Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
J Clin Invest ; 133(19)2023 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-37581936

RESUMEN

Secondary lung infection by inhaled Staphylococcus aureus (SA) is a common and lethal event for individuals infected with influenza A virus (IAV). How IAV disrupts host defense to promote SA infection in lung alveoli, where fatal lung injury occurs, is not known. We addressed this issue using real-time determinations of alveolar responses to IAV in live, intact, perfused lungs. Our findings show that IAV infection blocked defensive alveolar wall liquid (AWL) secretion and induced airspace liquid absorption, thereby reversing normal alveolar liquid dynamics and inhibiting alveolar clearance of inhaled SA. Loss of AWL secretion resulted from inhibition of the cystic fibrosis transmembrane conductance regulator (CFTR) ion channel in the alveolar epithelium, and airspace liquid absorption was caused by stimulation of the alveolar epithelial Na+ channel (ENaC). Loss of AWL secretion promoted alveolar stabilization of inhaled SA, but rescue of AWL secretion protected against alveolar SA stabilization and fatal SA-induced lung injury in IAV-infected mice. These findings reveal a central role for AWL secretion in alveolar defense against inhaled SA and identify AWL inhibition as a critical mechanism of IAV lung pathogenesis. AWL rescue may represent a new therapeutic approach for IAV-SA coinfection.


Asunto(s)
Coinfección , Virus de la Influenza A , Gripe Humana , Lesión Pulmonar , Ratones , Animales , Humanos , Gripe Humana/patología , Lesión Pulmonar/patología , Coinfección/patología , Alveolos Pulmonares/patología , Pulmón/patología
2.
Sci Adv ; 8(51): eadc8753, 2022 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-36542703

RESUMEN

Salivary gland acinar cells are severely depleted after radiotherapy for head and neck cancer, leading to loss of saliva and extensive oro-digestive complications. With no regenerative therapies available, organ dysfunction is irreversible. Here, using the adult murine system, we demonstrate that radiation-damaged salivary glands can be functionally regenerated via sustained delivery of the neurogenic muscarinic receptor agonist cevimeline. We show that endogenous gland repair coincides with increased nerve activity and acinar cell division that is limited to the first week after radiation, with extensive acinar cell degeneration, dysfunction, and cholinergic denervation occurring thereafter. However, we found that mimicking cholinergic muscarinic input via sustained local delivery of a cevimeline-alginate hydrogel was sufficient to regenerate innervated acini and retain physiological saliva secretion at nonirradiated levels over the long term (>3 months). Thus, we reveal a previously unknown regenerative approach for restoring epithelial organ structure and function that has extensive implications for human patients.

3.
Dev Cell ; 57(22): 2550-2565.e5, 2022 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-36413949

RESUMEN

Acinar cells are the principal secretory units of multiple exocrine organs. A single-cell, layered, lumenized acinus forms from a large cohort of epithelial progenitors that must initiate and coordinate three cellular programs of acinar specification, namely, lineage progression, secretion, and polarization. Despite this well-known outcome, the mechanism(s) that regulate these complex programs are unknown. Here, we demonstrate that neuronal-epithelial cross-talk drives acinar specification through neuregulin (NRG1)-ERBB3-mTORC2 signaling. Using single-cell and global RNA sequencing of developing murine salivary glands, we identified NRG1-ERBB3 to precisely overlap with acinar specification during gland development. Genetic deletion of Erbb3 prevented cell lineage progression and the establishment of lumenized, secretory acini. Conversely, NRG1 treatment of isolated epithelia was sufficient to recapitulate the development of secretory acini. Mechanistically, we found that NRG1-ERBB3 regulates each developmental program through an mTORC2 signaling pathway. Thus, we reveal that a neuronal-epithelial (NRG1/ERBB3/mTORC2) mechanism orchestrates the creation of functional acini.


Asunto(s)
Neurregulinas , Transducción de Señal , Humanos , Ratones , Animales , Diana Mecanicista del Complejo 2 de la Rapamicina , Células Acinares , Transporte Biológico , Neurregulina-1 , Receptor ErbB-3
4.
Sci Adv ; 8(34): eabm8757, 2022 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-36026444

RESUMEN

How novel gene functions evolve is a fundamental question in biology. Mucin proteins, a functionally but not evolutionarily defined group of proteins, allow the study of convergent evolution of gene function. By analyzing the genomic variation of mucins across a wide range of mammalian genomes, we propose that exonic repeats and their copy number variation contribute substantially to the de novo evolution of new gene functions. By integrating bioinformatic, phylogenetic, proteomic, and immunohistochemical approaches, we identified 15 undescribed instances of evolutionary convergence, where novel mucins originated by gaining densely O-glycosylated exonic repeat domains. Our results suggest that secreted proteins rich in proline are natural precursors for acquiring mucin function. Our findings have broad implications for understanding the role of exonic repeats in the parallel evolution of new gene functions, especially those involving protein glycosylation.


Asunto(s)
Variaciones en el Número de Copia de ADN , Mucinas , Animales , Glicosilación , Mamíferos , Filogenia , Proteómica
5.
Am J Med Genet A ; 188(6): 1667-1675, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35146895

RESUMEN

TRPM3 encodes a transient receptor potential cation channel of the melastatin family, expressed in the central nervous system and in peripheral sensory neurons of the dorsal root ganglia. The recurrent substitution in TRPM3: c.2509G>A, p.(Val837Met) has been associated with syndromic intellectual disability and seizures. In this report, we present the clinical and molecular features of seven previously unreported individuals, identified by exome sequencing, with the recurrent p.(Val837Met) variant and global developmental delay. Other shared clinical features included congenital hypotonia, dysmorphic facial features (broad forehead, deep-set eyes, and down turned mouth), exotropia, and musculoskeletal issues (hip dysplasia, hip dislocation, scoliosis). Seizures were observed in two of seven individuals (febrile seizure in one and generalized tonic-clonic seizures with atonic drops in another), and epileptiform activity was observed in an additional two individuals. This report extends the number of affected individuals to 16 who are heterozygous for the de novo recurrent substitution p.(Val837Met). In contrast with the initial report, epilepsy was not a mandatory feature observed in this series. TRPM3 pathogenic variation should be considered in individuals with global developmental delays, moderate-severe intellectual disability with, or without, childhood-onset epilepsy.


Asunto(s)
Epilepsia , Enfermedades del Recién Nacido , Discapacidad Intelectual , Canales Catiónicos TRPM , Niño , Discapacidades del Desarrollo/genética , Humanos , Recién Nacido , Discapacidad Intelectual/genética , Hipotonía Muscular/genética , Mutación Missense , Canales Catiónicos TRPM/genética , Secuenciación del Exoma
6.
J Child Neurol ; 37(3): 186-193, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35044272

RESUMEN

Objective: In a randomized trial, we aimed to evaluate the efficacy of cosyntropin injectable suspension, 1 mg/mL, compared to vigabatrin for infantile spasms syndrome. An additional arm was included to assess the efficacy of combination therapy (cosyntropin and vigabatrin) compared with cosyntropin monotherapy. Methods: Children (2 months to 2 years) with new-onset infantile spasms syndrome and hypsarhythmia were randomized into 3 arms: cosyntropin, vigabatrin, and cosyntropin and vigabatrin combined. Daily seizures and adverse events were recorded, and EEG was repeated at day 14 to assess for resolution of hypsarhythmia. The primary outcome measure was the composite of resolution of hypsarhythmia and absence of clinical spasms at day 14. Fisher exact test was used to compare outcomes. Results: 37 children were enrolled and 34 were included in the final efficacy analysis (1 withdrew prior to treatment and 2 did not return seizure diaries). Resolution of both hypsarhythmia and clinical spasms was achieved in in 9 of 12 participants (75%) treated with cosyntropin, 1/9 (11%) vigabatrin, and 5/13 (38%) cosyntropin and vigabatrin combined. The primary comparison of cosyntropin versus vigabatrin was significant (64% [95% confidence interval 21, 82], P < .01). Adverse events were reported in all 3 treatment arms: 31 (86%) had an adverse event, 7 (19%) had a serious adverse event, and 15 (42%) had an adverse event of special interest with no difference between treatment arms. Significance: This randomized trial was underpowered because of incomplete enrollment, yet it demonstrated that cosyntropin was more effective for short-term outcomes than vigabatrin as initial treatment for infantile spasms.


Asunto(s)
Espasmos Infantiles , Vigabatrin , Anticonvulsivantes/efectos adversos , Niño , Cosintropina/uso terapéutico , Humanos , Estudios Prospectivos , Espasmo/inducido químicamente , Espasmo/complicaciones , Espasmo/tratamiento farmacológico , Espasmos Infantiles/tratamiento farmacológico , Espasmos Infantiles/etiología , Resultado del Tratamiento , Vigabatrin/efectos adversos
7.
Nat Genet ; 53(7): 1006-1021, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34211179

RESUMEN

SPTBN1 encodes ßII-spectrin, the ubiquitously expressed ß-spectrin that forms micrometer-scale networks associated with plasma membranes. Mice deficient in neuronal ßII-spectrin have defects in cortical organization, developmental delay and behavioral deficiencies. These phenotypes, while less severe, are observed in haploinsufficient animals, suggesting that individuals carrying heterozygous SPTBN1 variants may also show measurable compromise of neural development and function. Here we identify heterozygous SPTBN1 variants in 29 individuals with developmental, language and motor delays; mild to severe intellectual disability; autistic features; seizures; behavioral and movement abnormalities; hypotonia; and variable dysmorphic facial features. We show that these SPTBN1 variants lead to effects that affect ßII-spectrin stability, disrupt binding to key molecular partners, and disturb cytoskeleton organization and dynamics. Our studies define SPTBN1 variants as the genetic basis of a neurodevelopmental syndrome, expand the set of spectrinopathies affecting the brain and underscore the critical role of ßII-spectrin in the central nervous system.


Asunto(s)
Genes Dominantes , Predisposición Genética a la Enfermedad , Variación Genética , Trastornos del Neurodesarrollo/genética , Espectrina/genética , Animales , Estudios de Asociación Genética/métodos , Heterocigoto , Humanos , Ratones , Trastornos del Neurodesarrollo/diagnóstico , Fenotipo , Espectrina/metabolismo
8.
Int Forum Allergy Rhinol ; 11(10): 1443-1451, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33956392

RESUMEN

BACKGROUND: Chronic rhinosinusitis (CRS) is characterized by significant accumulation and thickening of mucus in the sinonasal cavities. One contributor of aberrant mucus production and impaired mucociliary clearance (MCC) is altered function of the sinonasal submucosal glands (SMGs), yet contributions of SMGs to upper airway disease initiation and progression remain unknown. The objective of this study was to characterize the morphology and secretory cell identities of the nasal septum SMGs in both healthy and CRS adults. METHODS: Biopsies from adult participants with CRS without nasal polyps (CRSsNP, n = 4), CRS with nasal polyps (CRSwNP, n = 8), and non-CRS controls (n = 14) were collected from the posterior septum. Glandular morphology and mucus markers were investigated using histological techniques and high-resolution confocal microscopy. RESULTS: Analysis revealed a significant decrease in gland density in the posterior septum of CRSsNP (28% ± 6.15%) and CRSwNP (23% ± 3.09%) compared to control participants (53% ± 1.59%, p < 0.0001). Further analysis of the CRS SMG secretory function revealed an overall decrease in Mucin 5B+ gland mucus being produced. Dilated and cystic ductal structures filled with inspissated mucus were also common to CRS glands. CONCLUSION: Here, we describe a significant alteration in SMG structure and function in the adult CRS posterior septum suggesting reduced gland contribution to MCC. The SMGs of both the nose and sinuses may represent targets for future therapeutic approaches.


Asunto(s)
Pólipos Nasales , Rinitis , Sinusitis , Enfermedad Crónica , Humanos , Mucinas , Mucosa Nasal/patología , Pólipos Nasales/patología , Rinitis/patología , Sinusitis/patología
9.
Am J Hum Genet ; 108(4): 749-756, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33743206

RESUMEN

The DNA damage-binding protein 1 (DDB1) is part of the CUL4-DDB1 ubiquitin E3 ligase complex (CRL4), which is essential for DNA repair, chromatin remodeling, DNA replication, and signal transduction. Loss-of-function variants in genes encoding the complex components CUL4 and PHIP have been reported to cause syndromic intellectual disability with hypotonia and obesity, but no phenotype has been reported in association with DDB1 variants. Here, we report eight unrelated individuals, identified through Matchmaker Exchange, with de novo monoallelic variants in DDB1, including one recurrent variant in four individuals. The affected individuals have a consistent phenotype of hypotonia, mild to moderate intellectual disability, and similar facies, including horizontal or slightly bowed eyebrows, deep-set eyes, full cheeks, a short nose, and large, fleshy and forward-facing earlobes, demonstrated in the composite face generated from the cohort. Digital anomalies, including brachydactyly and syndactyly, were common. Three older individuals have obesity. We show that cells derived from affected individuals have altered DDB1 function resulting in abnormal DNA damage signatures and histone methylation following UV-induced DNA damage. Overall, our study adds to the growing family of neurodevelopmental phenotypes mediated by disruption of the CRL4 ubiquitin ligase pathway and begins to delineate the phenotypic and molecular effects of DDB1 misregulation.


Asunto(s)
Alelos , Reparación del ADN/genética , Proteínas de Unión al ADN/genética , Mutación , Trastornos del Neurodesarrollo/genética , Adolescente , Niño , Preescolar , Femenino , Humanos , Masculino , Fenotipo , Síndrome
10.
Cell Rep ; 33(7): 108402, 2020 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-33207190

RESUMEN

Salivary proteins are essential for maintaining health in the oral cavity and proximal digestive tract, and they serve as potential diagnostic markers for monitoring human health and disease. However, their precise organ origins remain unclear. Through transcriptomic analysis of major adult and fetal salivary glands and integration with the saliva proteome, the blood plasma proteome, and transcriptomes of 28+ organs, we link human saliva proteins to their source, identify salivary-gland-specific genes, and uncover fetal- and adult-specific gene repertoires. Our results also provide insights into the degree of gene retention during gland maturation and suggest that functional diversity among adult gland types is driven by specific dosage combinations of hundreds of transcriptional regulators rather than by a few gland-specific factors. Finally, we demonstrate the heterogeneity of the human acinar cell lineage. Our results pave the way for future investigations into glandular biology and pathology, as well as saliva's use as a diagnostic fluid.


Asunto(s)
Saliva/química , Saliva/metabolismo , Glándulas Salivales/metabolismo , Adulto , Anciano , Femenino , Feto , Expresión Génica/genética , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica/genética , Humanos , Masculino , Persona de Mediana Edad , Boca/metabolismo , Proteoma/metabolismo , Glándulas Salivales/fisiología , Proteínas y Péptidos Salivales/metabolismo , Relación Estructura-Actividad , Transcriptoma/genética
11.
Dev Biol ; 451(2): 158-166, 2019 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-30965042

RESUMEN

Mucus secretion and mucociliary clearance are crucial processes required to maintain pulmonary homeostasis. In the trachea and nasal passages, mucus is secreted by submucosal glands (SMGs) that line the airway, with an additional contribution from goblet cells of the surface airway epithelium. The SMG mucus is rich in mucins and antimicrobial enzymes. Defective tracheal SMGs contribute to hyper-secretory respiratory diseases, such as cystic fibrosis, asthma, and chronic obstructive pulmonary disease, however little is known about the signals that regulate their morphogenesis and patterning. Here, we show that Fgf10 is essential for the normal development of murine tracheal SMGs, with gland development arresting at the early bud stage in the absence of FGF10 signalling. As Fgf10 knockout mice are lethal at birth, inducible knockdown of Fgf10 at late embryonic stages was used to follow postnatal gland formation, confirming the essential role of FGF10 in SMG development. In heterozygous Fgf10 mice the tracheal glands formed but with altered morphology and restricted distribution. The reduction in SMG branching in Fgf10 heterozygous mice was not rescued with time and resulted in a reduction in overall tracheal mucus secretion. Fgf10 is therefore a key signal in SMG development, influencing both the number of glands and extent of branching morphogenesis, and is likely, therefore, to play a role in aspects of SMG-dependent respiratory health.


Asunto(s)
Glándulas Exocrinas/embriología , Factor 10 de Crecimiento de Fibroblastos/metabolismo , Mucosa Respiratoria/embriología , Tráquea/embriología , Animales , Cruzamientos Genéticos , Femenino , Factor 10 de Crecimiento de Fibroblastos/deficiencia , Factor 10 de Crecimiento de Fibroblastos/genética , Masculino , Ratones , Morfogénesis , Moco/metabolismo , Tráquea/metabolismo
12.
Development ; 145(21)2018 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-30305288

RESUMEN

The ductal system of the salivary gland has long been postulated to be resistant to radiation-induced damage, a common side effect incurred by head and neck cancer patients receiving radiotherapy. Yet, whether the ducts are capable of regenerating after genotoxic injury, or whether damage to ductal cells induces lineage plasticity, as has been reported in other organ systems, remains unknown. Here, using the murine salivary gland, we show that two ductal progenitor populations, marked exclusively by KRT14 and KIT, maintain non-overlapping ductal compartments after radiation exposure but do so through distinct cellular mechanisms. KRT14+ progenitor cells are fast-cycling cells that proliferate in response to radiation-induced damage in a sustained manner and divide asymmetrically to produce differentiated cells of the larger granulated ducts. Conversely, KIT+ intercalated duct cells are long-lived progenitors for the intercalated ducts that undergo few cell divisions either during homeostasis or after gamma radiation, thus maintaining ductal architecture with slow rates of cell turnover. Together, these data illustrate the regenerative capacity of the salivary ducts and highlight the heterogeneity in the damage responses used by salivary progenitor cells to maintain tissue architecture.


Asunto(s)
Traumatismos por Radiación/terapia , Conductos Salivales/patología , Conductos Salivales/efectos de la radiación , Trasplante de Células Madre , Células Madre/citología , Células Acinares/metabolismo , Animales , Animales Recién Nacidos , División Celular Asimétrica , Linaje de la Célula , Proliferación Celular , Células Epiteliales/metabolismo , Femenino , Humanos , Queratina-14/metabolismo , Masculino , Ratones Endogámicos C57BL , Modelos Biológicos , Proteínas Proto-Oncogénicas c-kit/metabolismo , Traumatismos por Radiación/patología , Conductos Salivales/metabolismo , Glándula Submandibular/metabolismo , Glándula Submandibular/patología , Glándula Submandibular/efectos de la radiación
13.
NCSL Legisbrief ; 26(31): 1-2, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30199174

RESUMEN

(1) Studies have found a $1.80 to $5.70 return on investment for every dollar spent on home visiting. (2) Nearly 4 million evidence-based home visits reached more than 300,000 families in 2016. (3) Approximately 40 percent of U.S. counties have at least one home visiting agency that offers an evidence-based program.


Asunto(s)
Protección a la Infancia , Servicios de Atención de Salud a Domicilio , Bienestar del Lactante , Preescolar , Análisis Costo-Beneficio , Medicina Basada en la Evidencia , Familia , Gobierno Federal , Servicios de Atención de Salud a Domicilio/economía , Servicios de Atención de Salud a Domicilio/legislación & jurisprudencia , Humanos , Lactante , Recién Nacido , Gobierno Estatal , Estados Unidos
14.
EMBO Mol Med ; 10(3)2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29335337

RESUMEN

Salivary gland acinar cells are routinely destroyed during radiation treatment for head and neck cancer that results in a lifetime of hyposalivation and co-morbidities. A potential regenerative strategy for replacing injured tissue is the reactivation of endogenous stem cells by targeted therapeutics. However, the identity of these cells, whether they are capable of regenerating the tissue, and the mechanisms by which they are regulated are unknown. Using in vivo and ex vivo models, in combination with genetic lineage tracing and human tissue, we discover a SOX2+ stem cell population essential to acinar cell maintenance that is capable of replenishing acini after radiation. Furthermore, we show that acinar cell replacement is nerve dependent and that addition of a muscarinic mimetic is sufficient to drive regeneration. Moreover, we show that SOX2 is diminished in irradiated human salivary gland, along with parasympathetic nerves, suggesting that tissue degeneration is due to loss of progenitors and their regulators. Thus, we establish a new paradigm that salivary glands can regenerate after genotoxic shock and do so through a SOX2 nerve-dependent mechanism.


Asunto(s)
Traumatismos por Radiación/patología , Traumatismos por Radiación/fisiopatología , Regeneración , Factores de Transcripción SOXB1/metabolismo , Glándulas Salivales/patología , Glándulas Salivales/fisiopatología , Acetilcolina/metabolismo , Células Acinares/metabolismo , Células Acinares/efectos de la radiación , Adulto , Anciano , Animales , Linaje de la Célula/efectos de la radiación , Proliferación Celular/efectos de la radiación , Nervio de la Cuerda del Tímpano/patología , Nervio de la Cuerda del Tímpano/efectos de la radiación , Femenino , Homeostasis , Humanos , Masculino , Ratones Endogámicos C57BL , Persona de Mediana Edad , Traumatismos por Radiación/metabolismo , Radiación Ionizante , Receptores Muscarínicos/metabolismo , Glándulas Salivales/efectos de la radiación , Transducción de Señal , Células Madre/metabolismo , Células Madre/efectos de la radiación
15.
Elife ; 62017 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-28623666

RESUMEN

Acinar cells play an essential role in the secretory function of exocrine organs. Despite this requirement, how acinar cells are generated during organogenesis is unclear. Using the acini-ductal network of the developing human and murine salivary gland, we demonstrate an unexpected role for SOX2 and parasympathetic nerves in generating the acinar lineage that has broad implications for epithelial morphogenesis. Despite SOX2 being expressed by progenitors that give rise to both acinar and duct cells, genetic ablation of SOX2 results in a failure to establish acini but not ducts. Furthermore, we show that SOX2 targets acinar-specific genes and is essential for the survival of acinar but not ductal cells. Finally, we illustrate an unexpected and novel role for peripheral nerves in the creation of acini throughout development via regulation of SOX2. Thus, SOX2 is a master regulator of the acinar cell lineage essential to the establishment of a functional organ.


Asunto(s)
Células Acinares/fisiología , Diferenciación Celular , Organogénesis , Factores de Transcripción SOXB1/metabolismo , Glándulas Salivales/citología , Glándulas Salivales/embriología , Animales , Técnicas de Inactivación de Genes , Humanos , Ratones
16.
Dev Biol ; 419(2): 348-356, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27590203

RESUMEN

Hypertrophy, hyperplasia and altered mucus secretion from the respiratory submucosal glands (SMG) are characteristics of airway diseases such as cystic fibrosis, asthma and chronic bronchitis. More commonly, hyper-secretion of the nasal SMGs contributes to allergic rhinitis and upper airway infection. Considering the role of these glands in disease states, there is a significant dearth in understanding the molecular signals that regulate SMG development and patterning. Due to the imperative role of FGF signalling during the development of other branched structures, we investigated the role of Fgf10 during initiation and branching morphogenesis of murine nasal SMGs. Fgf10 is expressed in the mesenchyme around developing SMGs while expression of its receptor Fgfr2 is seen within glandular epithelial cells. In the Fgf10 null embryo, Steno's gland and the maxillary sinus gland were completely absent while other neighbouring nasal glands showed normal duct elongation but defective branching. Interestingly, the medial nasal glands were present in Fgf10 homozygotes but missing in Fgfr2b mutants, with expression of Fgf7 specifically expressed around these developing glands, indicating that Fgf7 might compensate for loss of Fgf10 in this group of glands. Intriguingly the lateral nasal glands were only mildly affected by loss of FGF signalling, while these glands were missing in Eda mutant mice, where the Steno's and maxillary sinus gland developed as normal. This analysis reveals that regulation of nasal gland development is complex with different subsets of glands being regulated by different signalling pathways. This analysis helps shed light on the nasal gland defects observed in patients with hypohidrotic ectodermal dysplasia (HED) (defect EDA pathway) and LADD syndrome (defect FGFR2b pathway).


Asunto(s)
Ectodisplasinas/fisiología , Glándulas Exocrinas/embriología , Factor 10 de Crecimiento de Fibroblastos/fisiología , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/fisiología , Transducción de Señal/fisiología , Animales , Ectodisplasinas/deficiencia , Ectodisplasinas/genética , Resección Endoscópica de la Mucosa , Glándulas Exocrinas/metabolismo , Glándulas Exocrinas/ultraestructura , Femenino , Factor 10 de Crecimiento de Fibroblastos/deficiencia , Factor 10 de Crecimiento de Fibroblastos/genética , Factor 7 de Crecimiento de Fibroblastos/fisiología , Masculino , Seno Maxilar/embriología , Seno Maxilar/ultraestructura , Mesodermo/metabolismo , Ratones , Morfogénesis , Mucosa Nasal/embriología , Mucosa Nasal/ultraestructura , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/deficiencia , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética
17.
Blood ; 128(15): 1913-1917, 2016 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-27488349

RESUMEN

The congenital sideroblastic anemias (CSAs) are a heterogeneous group of inherited blood disorders characterized by pathological mitochondrial iron deposition in erythroid precursors. Each known cause has been attributed to a mutation in a protein associated with heme biosynthesis, iron-sulfur cluster biogenesis, mitochondrial translation, or a component of the mitochondrial respiratory chain. Here, we describe a recurring mutation, c.276_278del, p.F93del, in NDUFB11, a mitochondrial respiratory complex I-associated protein encoded on the X chromosome, in 5 males with a variably syndromic, normocytic CSA. The p.F93del mutation results in respiratory insufficiency and loss of complex I stability and activity in patient-derived fibroblasts. Targeted introduction of this allele into K562 erythroleukemia cells results in a proliferation defect with minimal effect on erythroid differentiation potential, suggesting the mechanism of anemia in this disorder.


Asunto(s)
Anemia Sideroblástica/genética , Secuencia de Bases , Cromosomas Humanos X/genética , Complejo I de Transporte de Electrón/genética , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Eliminación de Secuencia , Adolescente , Adulto , Anciano , Anemia Sideroblástica/metabolismo , Anemia Sideroblástica/patología , Niño , Preescolar , Cromosomas Humanos X/metabolismo , Complejo I de Transporte de Electrón/metabolismo , Femenino , Enfermedades Genéticas Ligadas al Cromosoma X/metabolismo , Humanos , Células K562 , Masculino , Persona de Mediana Edad
18.
Blood ; 126(25): 2734-8, 2015 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-26491070

RESUMEN

The congenital sideroblastic anemias (CSAs) are relatively uncommon diseases characterized by defects in mitochondrial heme synthesis, iron-sulfur (Fe-S) cluster biogenesis, or protein synthesis. Here we demonstrate that mutations in HSPA9, a mitochondrial HSP70 homolog located in the chromosome 5q deletion syndrome 5q33 critical deletion interval and involved in mitochondrial Fe-S biogenesis, result in CSA inherited as an autosomal recessive trait. In a fraction of patients with just 1 severe loss-of-function allele, expression of the clinical phenotype is associated with a common coding single nucleotide polymorphism in trans that correlates with reduced messenger RNA expression and results in a pseudodominant pattern of inheritance.


Asunto(s)
Anemia Sideroblástica/genética , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Proteínas HSP70 de Choque Térmico/genética , Proteínas Mitocondriales/genética , Adulto , Anciano , Secuencia de Bases , Análisis Mutacional de ADN , Femenino , Genotipo , Humanos , Lactante , Recién Nacido , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Mutación , Análisis de Secuencia por Matrices de Oligonucleótidos , Linaje , Polimorfismo de Nucleótido Simple , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Adulto Joven
20.
Dev Dyn ; 244(4): 525-39, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25648514

RESUMEN

BACKGROUND: The submucosal glands (SMGs) of the respiratory system are specialized structures essential for maintaining airway homeostasis. The significance of SMGs is highlighted by their involvement in respiratory diseases such as cystic fibrosis, asthma and chronic bronchitis, where their phenotype and function are severely altered. Uncovering the normal development of the airway SMGs is essential to elucidate their role in these disorders, however, very little is known about the cellular mechanisms and intracellular signals involved in their morphogenesis. RESULTS: This review describes in detail the embryonic developmental journey of the nasal SMGs and the postnatal development of the tracheal SMGs in the mouse. Current knowledge of the genes and signalling molecules involved in SMG organogenesis is also explored. CONCLUSION: Here we review the temporal localisation and development of the murine respiratory glands in the hope of stimulating further research into the mechanisms required for successful SMG patterning and function.


Asunto(s)
Glándulas Exocrinas/embriología , Sistema Respiratorio/embriología , Adipocitos/metabolismo , Animales , Animales Modificados Genéticamente , Membrana Basal/metabolismo , Colágeno Tipo IV/metabolismo , Drosophila melanogaster , Matriz Extracelular/metabolismo , Hemocitos/metabolismo , Ratones , Microscopía Electrónica de Rastreo , Microscopía Electrónica de Transmisión , Mutación , Necrosis , Osteonectina/metabolismo , Fenotipo , Temperatura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA