Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Annu Rev Physiol ; 83: 529-550, 2021 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-33074772

RESUMEN

The mammalian lung epithelium is composed of a wide array of specialized cells that have adapted to survive environmental exposure and perform the tasks necessary for respiration. Although the majority of these cells are remarkably quiescent during adult lung homeostasis, a growing body of literature has demonstrated the capacity of these epithelial lineages to proliferate in response to injury and regenerate lost or damaged cells. In this review, we focus on the regionally distinct lung epithelial cell types that contribute to repair after injury, and we address current controversies regarding whether elite stem cells or frequent facultative progenitors are the predominant participants. We also shed light on the newly emerging approaches for exogenously generating similar lung epithelial lineages from pluripotent stem cells.


Asunto(s)
Células Epiteliales/citología , Pulmón/citología , Pulmón/fisiología , Regeneración/fisiología , Células Madre/citología , Animales , Diferenciación Celular/fisiología , Linaje de la Célula/fisiología , Homeostasis/fisiología , Humanos
3.
PLoS Pathog ; 18(2): e1010268, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35120176

RESUMEN

Next generation sequencing has revealed the presence of numerous RNA viruses in animal reservoir hosts, including many closely related to known human pathogens. Despite their zoonotic potential, most of these viruses remain understudied due to not yet being cultured. While reverse genetic systems can facilitate virus rescue, this is often hindered by missing viral genome ends. A prime example is Lloviu virus (LLOV), an uncultured filovirus that is closely related to the highly pathogenic Ebola virus. Using minigenome systems, we complemented the missing LLOV genomic ends and identified cis-acting elements required for LLOV replication that were lacking in the published sequence. We leveraged these data to generate recombinant full-length LLOV clones and rescue infectious virus. Similar to other filoviruses, recombinant LLOV (rLLOV) forms filamentous virions and induces the formation of characteristic inclusions in the cytoplasm of the infected cells, as shown by electron microscopy. Known target cells of Ebola virus, including macrophages and hepatocytes, are permissive to rLLOV infection, suggesting that humans could be potential hosts. However, inflammatory responses in human macrophages, a hallmark of Ebola virus disease, are not induced by rLLOV. Additional tropism testing identified pneumocytes as capable of robust rLLOV and Ebola virus infection. We also used rLLOV to test antivirals targeting multiple facets of the replication cycle. Rescue of uncultured viruses of pathogenic concern represents a valuable tool in our arsenal for pandemic preparedness.


Asunto(s)
Ebolavirus/genética , Infecciones por Filoviridae/virología , Filoviridae/genética , Replicación Viral , Animales , Línea Celular , Chlorocebus aethiops , Prueba de Complementación Genética , Genoma Viral , Fiebre Hemorrágica Ebola/virología , Interacciones Microbiota-Huesped , Humanos , Cuerpos de Inclusión/virología , Células Madre Pluripotentes Inducidas/virología , Macrófagos/virología , ARN Viral , Genética Inversa , Células Vero , Virión/genética
4.
Proc Natl Acad Sci U S A ; 118(16)2021 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-33811184

RESUMEN

Coronaviruses are adept at evading host antiviral pathways induced by viral double-stranded RNA, including interferon (IFN) signaling, oligoadenylate synthetase-ribonuclease L (OAS-RNase L), and protein kinase R (PKR). While dysregulated or inadequate IFN responses have been associated with severe coronavirus infection, the extent to which the recently emerged SARS-CoV-2 activates or antagonizes these pathways is relatively unknown. We found that SARS-CoV-2 infects patient-derived nasal epithelial cells, present at the initial site of infection; induced pluripotent stem cell-derived alveolar type 2 cells (iAT2), the major cell type infected in the lung; and cardiomyocytes (iCM), consistent with cardiovascular consequences of COVID-19 disease. Robust activation of IFN or OAS-RNase L is not observed in these cell types, whereas PKR activation is evident in iAT2 and iCM. In SARS-CoV-2-infected Calu-3 and A549ACE2 lung-derived cell lines, IFN induction remains relatively weak; however, activation of OAS-RNase L and PKR is observed. This is in contrast to Middle East respiratory syndrome (MERS)-CoV, which effectively inhibits IFN signaling and OAS-RNase L and PKR pathways, but is similar to mutant MERS-CoV lacking innate immune antagonists. Remarkably, OAS-RNase L and PKR are activated in MAVS knockout A549ACE2 cells, demonstrating that SARS-CoV-2 can induce these host antiviral pathways despite minimal IFN production. Moreover, increased replication and cytopathic effect in RNASEL knockout A549ACE2 cells implicates OAS-RNase L in restricting SARS-CoV-2. Finally, while SARS-CoV-2 fails to antagonize these host defense pathways, which contrasts with other coronaviruses, the IFN signaling response is generally weak. These host-virus interactions may contribute to the unique pathogenesis of SARS-CoV-2.


Asunto(s)
Células Epiteliales/inmunología , Células Epiteliales/virología , Inmunidad Innata , Pulmón/patología , Miocitos Cardíacos/inmunología , Miocitos Cardíacos/virología , ARN Bicatenario/metabolismo , SARS-CoV-2/inmunología , Células A549 , Endorribonucleasas/metabolismo , Humanos , Coronavirus del Síndrome Respiratorio de Oriente Medio/inmunología , Coronavirus del Síndrome Respiratorio de Oriente Medio/fisiología , Nariz/virología , Replicación Viral , eIF-2 Quinasa
5.
Development ; 144(21): 3879-3893, 2017 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-28947536

RESUMEN

The in vitro-directed differentiation of pluripotent stem cells (PSCs) through stimulation of developmental signaling pathways can generate mature somatic cell types for basic laboratory studies or regenerative therapies. However, there has been significant uncertainty regarding a method to separately derive lung versus thyroid epithelial lineages, as these two cell types each originate from Nkx2-1+ foregut progenitors and the minimal pathways claimed to regulate their distinct lineage specification in vivo or in vitro have varied in previous reports. Here, we employ PSCs to identify the key minimal signaling pathways (Wnt+BMP versus BMP+FGF) that regulate distinct lung- versus thyroid-lineage specification, respectively, from foregut endoderm. In contrast to most previous reports, these minimal pathways appear to be evolutionarily conserved between mice and humans, and FGF signaling, although required for thyroid specification, unexpectedly appears to be dispensable for lung specification. Once specified, distinct Nkx2-1+ lung or thyroid progenitor pools can now be independently derived for functional 3D culture maturation, basic developmental studies or future regenerative therapies.


Asunto(s)
Tipificación del Cuerpo , Diferenciación Celular , Pulmón/citología , Pulmón/embriología , Células Madre Pluripotentes/citología , Transducción de Señal , Glándula Tiroides/citología , Animales , Biomarcadores/metabolismo , Tipificación del Cuerpo/genética , Proteínas Morfogenéticas Óseas/metabolismo , Linaje de la Célula , Embrión de Mamíferos/citología , Desarrollo Embrionario , Endodermo/citología , Endodermo/metabolismo , Células Epiteliales/citología , Factores de Crecimiento de Fibroblastos/metabolismo , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Proteínas Fluorescentes Verdes/metabolismo , Proteínas de Homeodominio/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/metabolismo , Reproducibilidad de los Resultados , Esferoides Celulares/citología , Esferoides Celulares/metabolismo , Glándula Tiroides/embriología , Transcriptoma/genética , Proteínas Wnt/metabolismo
6.
Eur J Nutr ; 56(1): 161-170, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26525562

RESUMEN

OBJECTIVE: We examined associations between body weight and plasma 25-hydroxyvitamin D concentration (25OHD) in prediabetes and sought to estimate the impact of adiposity on these associations. METHODS: The study was conducted in the placebo (n = 1082) and intensive lifestyle (n = 1079) groups of the Diabetes Prevention Program (DPP), a multicenter trial to prevent type 2 diabetes in adults with prediabetes. Weight and 25OHD were measured at baseline, month 6, years 1 and 2. In a subset (n = 584), visceral (VAT) and subcutaneous (SAT) adiposity were assessed by computed tomography at baseline and year 1. RESULTS: In cross-sectional analyses, baseline body weight, total fat, VAT, and SAT were inversely associated with plasma 25OHD concentration after multivariable adjustment. VAT accounted for 40 % [95 % CI 11, 69] of the association of body weight with plasma 25OHD concentration. There was no significant contribution by total fat or SAT. Two-year changes in plasma 25OHD concentration varied inversely with changes in body weight (p < 0.0001). One-year changes in total fat, VAT, or SAT were not significant mediators of the association between change in plasma 25OHD concentration and body weight. CONCLUSION: Our study found an inverse association between body weight and plasma 25OHD concentration at baseline and over a 2-year period in adults with prediabetes. These findings in the DPP, a weight loss intervention study, raise the possibility that weight loss increases plasma 25OHD concentration. Whether adiposity mediates this association remains inconclusive.


Asunto(s)
Composición Corporal , Peso Corporal , Diabetes Mellitus Tipo 2/sangre , Vitamina D/análogos & derivados , Adiposidad , Adulto , Índice de Masa Corporal , Estudios Transversales , Diabetes Mellitus Tipo 2/prevención & control , Femenino , Estudios de Seguimiento , Humanos , Grasa Intraabdominal/metabolismo , Estilo de Vida , Modelos Lineales , Estudios Longitudinales , Masculino , Persona de Mediana Edad , Obesidad/sangre , Estado Prediabético/sangre , Estado Prediabético/tratamiento farmacológico , Estaciones del Año , Grasa Subcutánea Abdominal/metabolismo , Vitamina D/sangre , Circunferencia de la Cintura
7.
Cell Stem Cell ; 31(4): 439-454, 2024 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-38492572

RESUMEN

The respiratory system acts as both the primary site of gas exchange and an important sensor and barrier to the external environment. The increase in incidences of respiratory disease over the past decades has highlighted the importance of developing improved therapeutic approaches. This review will summarize recent research on the cellular complexity of the mammalian respiratory system with a focus on gas exchange and immunological defense functions of the lung. Different models of repair and regeneration will be discussed to help interpret human and animal data and spur the investigation of models and assays for future drug development.


Asunto(s)
Pulmón , Mamíferos , Animales , Humanos
8.
EBioMedicine ; 104: 105135, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38718684

RESUMEN

Interstitial lung diseases (ILDs) in adults and children (chILD) are a heterogeneous group of lung disorders leading to inflammation, abnormal tissue repair and scarring of the lung parenchyma often resulting in respiratory failure and death. Inherited factors directly cause, or contribute significantly to the risk of developing ILD, so called familial pulmonary fibrosis (FPF), and monogenic forms may have a poor prognosis and respond poorly to current treatments. Specific, variant-targeted or precision treatments are lacking. Clinical trials of repurposed drugs, anti-fibrotic medications and specific treatments are emerging but for many patients no interventions exist. We convened an expert working group to develop an overarching framework to address the existing research gaps in basic, translational, and clinical research and identified areas for future development of preclinical models, candidate medications and innovative clinical trials. In this Position Paper, we summarise working group discussions, recommendations, and unresolved questions concerning precision treatments for FPF.


Asunto(s)
Medicina de Precisión , Humanos , Medicina de Precisión/métodos , Animales , Manejo de la Enfermedad , Fibrosis Pulmonar/terapia , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/etiología , Ensayos Clínicos como Asunto
9.
bioRxiv ; 2024 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-38562781

RESUMEN

Human induced pluripotent stem cell (iPSC) derived alveolar organoids have emerged as a system to model the alveolar epithelium in homeostasis and disease. However, alveolar organoids are typically grown in Matrigel, a mouse-sarcoma derived basement membrane matrix that offers poor control over matrix properties, prompting the development of synthetic hydrogels as a Matrigel alternative. Here, we develop a two-step culture method that involves pre-aggregation of organoids in hydrogel-based microwells followed by embedding in a synthetic hydrogel that supports alveolar organoid growth, while also offering considerable control over organoid and hydrogel properties. We find that the aggregated organoids secrete their own nascent extracellular matrix (ECM) both in the microwells and upon embedding in the synthetic hydrogels. Thus, the synthetic gels described here allow us to de-couple exogenous and nascent ECM in order to interrogate the role of ECM in organoid formation.

10.
Cell Stem Cell ; 31(5): 657-675.e8, 2024 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-38642558

RESUMEN

Alveolar epithelial type I cells (AT1s) line the gas exchange barrier of the distal lung and have been historically challenging to isolate or maintain in cell culture. Here, we engineer a human in vitro AT1 model system via directed differentiation of induced pluripotent stem cells (iPSCs). We use primary adult AT1 global transcriptomes to suggest benchmarks and pathways, such as Hippo-LATS-YAP/TAZ signaling, enriched in these cells. Next, we generate iPSC-derived alveolar epithelial type II cells (AT2s) and find that nuclear YAP signaling is sufficient to promote a broad transcriptomic shift from AT2 to AT1 gene programs. The resulting cells express a molecular, morphologic, and functional phenotype reminiscent of human AT1 cells, including the capacity to form a flat epithelial barrier producing characteristic extracellular matrix molecules and secreted ligands. Our results provide an in vitro model of human alveolar epithelial differentiation and a potential source of human AT1s.


Asunto(s)
Células Epiteliales Alveolares , Diferenciación Celular , Células Madre Pluripotentes Inducidas , Humanos , Células Epiteliales Alveolares/citología , Células Epiteliales Alveolares/metabolismo , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Transducción de Señal , Células Cultivadas , Transcriptoma/genética , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo
11.
bioRxiv ; 2024 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-38585863

RESUMEN

Alveolar epithelial type II (AT2) cell dysfunction is implicated in the pathogenesis of familial and sporadic idiopathic pulmonary fibrosis (IPF). We previously described that expression of an AT2 cell exclusive disease-associated protein isoform (SP-CI73T) in murine and patient-specific induced pluripotent stem cell (iPSC)-derived AT2 cells leads to a block in late macroautophagy and promotes time-dependent mitochondrial impairments; however, how a metabolically dysfunctional AT2 cell results in fibrosis remains elusive. Here using murine and human iPSC-derived AT2 cell models expressing SP-CI73T, we characterize the molecular mechanisms governing alterations in AT2 cell metabolism that lead to increased glycolysis, decreased mitochondrial biogenesis, disrupted fatty acid oxidation, accumulation of impaired mitochondria, and diminished AT2 cell progenitor capacity manifesting as reduced AT2 self-renewal and accumulation of transitional epithelial cells. We identify deficient AMP-kinase signaling as a key upstream signaling hub driving disease in these dysfunctional AT2 cells and augment this pathway to restore alveolar epithelial metabolic function, thus successfully alleviating lung fibrosis in vivo.

12.
Biochim Biophys Acta ; 1822(1): 34-41, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21193035

RESUMEN

Autism spectrum disorders (ASD) are neurodevelopmental disorders characterized by varying degrees of dysfunctional communication and social interactions, repetitive and stereotypic behaviors, as well as learning and sensory deficits. Despite the impressive rise in the prevalence of autism during the last two decades, there are few if any clues for its pathogenesis, early detection or treatment. Increasing evidence indicates high brain expression of pro-inflammatory cytokines and the presence of circulating antibodies against brain proteins. A number of papers, mostly based on parental reporting on their children's health problems, suggest that ASD children may present with "allergic-like" problems in the absence of elevated serum IgE and chronic urticaria. These findings suggest non-allergic mast cell activation, probably in response to environmental and stress triggers that could contribute to inflammation. In utero inflammation can lead to preterm labor and has itself been strongly associated with adverse neurodevelopmental outcomes. Premature babies have about four times higher risk of developing ASD and are also more vulnerable to infections, while delayed development of their gut-blood-brain barriers makes exposure to potential neurotoxins likely. Perinatal mast cell activation by infectious, stress-related, environmental or allergic triggers can lead to release of pro-inflammatory and neurotoxic molecules, thus contributing to brain inflammation and ASD pathogenesis, at least in a subgroup of ASD patients. This article is part of a Special Issue entitled: Mast cells in inflammation.


Asunto(s)
Trastornos Generalizados del Desarrollo Infantil/inmunología , Inflamación/inmunología , Mastocitos/inmunología , Animales , Ansiedad/complicaciones , Barrera Hematoencefálica/patología , Niño , Trastornos Generalizados del Desarrollo Infantil/epidemiología , Trastornos Generalizados del Desarrollo Infantil/etiología , Trastornos Generalizados del Desarrollo Infantil/patología , Estudios Transversales , Humanos , Hipersensibilidad/complicaciones , Hipersensibilidad/inmunología , Hipersensibilidad/patología , Inflamación/complicaciones , Inflamación/patología , Prevalencia , Estrés Psicológico/complicaciones
13.
Biochim Biophys Acta ; 1822(1): 21-33, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21185371

RESUMEN

Mast cells are well known for their role in allergic and anaphylactic reactions, as well as their involvement in acquired and innate immunity. Increasing evidence now implicates mast cells in inflammatory diseases where they are activated by non-allergic triggers, such as neuropeptides and cytokines, often exerting synergistic effects as in the case of IL-33 and neurotensin. Mast cells can also release pro-inflammatory mediators selectively without degranulation. In particular, IL-1 induces selective release of IL-6, while corticotropin-releasing hormone secreted under stress induces the release of vascular endothelial growth factor. Many inflammatory diseases involve mast cells in cross-talk with T cells, such as atopic dermatitis, psoriasis and multiple sclerosis, which all worsen by stress. How mast cell differential responses are regulated is still unresolved. Preliminary evidence suggests that mitochondrial function and dynamics control mast cell degranulation, but not selective release. Recent findings also indicate that mast cells have immunomodulatory properties. Understanding selective release of mediators could explain how mast cells participate in numerous diverse biologic processes, and how they exert both immunostimulatory and immunosuppressive actions. Unraveling selective mast cell secretion could also help develop unique mast cell inhibitors with novel therapeutic applications. This article is part of a Special Issue entitled: Mast cells in inflammation.


Asunto(s)
Inflamación/patología , Mastocitos/metabolismo , Animales , Hormona Liberadora de Corticotropina/metabolismo , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Mediadores de Inflamación/metabolismo , Mastocitos/inmunología , Mastocitos/patología , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/patología , Enfermedades de la Piel/inmunología , Enfermedades de la Piel/patología , Estrés Fisiológico
14.
Proc Natl Acad Sci U S A ; 107(9): 4448-53, 2010 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-20160089

RESUMEN

The peptide substance P (SP) has been implicated in inflammatory conditions, such as psoriasis, where mast cells and VEGF are increased. A relationship between SP and VEGF has not been well studied, nor has any interaction with the proinflammatory cytokines, especially IL-33. Here we report that SP (0.1-10 microM) induces gene expression and secretion of VEGF from human LAD2 mast cells and human umbilical core blood-derived cultured mast cells (hCBMCs). This effect is significantly increased by coadministration of IL-33 (5-100 ng/mL) in both cell types. The effect of SP on VEGF release is inhibited by treatment with the NK-1 receptor antagonist 733,060. SP rapidly increases cytosolic calcium, and so does IL-33 to a smaller extent; the addition of IL-33 augments the calcium increase. SP-induced VEGF production involves calcium-dependent PKC isoforms, as well as the ERK and JNK MAPKs. Gene expression of IL-33 and histidine decarboxylase (HDC), an indicator of mast cell presence/activation, is significantly increased in affected and unaffected (at least 15 cm away from the lesion) psoriatic skin, as compared with normal control skin. Immunohistochemistry indicates that IL-33 is associated with endothelial cells in both the unaffected and affected sites, but is stronger and also associated with immune cells in the affected site. These results imply that functional interactions among SP, IL-33, and mast cells leading to VEGF release contribute to inflammatory conditions, such as the psoriasis, a nonallergic hyperproliferative skin inflammatory disorder with a neurogenic component.


Asunto(s)
Interleucinas/farmacología , Mastocitos/metabolismo , Psoriasis/metabolismo , Piel/efectos de los fármacos , Sustancia P/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Calcio/metabolismo , Citosol/metabolismo , Humanos , Inmunohistoquímica , Interleucina-33 , Antagonistas del Receptor de Neuroquinina-1 , Piperidinas/farmacología , ARN Mensajero/genética , Piel/metabolismo , Sustancia P/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética
15.
JCI Insight ; 8(1)2023 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-36454643

RESUMEN

Dysfunction of alveolar epithelial type 2 cells (AEC2s), the facultative progenitors of lung alveoli, is implicated in pulmonary disease pathogenesis, highlighting the importance of human in vitro models. However, AEC2-like cells in culture have yet to be directly compared to their in vivo counterparts at single-cell resolution. Here, we performed head-to-head comparisons among the transcriptomes of primary (1°) adult human AEC2s, their cultured progeny, and human induced pluripotent stem cell-derived AEC2s (iAEC2s). We found each population occupied a distinct transcriptomic space with cultured AEC2s (1° and iAEC2s) exhibiting similarities to and differences from freshly purified 1° cells. Across each cell type, we found an inverse relationship between proliferative and maturation states, with preculture 1° AEC2s being most quiescent/mature and iAEC2s being most proliferative/least mature. Cultures of either type of human AEC2s did not generate detectable alveolar type 1 cells in these defined conditions; however, a subset of iAEC2s cocultured with fibroblasts acquired a transitional cell state described in mice and humans to arise during fibrosis or following injury. Hence, we provide direct comparisons of the transcriptomic programs of 1° and engineered AEC2s, 2 in vitro models that can be harnessed to study human lung health and disease.


Asunto(s)
Células Madre Pluripotentes Inducidas , Humanos , Animales , Ratones , Transcriptoma , Células Epiteliales Alveolares/metabolismo , Pulmón/patología , Alveolos Pulmonares/patología
16.
bioRxiv ; 2023 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-36711505

RESUMEN

In the distal lung, alveolar epithelial type I cells (AT1s) comprise the vast majority of alveolar surface area and are uniquely flattened to allow the diffusion of oxygen into the capillaries. This structure along with a quiescent, terminally differentiated phenotype has made AT1s particularly challenging to isolate or maintain in cell culture. As a result, there is a lack of established models for the study of human AT1 biology, and in contrast to alveolar epithelial type II cells (AT2s), little is known about the mechanisms regulating their differentiation. Here we engineer a human in vitro AT1 model system through the directed differentiation of induced pluripotent stem cells (iPSC). We first define the global transcriptomes of primary adult human AT1s, suggesting gene-set benchmarks and pathways, such as Hippo-LATS-YAP/TAZ signaling, that are enriched in these cells. Next, we generate iPSC-derived AT2s (iAT2s) and find that activating nuclear YAP signaling is sufficient to promote a broad transcriptomic shift from AT2 to AT1 gene programs. The resulting cells express a molecular, morphologic, and functional phenotype reminiscent of human AT1 cells, including the capacity to form a flat epithelial barrier which produces characteristic extracellular matrix molecules and secreted ligands. Our results indicate a role for Hippo-LATS-YAP signaling in the differentiation of human AT1s and demonstrate the generation of viable AT1-like cells from iAT2s, providing an in vitro model of human alveolar epithelial differentiation and a potential source of human AT1s that until now have been challenging to viably obtain from patients.

17.
Int Arch Allergy Immunol ; 159(1): 23-32, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22555146

RESUMEN

BACKGROUND: Mast cells are immune cells derived from hematopoietic precursors that mature in the tissue microenvironment. Mast cells are critical for allergic, immune and inflammatory processes, many of which involve tumor necrosis factor (TNF). These cells uniquely store TNF in their secretory granules. Upon stimulation, mast cells rapidly (30 min) secrete ß-hexosaminidase and granule-stored TNF through degranulation, but also increase TNF mRNA and release de novo synthesized TNF 24 h later. The regulation of these two distinct pathways is poorly understood. METHODS: Human LAD2 leukemic mast cells are stimulated by substance P. TNF secretion and gene expression were measured by ELISA and real-time PCR, and mitochondrial dynamics was observed in live cells under confocal microscopy. Cell energy consumption was measured in terms of oxygen consumption rate. RESULTS: Here, we show that granule-stored TNF is preformed, and its secretion from LAD2 mast cells stimulated by substance P (1) exhibits higher energy consumption and is inhibited by the mitochondrial ATP pump blocker oligomycin, (2) shows rapid increase in intracellular calcium levels, and (3) exhibits reversible mitochondrial translocation, from a perinuclear distribution to the cell surface, as compared to de novo synthesized TNF release induced by lipopolysaccharide. This mitochondrial translocation is confirmed using primary human umbilical cord blood-derived mast cells stimulated by an allergic trigger (IgE/streptavidin). CONCLUSION: Our findings indicate that unique mitochondrial functions distinguish granule-stored from newly synthesized TNF release from human mast cells, thus permitting the versatile involvement of mast cells in different biological processes.


Asunto(s)
Mastocitos/fisiología , Mitocondrias/inmunología , Vesículas Secretoras/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Calcio/metabolismo , Degranulación de la Célula/efectos de los fármacos , Línea Celular Tumoral , Células Cultivadas , Humanos , Lipopolisacáridos/farmacología , Mastocitos/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Oligomicinas/farmacología , ARN Mensajero/metabolismo , Sustancia P/farmacología , Factor de Necrosis Tumoral alfa/genética , Desacopladores/farmacología
18.
Ann Allergy Asthma Immunol ; 109(1): 14-9, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22727152

RESUMEN

OBJECTIVE: To review the available evidence linking stress to asthma and to investigate whether mast cells contribute to the effect of stress through activation by corticotropin-releasing hormone (CRH). DATA SOURCE: The PubMed database was searched for articles (1998-2011) using the keywords anxiety, asthma, exacerbation, inflammation, mast cells, socioeconomic status, stress, violence, and worsening. STUDY SELECTION: Articles were selected based on their relevance to the topic, with emphasis on clinical or epidemiologic data linking stress to asthma and studies that offered possible explanations for how stress may affect asthma. RESULTS: Many articles point to an association between stress (socioeconomic status, interpersonal conflicts, emotional distress, terrorism) and asthma exacerbations but without any distinct pathogenetic mechanism. A few articles have reported reduced circulating cortisol and/or sensitivity to corticosteroids. We propose that mast cells, known to be involved in the pathophysiology of asthma, can be activated by CRH, which is secreted under stress in the lungs, leading to selective release of proinflammatory mediators. This effect may be augmented by neuropeptides or cytokines. CRH also reduces T-regulatory cell production of interleukin 10, which in known to inhibit allergic mast cell activation. CONCLUSION: More studies are required to investigate lung levels of CRH and selective mast cell mediators. Reducing stress and using CRH receptor antagonists and/or mast cell blockers may serve as possible new therapeutic approaches for asthma.


Asunto(s)
Asma/inmunología , Hormona Liberadora de Corticotropina/inmunología , Hormona Liberadora de Corticotropina/metabolismo , Mastocitos/inmunología , Estrés Psicológico/inmunología , Adolescente , Adulto , Asma/etiología , Asma/terapia , Niño , Preescolar , Hormona Liberadora de Corticotropina/análisis , Femenino , Humanos , Hipersensibilidad/inmunología , Interleucina-10/inmunología , Interleucina-10/metabolismo , Pulmón/química , Masculino , Mastocitos/metabolismo , Neuropéptidos/inmunología , Neuropéptidos/metabolismo , Receptores de Hormona Liberadora de Corticotropina/antagonistas & inhibidores , Estrés Psicológico/complicaciones , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo
19.
BMC Pediatr ; 12: 89, 2012 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-22747567

RESUMEN

BACKGROUND: Autism Spectrum Disorders (ASD) are neurodevelopmental disorders characterized by varying deficits in social interactions, communication, and learning, as well as stereotypic behaviors. Despite the significant increase in ASD, there are few if any clues for its pathogenesis, hampering early detection or treatment. Premature babies are also more vulnerable to infections and inflammation leading to neurodevelopmental problems and higher risk of developing ASD. Many autism "susceptibility" genes have been identified, but "environmental" factors appear to play a significant role. Increasing evidence suggests that there are different ASD endophenotypes. DISCUSSION: We review relevant literature suggesting in utero inflammation can lead to preterm labor, while insufficient development of the gut-blood-brain barriers could permit exposure to potential neurotoxins. This risk apparently may increase in parents with "allergic" or autoimmune problems during gestation, or if they had been exposed to stressors. The presence of circulating auto-antibodies against fetal brain proteins in mothers is associated with higher risk of autism and suggests disruption of the blood-brain-barrier (BBB). A number of papers have reported increased brain expression or cerebrospinal fluid (CSF) levels of pro-inflammatory cytokines, especially TNF, which is preformed in mast cells. Recent evidence also indicates increased serum levels of the pro-inflammatory mast cell trigger neurotensin (NT), and of extracellular mitochondrial DNA (mtDNA), which is immunogenic. Gene mutations of phosphatase and tensin homolog (PTEN), the negative regulator of the mammalian target of rapamycin (mTOR), have been linked to higher risk of autism, but also to increased proliferation and function of mast cells. SUMMARY: Premature birth and susceptibility genes may make infants more vulnerable to allergic, environmental, infectious, or stress-related triggers that could stimulate mast cell release of pro-inflammatory and neurotoxic molecules, thus contributing to brain inflammation and ASD pathogenesis, at least in an endophenotype of ASD patients.


Asunto(s)
Trastorno Autístico/etiología , Encefalitis/complicaciones , Enfermedades Fetales , Nacimiento Prematuro , Estrés Fisiológico , Femenino , Humanos , Recién Nacido , Embarazo , Factores de Riesgo
20.
J Allergy Clin Immunol ; 127(6): 1522-31.e8, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21453958

RESUMEN

BACKGROUND: Mast cells derive from hematopoietic cell precursors and participate in tissue allergic, immune, and inflammatory processes. They secrete many mediators, including preformed TNF, in response to allergic, neuropeptide, and environmental triggers. However, regulation of mast cell degranulation is not well understood. OBJECTIVE: We investigated the role of mitochondrial dynamics in degranulation of human cultured mast cells. METHODS: Human umbilical cord blood-derived mast cells (hCBMCs) and Laboratory of Allergic Diseases 2 (LAD2) mast cells were examined by confocal and differential interference contrast microscopy during activation by IgE/antigen and substance P (SP). Mast cells in control and atopic dermatitis (AD) skin were evaluated by transmission electron microscopy. LAD2 cells were pretreated with mitochondrial division inhibitor, a dynamin-related protein 1 (Drp1) inhibitor, and small interfering RNA for Drp1, which is necessary for mitochondrial fission and translocation. Calcineurin and Drp1 gene expression was analyzed in stimulated LAD2 cells and AD skin biopsies. RESULTS: Stimulation of hCBMCs with IgE/antigen or LAD2 cells with SP leads to rapid (30 minutes) secretion of preformed TNF. Degranulation is accompanied by mitochondrial translocation from a perinuclear location to exocytosis sites. Extracellular calcium depletion prevents these effects, indicating calcium requirement. The calcium-dependent calcineurin and Drp1 are activated 30 minutes after SP stimulation. Reduction of Drp1 activity by mitochondrial division inhibitor and decrease of Drp1 expression using small interfering RNA inhibit mitochondrial translocation, degranulation, and TNF secretion. Mitochondrial translocation is also evident by transmission electron microscopy in skin mast cells from AD biopsies, in which gene expression of calcineurin, Drp1, and SP is higher than in normal skin. CONCLUSION: Human mast cell degranulation requires mitochondrial dynamics, also implicated in AD.


Asunto(s)
Degranulación de la Célula/fisiología , Dermatitis Atópica/fisiopatología , Mastocitos/fisiología , Factor de Necrosis Tumoral alfa/fisiología , Adolescente , Adulto , Antígenos/administración & dosificación , Transporte Biológico Activo , Calcineurina/genética , Calcineurina/metabolismo , Calcio/metabolismo , Estudios de Casos y Controles , Degranulación de la Célula/efectos de los fármacos , Degranulación de la Célula/inmunología , Células Cultivadas , Niño , Dermatitis Atópica/genética , Dermatitis Atópica/inmunología , Dinaminas , Exocitosis/fisiología , Femenino , GTP Fosfohidrolasas/antagonistas & inhibidores , GTP Fosfohidrolasas/genética , GTP Fosfohidrolasas/metabolismo , Humanos , Inmunoglobulina E/administración & dosificación , Masculino , Mastocitos/efectos de los fármacos , Mastocitos/inmunología , Mastocitos/ultraestructura , Microscopía Electrónica de Transmisión , Proteínas Asociadas a Microtúbulos/antagonistas & inhibidores , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Persona de Mediana Edad , Mitocondrias/fisiología , Proteínas Mitocondriales/antagonistas & inhibidores , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , ARN Interferente Pequeño/genética , Sustancia P/administración & dosificación , Sustancia P/genética , Adulto Joven
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda