUMEN

Hereditary paraganglioma (PGL) is characterized by the development of highly vascularized paraganglionic tumors as a result of germline mutations in the SDHB, SDHC or SDHD subunit genes of succinate dehydrogenase (SDH; mitochondrial complex II), or in the Von Hippel-Lindau tumor-suppressor gene. Although many PGL mutations have been described, gross SDHD deletions have not yet been implicated as founder mutations and are rarely characterized at the DNA sequence level. We investigated the genetic basis of head and neck PGLs observed in 20 subjects from two unrelated multiplex pedigrees from Austria and identified a 4944-base pair partial SDHD deletion, which escaped PCR-based detection methods. The deletion occurred between Alu elements and was present within the same haplotype context in both pedigrees, indicating a founder effect. The deletion caused tumors only after a paternal transmission similar to other conventional SDHD mutations, suggesting preservation of genomic imprinting mechanisms operating at this locus. These data describe a large SDHD deletion at the genomic sequence level and indicate that gross SDHD deletions could be a founder PGL mutation in certain populations.


Asunto(s)
Elementos Alu/genética , Emparejamiento Base/genética , Efecto Fundador , Eliminación de Gen , Paraganglioma/enzimología , Paraganglioma/genética , Succinato Deshidrogenasa/genética , Austria , Secuencia de Bases , Análisis Mutacional de ADN , Exones/genética , Familia , Femenino , Haplotipos/genética , Humanos , Masculino , Datos de Secuencia Molecular , Linaje
15.
Neurosci Lett ; 738: 135389, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-32949661

RESUMEN

Despite widely known detrimental effects on the developing brain, supplemental oxygen is still irreplaceable in the management of newborn infants with respiratory distress. Identifying downstream mechanisms underlying oxygen toxicity is a key step for development of new neuroprotective strategies. Main purpose of this study is to investigate whether NLRP3 inflammasome activation has a role in the pathogenesis of hyperoxia-induced preterm brain injury. C57BL6 pups were randomly divided into either a hyperoxia group (exposed to 90 % oxygen from birth until postnatal day 7) or control group (maintained in room air; 21 % O2). At postnatal day 7, all animals were sacrificed. Immunohistochemical examination revealed that hyperoxic exposure for seven days resulted in a global increase in NLRP3 and IL-1ß immunopositive cells in neonatal mouse brain (p ≤ 0.001). There was a significant rise in Caspase-1 positive cell count in prefrontal and parietal area in the hyperoxia group when compared with controls (p ≤ 0.001). Western blot analysis of brain tissues showed elevated NLRP3, IL-1ß and Caspase-1 protein levels in the hyperoxia group when compared with controls (p ≤ 0.001). To the best of our knowledge, this is the first study that investigates an association between hyperoxia and establishment of NLRP3 inflammasome in preterm brain.


Asunto(s)
Encéfalo/efectos de los fármacos , Hiperoxia/metabolismo , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/efectos de los fármacos , Oxígeno , Animales , Animales Recién Nacidos , Encéfalo/metabolismo , Humanos , Recién Nacido , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Oxígeno/metabolismo , Oxígeno/farmacología
16.
Placenta ; 97: 1-5, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32501218

RESUMEN

Although many pregnant women have been infected by coronavirus, the presence of intrauterine vertical transmission has not been conclusively reported yet. What prevents this highly contagious virus from reaching the fetus? Is it only the presence of a strong placental barrier, or is it the natural absence of the some receptor that the viruses use for transmission? We, therefore, need to comprehensively understand the mechanism of action of the mammalian epithelial barriers located in two different organs with functional similarity. The barriers selected as potential targets by SARS-CoV-2 are the alveolo-capillary barrier (ACB), and the syncytio-capillary barrier (SCB). Caveolae are omega-shaped structures located on the cell membrane. They consist of caveolin-1 protein (Cav-1) and are involved in the internalisation of some viruses. By activating leukocytes and nuclear factor-κB, Cav-1 initiates inflammatory reactions. The presence of more than one Cav-1 binding sites on coronavirus is an important finding supporting the possible relationship between SARS-CoV-2-mediated lung injury. While the ACB cells express Cav-1 there is no caveolin expression in syncytiotrophoblasts. In this short review, we will try to explain our hypothesis that the lack of caveolin expression in the SCB is one of the most important physiological mechanisms that prevents vertical transmission of SARS-CoV-2. Since the physiological Cav-1 deficiency appears to prevent acute cell damage treatment algorithms could potentially be developed to block this pathway in the non-pregnant population affected by SARS-CoV-2.


Asunto(s)
Betacoronavirus/fisiología , Infecciones por Coronavirus/prevención & control , Infecciones por Coronavirus/transmisión , Enfermedades Fetales/prevención & control , Transmisión Vertical de Enfermedad Infecciosa/prevención & control , Intercambio Materno-Fetal/inmunología , Pandemias/prevención & control , Neumonía Viral/prevención & control , Neumonía Viral/transmisión , Betacoronavirus/inmunología , COVID-19 , Caveolina 1/fisiología , Infecciones por Coronavirus/inmunología , Epitelio/fisiología , Epitelio/virología , Femenino , Enfermedades Fetales/inmunología , Enfermedades Fetales/virología , Células Gigantes/fisiología , Células Gigantes/virología , Humanos , Inmunidad Innata/fisiología , Neumonía Viral/inmunología , Embarazo , Factores de Riesgo , SARS-CoV-2 , Internalización del Virus
17.
Genome Biol ; 20(1): 37, 2019 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-30791937

RESUMEN

BACKGROUND: Protein recoding by RNA editing is required for normal health and evolutionary adaptation. However, de novo induction of RNA editing in response to environmental factors is an uncommon phenomenon. While APOBEC3A edits many mRNAs in monocytes and macrophages in response to hypoxia and interferons, the physiological significance of such editing is unclear. RESULTS: Here, we show that the related cytidine deaminase, APOBEC3G, induces site-specific C-to-U RNA editing in natural killer cells, lymphoma cell lines, and, to a lesser extent, CD8-positive T cells upon cellular crowding and hypoxia. In contrast to expectations from its anti-HIV-1 function, the highest expression of APOBEC3G is shown to be in cytotoxic lymphocytes. RNA-seq analysis of natural killer cells subjected to cellular crowding and hypoxia reveals widespread C-to-U mRNA editing that is enriched for genes involved in mRNA translation and ribosome function. APOBEC3G promotes Warburg-like metabolic remodeling in HuT78 T cells under similar conditions. Hypoxia-induced RNA editing by APOBEC3G can be mimicked by the inhibition of mitochondrial respiration and occurs independently of HIF-1α. CONCLUSIONS: APOBEC3G is an endogenous RNA editing enzyme in primary natural killer cells and lymphoma cell lines. This RNA editing is induced by cellular crowding and mitochondrial respiratory inhibition to promote adaptation to hypoxic stress.


Asunto(s)
Desaminasa APOBEC-3G/metabolismo , Hipoxia/metabolismo , Células Asesinas Naturales/metabolismo , Edición de ARN , Linfocitos T/metabolismo , Línea Celular Tumoral , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Mitocondrias/metabolismo , Análisis de Secuencia de ARN , Estrés Fisiológico
18.
PLoS One ; 14(6): e0218002, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31170237

RESUMEN

BACKGROUND: Currently, there is a lack of clear definition for neonatal sepsis. The Pediatric Committee of the European Medicines Agency (EMA) developed consensus criteria to ensure a standardization for neonatal sepsis definition. However, there is no evidence supporting the accuracy of the EMA sepsis criteria in neonatal sepsis diagnosis. The main objective of this study was to evaluate the diagnostic accuracy of EMA sepsis criteria for proven neonatal sepsis. METHODS: A multicenter prospective cohort study was conducted from October 2015 to November 2018. Infants with a gestational age over 34th weeks, diagnosed with clinical sepsis and received antibiotics according to the EMA criteria or experienced neonatologists' opinion were included. Blood culture or multiplex real time-PCR or 16S-rRNA positive infants were accepted as "proven sepsis". The predictive performance of EMA criteria for proven sepsis was evaluated by sensitivity, specificity, accuracy, and area under the curve measures of receiver operator characteristic curves. Data-mining methods were used for further analysis. RESULTS: Among the 245 included infants, the EMA criteria were positive in 97 infants (39.6%), while proven sepsis was diagnosed in 113 infants (46.1%). The sensitivity, specificity, and accuracy of the EMA criteria for proven sepsis were 44.2% (95%CI: 34.9-53.9), 64.4% (95%CI: 55.6-72.5), 55.1% (95%CI: 46.6-59.4) respectively. None of the clinical and laboratory parameters had sufficient performance individually in terms of sensitivity, specificity and accuracy measures. The diagnostic performance was similar when different clinical findings were added to the EMA sepsis criteria or assessment of the score was interpreted in different ways. CONCLUSIONS: Results highlighted that clinician opinion and standard laboratory tests are limited in the neonatal sepsis diagnosis. The EMA criteria also did not efficiently meet the diagnostic accuracy measures for neonatal sepsis. A predictive sepsis definition and rapid bedside point-of care tests are urgently needed.


Asunto(s)
Sepsis Neonatal/diagnóstico , Sociedades Médicas , Área Bajo la Curva , Europa (Continente) , Humanos , Recién Nacido
19.
J Clin Endocrinol Metab ; 93(12): 4826-32, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18840642

RESUMEN

CONTEXT: Patients with adrenal and extra-adrenal abdominal paraganglioma (PGL) almost invariably have increased plasma and urine concentrations of metanephrines, the O-methylated metabolites of catecholamines. We report four cases of biochemically silent abdominal PGL, in which metanephrines were normal despite extensive disease. OBJECTIVE: Our objective was to identify the mechanism underlying the lack of catecholamine hypersecretion and metabolism to metanephrines in biochemically silent PGL. DESIGN: This is a descriptive study. SETTING: The study was performed at a referral center. PATIENTS: One index case and three additional patients with large abdominal PGL and metastases but with the lack of evidence of catecholamine production, six patients with metastatic catecholamine-producing PGL and a mutation of the succinate dehydrogenase subunit B (SDHB) gene, and 136 random patients with catecholamine-producing PGL were included in the study. MAIN OUTCOME MEASURES: Plasma, urine, and tumor tissue concentrations of catecholamines and metabolites were calculated with electron microscopy and tyrosine hydroxylase immunohistochemistry. RESULTS: All four patients with biochemically silent PGL had an underlying SDHB mutation. In the index case, the tumor tissue concentration of catecholamines (1.8 nmol/g) was less than 0.01% that of the median (20,410 nmol/g) for the 136 patients with catecholamine-producing tumors. Electron microscopy showed the presence of normal secretory granules in all four biochemically silent PGLs. Tyrosine hydroxylase immunoreactivity was negligible in the four biochemically silent PGLs but abundant in catecholamine-producing PGLs. CONCLUSIONS: Patients with SDHB mutations may present with biochemically silent abdominal PGLs due to defective catecholamine synthesis resulting from the absence of tyrosine hydroxylase. Screening for tumors in patients with SDHB mutations should not be limited to biochemical tests of catecholamine excess.


Asunto(s)
Neoplasias Abdominales/genética , Paraganglioma/genética , Succinato Deshidrogenasa/genética , Neoplasias Abdominales/patología , Adulto , Biomarcadores de Tumor , Catecolaminas/metabolismo , Niño , Femenino , Humanos , Inmunohistoquímica , Masculino , Metanefrina/sangre , Metanefrina/orina , Microscopía Electrónica , Persona de Mediana Edad , Mutación/genética , Mutación/fisiología , Paraganglioma/patología , Tomografía Computarizada por Rayos X , Tirosina 3-Monooxigenasa/metabolismo , Adulto Joven
20.
Oral Oncol ; 44(6): 532-7, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17936061

RESUMEN

Paragangliomas of the head and neck are uncommon, slow-growing, multicentric and are usually benign. Ever since familial paragangliomas were first described a genetic explanation for their existence has been sought. An international collaboration finally elucidated the SDHB, SDHC and SDHD genes for three paraganglioma syndromes (PGL 4, 3, 1). A familial origin should be suspected if other family members have paraganglioma, paragangliomas are multiple, the patient is young or the patient has a vagal paraganglioma. Once familial disease is suspected the best initial screening method is by genetic testing of the patient in question. If genetic testing detects PGL 1, 3 or 4 mutations then the patient's siblings and children should be tested. All genotypically positive patients should be followed periodically as soon as detected. Surveillance is best performed with periodic radionuclide imaging and by directed magnetic resonance imaging. The purpose of surveillance is early detection and consequently earlier treatment. Abundant evidence exists that the risk of complications from surgical intervention increases with increasing tumor size. If tumors are detected and eradicated before they become large, then younger patients can be spared the dysphagia, dysphonia, dysarthria and stroke that have plagued patients undergoing surgery for these tumors.


Asunto(s)
Neoplasias de Cabeza y Cuello/genética , Paraganglioma/genética , Diagnóstico Precoz , Pruebas Genéticas , Neoplasias de Cabeza y Cuello/terapia , Humanos , Mutación , Paraganglioma/terapia , Succinato Deshidrogenasa/genética
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda