Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Semin Cell Dev Biol ; 115: 70-76, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33158728

RESUMEN

Cadmium (Cd) is a transition metal, also referred to as a heavy metal, that is naturally abundant in the earth's crust. It has no known benefit to humans. It is primarily released into our environment through mining and smelting in industrial processes and enters the food chain through uptake by plants from contaminated soil and water. In humans, Cd primarily enters the body through ingestion of foods and cigarette smoke and has an extremely long resident half-life in the body compared to other transition metals. Environmental workplace exposure is also a source through inhalation, although much less common. The principal organs adversely affected by Cd following acute and chronic exposure are the kidneys, bone, vasculature and lung. Cd adversely impacts cell function through changes in gene expression and signal transduction and is recognized as a carcinogen. Despite a substantial body of mechanistic studies in cells and animal models, the overall impact of Cd on innate immune function in humans remains poorly understood. The best evidence is perhaps alteration of reactive oxygen species balance and signaling in cells that regulate innate immunity causing alteration of the inflammatory response that is postulated to contribute to chronic diseases. Epidemiologic studies support this possibility since increased tissue levels in humans are strongly associated with leading chronic diseases including chronic obstructive pulmonary disease (COPD), which will be discussed in depth. Additional studies are required to understand how chronic exposure and accumulation of this leading environmental toxicant in vital organs negatively impact innate immune function and host defense leading to chronic disease in humans.


Asunto(s)
Cadmio/efectos adversos , Inmunidad Innata/efectos de los fármacos , Pulmón/efectos de los fármacos , Humanos
2.
J Immunol ; 207(5): 1357-1370, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34380651

RESUMEN

Zinc (Zn) is required for proper immune function and host defense. Zn homeostasis is tightly regulated by Zn transporters that coordinate biological processes through Zn mobilization. Zn deficiency is associated with increased susceptibility to bacterial infections, including Streptococcus pneumoniae, the most commonly identified cause of community-acquired pneumonia. Myeloid cells, including macrophages and dendritic cells (DCs), are at the front line of host defense against invading bacterial pathogens in the lung and play a critical role early on in shaping the immune response. Expression of the Zn transporter ZIP8 is rapidly induced following bacterial infection and regulates myeloid cell function in a Zn-dependent manner. To what extent ZIP8 is instrumental in myeloid cell function requires further study. Using a novel, myeloid-specific, Zip8 knockout model, we identified vital roles of ZIP8 in macrophage and DC function upon pneumococcal infection. Administration of S. pneumoniae into the lung resulted in increased inflammation, morbidity, and mortality in Zip8 knockout mice compared with wild-type counterparts. This was associated with increased numbers of myeloid cells, cytokine production, and cell death. In vitro analysis of macrophage and DC function revealed deficits in phagocytosis and increased cytokine production upon bacterial stimulation that was, in part, due to increased NF-κB signaling. Strikingly, alteration of myeloid cell function resulted in an imbalance of Th17/Th2 responses, which is potentially detrimental to host defense. These results (for the first time, to our knowledge) reveal a vital ZIP8- and Zn-mediated axis that alters the lung myeloid cell landscape and the host response against pneumococcus.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Células Dendríticas/inmunología , Macrófagos/inmunología , Células Mieloides/inmunología , Neumonía Neumocócica/inmunología , Streptococcus pneumoniae/fisiología , Células Th17/inmunología , Células Th2/inmunología , Animales , Proteínas de Transporte de Catión/genética , Células Cultivadas , Citocinas/metabolismo , Modelos Animales de Enfermedad , Humanos , Inmunidad Innata , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/metabolismo , Fagocitosis/genética , Transducción de Señal
3.
Int J Mol Sci ; 23(3)2022 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-35162945

RESUMEN

Pneumococcal pneumonia is a leading cause of morbidity and mortality worldwide. An increased susceptibility is due, in part, to compromised immune function. Zinc is required for proper immune function, and an insufficient dietary intake increases the risk of pneumonia. Our group was the first to reveal that the Zn transporter, ZIP8, is required for host defense. Furthermore, the gut microbiota that is essential for lung immunity is adversely impacted by a commonly occurring defective ZIP8 allele in humans. Taken together, we hypothesized that loss of the ZIP8 function would lead to intestinal dysbiosis and impaired host defense against pneumonia. To test this, we utilized a novel myeloid-specific Zip8KO mouse model in our studies. The comparison of the cecal microbial composition of wild-type and Zip8KO mice revealed significant differences in microbial community structure. Most strikingly, upon a S. pneumoniae lung infection, mice recolonized with Zip8KO-derived microbiota exhibited an increase in weight loss, bacterial dissemination, and lung inflammation compared to mice recolonized with WT microbiota. For the first time, we reveal the critical role of myeloid-specific ZIP8 on the maintenance of the gut microbiome structure, and that loss of ZIP8 leads to intestinal dysbiosis and impaired host defense in the lung. Given the high incidence of dietary Zn deficiency and the ZIP8 variant allele in the human population, additional investigation is warranted to improve surveillance and treatment strategies.


Asunto(s)
Bacterias/clasificación , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Disbiosis/metabolismo , Pulmón/microbiología , Neumonía Neumocócica/metabolismo , Streptococcus pneumoniae/patogenicidad , Animales , Bacterias/genética , ADN Bacteriano/genética , ADN Ribosómico/genética , Modelos Animales de Enfermedad , Disbiosis/genética , Femenino , Microbioma Gastrointestinal , Técnicas de Inactivación de Genes , Secuenciación de Nucleótidos de Alto Rendimiento , Pulmón/metabolismo , Ratones , Neumonía Neumocócica/microbiología , ARN Ribosómico 16S/genética , Análisis de Secuencia de ADN , Zinc/metabolismo
4.
Int J Mol Sci ; 18(11)2017 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-29120360

RESUMEN

Tuberculosis (TB) is a global epidemic caused by the infection of human macrophages with the world's most deadly single bacterial pathogen, Mycobacterium tuberculosis (M.tb). M.tb resides in a phagosomal niche within macrophages, where trace element concentrations impact the immune response, bacterial metal metabolism, and bacterial survival. The manipulation of micronutrients is a critical mechanism of host defense against infection. In particular, the human zinc transporter Zrt-/Irt-like protein 8 (ZIP8), one of 14 ZIP family members, is important in the flux of divalent cations, including zinc, into the cytoplasm of macrophages. It also has been observed to exist on the membrane of cellular organelles, where it can serve as an efflux pump that transports zinc into the cytosol. ZIP8 is highly inducible in response to M.tb infection of macrophages, and we have observed its localization to the M.tb phagosome. The expression, localization, and function of ZIP8 and other divalent cation transporters within macrophages have important implications for TB prevention and dissemination and warrant further study. In particular, given the importance of zinc as an essential nutrient required for humans and M.tb, it is not yet clear whether ZIP-guided zinc transport serves as a host protective factor or, rather, is targeted by M.tb to enable its phagosomal survival.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Interacciones Huésped-Patógeno , Macrófagos/inmunología , Mycobacterium tuberculosis/inmunología , Fagosomas/metabolismo , Tuberculosis/inmunología , Zinc/metabolismo , Citosol/metabolismo , Humanos , Inmunidad Innata , Macrófagos/metabolismo , Tuberculosis/microbiología
5.
Am J Physiol Lung Cell Mol Physiol ; 311(4): L754-L765, 2016 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-27496894

RESUMEN

Chronic obstructive pulmonary disease (COPD) in the U.S. is primarily caused by cigarette smoking. COPD patients are highly susceptible to respiratory infections in part due to alveolar macrophage dysfunction despite a substantial increase in macrophages in the lung. Cadmium (Cd) is a toxic metal that is concentrated within tobacco and accumulates in the lung of smokers. We hypothesized that Cd uptake into macrophages alters immune function thereby impairing the macrophage response to invading pathogens. Our hypothesis was tested by comparing primary human monocytes and macrophages, primary mouse bronchoalveolar lavage myeloid cells, and related cell lines. Strikingly, Cd exposure followed by LPS stimulation resulted in a dose-dependent, significant decrease in nuclear p65 activity in macrophages that was not observed in monocytes. This corresponded with Cd-mediated inhibition of IKKß and an impaired ability to transcribe and release cytokines in response to LPS challenge in vivo. These findings provide novel evidence that Cd has the capacity to disrupt macrophage immune function compared with monocytes. Importantly, Cd results in immune dysfunction in macrophages through inhibition of the NF-κB signaling pathway. Based on these findings, we provide new evidence that Cd contributes to immune dysfunction in the lung of COPD subjects and may increase susceptibility to infection.


Asunto(s)
Cadmio/toxicidad , Lipopolisacáridos/farmacología , Macrófagos Alveolares/metabolismo , FN-kappa B/metabolismo , Animales , Línea Celular , Polaridad Celular , Citocinas/biosíntesis , Citocinas/genética , Femenino , Humanos , Macrófagos Alveolares/efectos de los fármacos , Macrófagos Alveolares/inmunología , Metalotioneína/biosíntesis , Metalotioneína/genética , Ratones Endogámicos C57BL , Enfermedad Pulmonar Obstructiva Crónica/etiología , Enfermedad Pulmonar Obstructiva Crónica/inmunología , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Transducción de Señal , Fumar/efectos adversos , Activación Transcripcional
6.
J Biol Chem ; 288(6): 3691-5, 2013 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-23269671

RESUMEN

Burkholderia cenocepacia, the causative agent of cepacia syndrome, primarily affects cystic fibrosis patients, often leading to death. In the lung, epithelial cells serve as the initial barrier to airway infections, yet their responses to B. cenocepacia have not been fully investigated. Here, we examined the molecular responses of human airway epithelial cells to B. cenocepacia infection. Infection led to early signaling events such as activation of Erk, Akt, and NF-κB. Further, TNFα, IL-6, IL-8, and IL-1ß were all significantly induced upon infection, but no IL-1ß was detected in the supernatants. Because caspase-1 is required for IL-1ß processing and release, we examined its expression in airway epithelial cells. Interestingly, little to no caspase-1 was detectable in airway epithelial cells. Transfection of caspase-1 into airway epithelial cells restored their ability to secrete IL-1ß following B. cenocepacia infection, suggesting that a deficiency in caspase-1 is responsible, at least in part, for the attenuated IL-1ß secretion.


Asunto(s)
Bronquios/metabolismo , Infecciones por Burkholderia/metabolismo , Burkholderia cenocepacia , Células Epiteliales/metabolismo , Interleucina-1beta/metabolismo , Mucosa Respiratoria/metabolismo , Bronquios/microbiología , Bronquios/patología , Infecciones por Burkholderia/genética , Infecciones por Burkholderia/microbiología , Infecciones por Burkholderia/patología , Caspasa 1/biosíntesis , Caspasa 1/genética , Línea Celular , Citocinas/biosíntesis , Citocinas/genética , Células Epiteliales/microbiología , Células Epiteliales/patología , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Interleucina-1beta/genética , FN-kappa B/genética , FN-kappa B/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Mucosa Respiratoria/microbiología , Mucosa Respiratoria/patología , Transfección
7.
J Immunol ; 189(1): 433-43, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-22675199

RESUMEN

Plasmacytoid dendritic cells (pDC) are potent APCs known to regulate immune responses to self-Ags, particularly DNA. The mitochondrial fraction of necrotic cells was found to most potently promote human pDC activation, as reflected by type I IFN release, which was dependent upon the presence of mitochondrial DNA and involved TLR9 and receptors for advanced glycation end products. Mitochondrial transcription factor A (TFAM), a highly abundant mitochondrial protein that is functionally and structurally homologous to high mobility group box protein 1, was observed to synergize with CpG-containing oligonucleotide, type A, DNA to promote human pDC activation. pDC type I IFN responses to TFAM and CpG-containing oligonucleotide, type A, DNA indicated their engagement with receptors for advanced glycation end products and TLR9, respectively, and were dependent upon endosomal processing and PI3K, ERK, and NF-κB signaling. Taken together, these results indicate that pDC contribute to sterile immune responses by recognizing the mitochondrial component of necrotic cells and further incriminate TFAM and mitochondrial DNA as likely mediators of pDC activation under these circumstances.


Asunto(s)
Adyuvantes Inmunológicos/fisiología , Islas de CpG/inmunología , ADN Mitocondrial/genética , Proteínas de Unión al ADN/fisiología , Células Dendríticas/inmunología , Células Dendríticas/patología , Proteínas Mitocondriales/fisiología , Transducción de Señal/inmunología , Factores de Transcripción/fisiología , Adyuvantes Inmunológicos/genética , Adyuvantes Inmunológicos/metabolismo , Animales , Islas de CpG/genética , Proteínas de Unión al ADN/genética , Células Dendríticas/metabolismo , Amplificación de Genes/inmunología , Células Hep G2 , Humanos , Interferón-alfa/metabolismo , Ratones , Proteínas Mitocondriales/genética , Necrosis , Procesamiento Proteico-Postraduccional/genética , Procesamiento Proteico-Postraduccional/inmunología , Receptor para Productos Finales de Glicación Avanzada , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Transducción de Señal/genética , Receptor Toll-Like 9/fisiología , Factores de Transcripción/genética
8.
Int J Toxicol ; 33(3): 246-258, 2014 05.
Artículo en Inglés | MEDLINE | ID: mdl-24728862

RESUMEN

Mouse Slc39a8 and Slc39a14 genes encode ZIP8 and ZIP14, respectively, which are ubiquitous divalent cation/(HCO3-)2 symporters responsible for uptake of Zn2+, Fe2+, and Mn2+ into cells. Cd2+ and other toxic nonessential metals can displace essential cations, thereby entering vertebrate cells. Whereas Slc39a8 encodes a single protein, Slc39a14 has 2 exons 4 which, via alternative splicing, give rise to ZIP14A and ZIP14B; why differences exist in cell type-specific expression of ZIP14A and ZIP14B remains unknown. Inflammatory stimuli have been associated with upregulation of ZIP8 and ZIP14, but a systematic study of many tissues simultaneously in a laboratory animal following inflammatory cytokine exposure has not yet been reported. Herein, we show that C57BL/6J male mice--treated intraperitoneally with lipopolysaccharide or the proinflammatory cytokines tumor necrosis factor (TNF) or interleukin-6 (IL6)--exhibited quantatively very different, highly tissue-specific, and markedly time-dependent up- and downregulation of ZIP8, ZIP14A, and ZIP14B messenger RNA (mRNA) levels in 12 tissues. The magnitude of inflammatory response was confirmed by measuring the proinflammatory cytokine TNF, IL6, and interleukin-1ß mRNA levels in the same tissues of these animals. Our data suggest that most if not all tissues use ZIP8, ZIP14A, and/or ZIP14B for Zn2+ uptake, some tissues under basal conditions and others moreso when inflammatory stressors are present; collectively, this might lead to substantial alterations in plasma Zn2+ levels due to Zn2+ redistribution not just in liver but across many vital organs. In the context of cadmium-mediated toxicity, our data suggest that tissues other than liver, kidney, and lung should also be considered.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Modelos Animales de Enfermedad , Endotoxemia/metabolismo , Regulación de la Expresión Génica , Hígado/metabolismo , Zinc/metabolismo , Empalme Alternativo , Animales , Proteínas de Transporte de Catión/genética , Citocinas/metabolismo , Regulación hacia Abajo , Endotoxemia/sangre , Endotoxemia/inmunología , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-6/administración & dosificación , Interleucina-6/genética , Interleucina-6/metabolismo , Cinética , Lipopolisacáridos/administración & dosificación , Hígado/inmunología , Masculino , Ratones Endogámicos C57BL , Especificidad de Órganos , ARN Mensajero/metabolismo , Factor de Necrosis Tumoral alfa/administración & dosificación , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Regulación hacia Arriba , Zinc/sangre
9.
Am J Respir Cell Mol Biol ; 49(3): 368-83, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23590305

RESUMEN

In this study, a genetically diverse panel of 43 mouse strains was exposed to phosgene and genome-wide association mapping performed using a high-density single nucleotide polymorphism (SNP) assembly. Transcriptomic analysis was also used to improve the genetic resolution in the identification of genetic determinants of phosgene-induced acute lung injury (ALI). We prioritized the identified genes based on whether the encoded protein was previously associated with lung injury or contained a nonsynonymous SNP within a functional domain. Candidates were selected that contained a promoter SNP that could alter a putative transcription factor binding site and had variable expression by transcriptomic analyses. The latter two criteria also required that ≥10% of mice carried the minor allele and that this allele could account for ≥10% of the phenotypic difference noted between the strains at the phenotypic extremes. This integrative, functional approach revealed 14 candidate genes that included Atp1a1, Alox5, Galnt11, Hrh1, Mbd4, Phactr2, Plxnd1, Ptprt, Reln, and Zfand4, which had significant SNP associations, and Itga9, Man1a2, Mapk14, and Vwf, which had suggestive SNP associations. Of the genes with significant SNP associations, Atp1a1, Alox5, Plxnd1, Ptprt, and Zfand4 could be associated with ALI in several ways. Using a competitive electrophoretic mobility shift analysis, Atp1a1 promoter (rs215053185) oligonucleotide containing the minor G allele formed a major distinct faster-migrating complex. In addition, a gene with a suggestive SNP association, Itga9, is linked to transforming growth factor ß1 signaling, which previously has been associated with the susceptibility to ALI in mice.


Asunto(s)
Lesión Pulmonar Aguda/genética , Sustancias para la Guerra Química/toxicidad , Expresión Génica/efectos de los fármacos , Genoma , Pulmón/metabolismo , Fosgeno/toxicidad , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/metabolismo , Lesión Pulmonar Aguda/patología , Alelos , Animales , Mapeo Cromosómico , Ensayo de Cambio de Movilidad Electroforética , Femenino , Perfilación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Genómica , Genotipo , Integrinas/genética , Integrinas/metabolismo , Pulmón/efectos de los fármacos , Pulmón/patología , Ratones , Ratones Endogámicos , Análisis de Secuencia por Matrices de Oligonucleótidos , Polimorfismo de Nucleótido Simple , Regiones Promotoras Genéticas , Proteína Reelina , ATPasa Intercambiadora de Sodio-Potasio/genética , ATPasa Intercambiadora de Sodio-Potasio/metabolismo
10.
J Nutr ; 143(7): 1036-45, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23700340

RESUMEN

Zinc (Zn) deficiency and obesity are global public health problems. Zn deficiency is associated with obesity and comorbid conditions that include insulin resistance and type 2 diabetes. However, the function of Zn in obesity remains unclear. Using a mouse model of combined high-fat and low-Zn intake (0.5-1.5 mg/kg), we investigated whether Zn deficiency exacerbates the extent of adiposity as well as perturbations in metabolic and immune function. C57BL/6 mice were randomly assigned to receive either a high-fat diet (HFD) or a control (C) diet for 6 wk, followed by further subdivision into 2 additional groups fed Zn-deficient diets (C-Zn, HFD-Zn), along with a C diet and an HFD, for 3 wk (n = 8-9 mice/group). The extent of visceral fat, insulin resistance, or systemic inflammation was unaffected by Zn deficiency. Strikingly, Zn deficiency significantly augmented circulating leptin concentrations (HFD-Zn vs. HFD: 3.15 ± 0.16 vs. 2.59 ± 0.12 µg/L, respectively) and leptin signaling in the liver of obese mice. Furthermore, gene expression of macrophage-specific markers ADAM8 (A disintegrin and metalloproteinase domain-containing protein 8) and CD68 (cluster of differentiation 68) was significantly greater in adipose tissue in the HFD-Zn group than in the HFD group, as confirmed by CD68 protein analysis, indicative of increased macrophage infiltration. Inspection of Zn content and mRNA profiles of all Zn transporters in the adipose tissue revealed alterations of Zn metabolism to obesity and Zn deficiency. Our results demonstrate that Zn deficiency increases leptin production and exacerbates macrophage infiltration into adipose tissue in obese mice, indicating the importance of Zn in metabolic and immune dysregulation in obesity.


Asunto(s)
Dieta Alta en Grasa , Grasa Intraabdominal/metabolismo , Leptina/biosíntesis , Macrófagos/metabolismo , Zinc/sangre , Zinc/deficiencia , Adipoquinas/sangre , Adiposidad , Animales , Antígenos CD/genética , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/genética , Antígenos de Diferenciación Mielomonocítica/metabolismo , Biomarcadores/sangre , Western Blotting , Citocinas/sangre , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/fisiopatología , Inmunohistoquímica , Inflamación/metabolismo , Inflamación/fisiopatología , Resistencia a la Insulina , Hígado/metabolismo , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , FN-kappa B/metabolismo , Células 3T3 NIH , Obesidad/fisiopatología , PPAR gamma/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Transfección
11.
Pathogens ; 12(5)2023 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-37242309

RESUMEN

Intestinal dysbiosis increases susceptibility to infection through the alteration of metabolic profiles, which increases morbidity. Zinc (Zn) homeostasis in mammals is tightly regulated by 24 Zn transporters. ZIP8 is unique in that it is required by myeloid cells to maintain proper host defense against bacterial pneumonia. In addition, a frequently occurring ZIP8 defective variant (SLC39A8 rs13107325) is strongly associated with inflammation-based disorders and bacterial infection. In this study, we developed a novel model to study the effects of ZIP8-mediated intestinal dysbiosis on pulmonary host defense independent of the genetic effects. Cecal microbial communities from a myeloid-specific Zip8 knockout mouse model were transplanted into germ-free mice. Conventionalized ZIP8KO-microbiota mice were then bred to produce F1 and F2 generations of ZIP8KO-microbiota mice. F1 ZIP8KO-microbiota mice were also infected with S. pneumoniae, and pulmonary host defense was assessed. Strikingly, the instillation of pneumococcus into the lung of F1 ZIP8KO-microbiota mice resulted in a significant increase in weight loss, inflammation, and mortality when compared to F1 wild-type (WT)-microbiota recipients. Similar defects in pulmonary host defense were observed in both genders, although consistently greater in females. From these results, we conclude that myeloid Zn homeostasis is not only critical for myeloid function but also plays a significant role in the maintenance and control of gut microbiota composition. Further, these data demonstrate that the intestinal microbiota, independent of host genetics, play a critical role in governing host defense in the lung against infection. Finally, these data strongly support future microbiome-based interventional studies, given the high incidence of zinc deficiency and the rs13107325 allele in humans.

12.
Cells ; 12(18)2023 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-37759490

RESUMEN

Preclinical studies have shown that chronic alcohol abuse leads to alterations in the gastrointestinal microbiota that are associated with behavior changes, physiological alterations, and immunological effects. However, such studies have been limited in their ability to evaluate the direct effects of alcohol-associated dysbiosis. To address this, we developed a humanized alcohol-microbiota mouse model to systematically evaluate the immunological effects of chronic alcohol abuse mediated by intestinal dysbiosis. Germ-free mice were colonized with human fecal microbiota from individuals with high and low Alcohol Use Disorders Identification Test (AUDIT) scores and bred to produce human alcohol-associated microbiota or human control-microbiota F1 progenies. F1 offspring colonized with fecal microbiota from individuals with high AUDIT scores had increased susceptibility to Klebsiella pneumoniae and Streptococcus pneumoniae pneumonia, as determined by increased mortality rates, pulmonary bacterial burden, and post-infection lung damage. These findings highlight the importance of considering both the direct effects of alcohol and alcohol-induced dysbiosis when investigating the mechanisms behind alcohol-related disorders and treatment strategies.


Asunto(s)
Alcoholismo , Microbiota , Neumonía Bacteriana , Humanos , Animales , Ratones , Alcoholismo/complicaciones , Disbiosis/complicaciones , Etanol
13.
Am J Respir Cell Mol Biol ; 47(2): 234-44, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22447970

RESUMEN

The genetic basis for the underlying individual susceptibility to chlorine-induced acute lung injury is unknown. To uncover the genetic basis and pathophysiological processes that could provide additional homeostatic capacities during lung injury, 40 inbred murine strains were exposed to chlorine, and haplotype association mapping was performed. The identified single-nucleotide polymorphism (SNP) associations were evaluated through transcriptomic and metabolomic profiling. Using ≥ 10% allelic frequency and ≥ 10% phenotype explained as threshold criteria, promoter SNPs that could eliminate putative transcriptional factor recognition sites in candidate genes were assessed by determining transcript levels through microarray and reverse real-time PCR during chlorine exposure. The mean survival time varied by approximately 5-fold among strains, and SNP associations were identified for 13 candidate genes on chromosomes 1, 4, 5, 9, and 15. Microarrays revealed several differentially enriched pathways, including protein transport (decreased more in the sensitive C57BLKS/J lung) and protein catabolic process (increased more in the resistant C57BL/10J lung). Lung metabolomic profiling revealed 95 of the 280 metabolites measured were altered by chlorine exposure, and included alanine, which decreased more in the C57BLKS/J than in the C57BL/10J strain, and glutamine, which increased more in the C57BL/10J than in the C57BLKS/J strain. Genetic associations from haplotype mapping were strengthened by an integrated assessment using transcriptomic and metabolomic profiling. The leading candidate genes associated with increased susceptibility to acute lung injury in mice included Klf4, Sema7a, Tns1, Aacs, and a gene that encodes an amino acid carrier, Slc38a4.


Asunto(s)
Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/genética , Cloro/farmacología , Animales , Mapeo Cromosómico/métodos , Femenino , Perfilación de la Expresión Génica/métodos , Predisposición Genética a la Enfermedad , Haplotipos , Factor 4 Similar a Kruppel , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Metaboloma , Ratones , Ratones Endogámicos C57BL , Fenotipo , Polimorfismo de Nucleótido Simple , Transcriptoma/genética
14.
Am J Physiol Lung Cell Mol Physiol ; 302(3): L287-99, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22037358

RESUMEN

The phosphoinositide-3 kinase/Akt pathway is a vital survival axis in lung epithelia. We previously reported that inhibition of phosphatase and tensin homolog deleted on chromosome 10 (PTEN), a major suppressor of this pathway, results in enhanced wound repair following injury. However, the precise cellular and biomechanical mechanisms responsible for increased wound repair during PTEN inhibition are not yet well established. Using primary human lung epithelia and a related lung epithelial cell line, we first determined whether changes in migration or proliferation account for wound closure. Strikingly, we observed that cell migration accounts for the majority of wound recovery following PTEN inhibition in conjunction with activation of the Akt and ERK signaling pathways. We then used fluorescence and atomic force microscopy to investigate how PTEN inhibition alters the cytoskeletal and mechanical properties of the epithelial cell. PTEN inhibition did not significantly alter cytoskeletal structure but did result in large spatial variations in cell stiffness and in particular a decrease in cell stiffness near the wound edge. Biomechanical changes, as well as migration rates, were mediated by both the Akt and ERK pathways. Our results indicate that PTEN inhibition rapidly alters biochemical signaling events that in turn provoke alterations in biomechanical properties that enhance cell migration. Specifically, the reduced stiffness of PTEN-inhibited cells promotes larger deformations, resulting in a more migratory phenotype. We therefore conclude that increased wound closure consequent to PTEN inhibition occurs through enhancement of cell migration that is due to specific changes in the biomechanical properties of the cell.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Células Epiteliales/fisiología , Pulmón/citología , Fosfohidrolasa PTEN/antagonistas & inhibidores , Compuestos de Vanadio/farmacología , Cicatrización de Heridas/efectos de los fármacos , Citoesqueleto de Actina/metabolismo , Fenómenos Biomecánicos , Línea Celular , Proliferación Celular , Impedancia Eléctrica , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Lesión Pulmonar/tratamiento farmacológico , Lesión Pulmonar/prevención & control , Sistema de Señalización de MAP Quinasas , Microscopía de Fuerza Atómica , Fosfohidrolasa PTEN/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo
15.
Am J Physiol Lung Cell Mol Physiol ; 302(9): L909-18, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22345571

RESUMEN

Cadmium (Cd), a toxic heavy metal and carcinogen that is abundantly present in cigarette smoke, is a cause of smoking-induced lung disease. SLC39A8 (ZIP8), a zinc transporter, is a major portal for Cd uptake into cells. We have recently identified that ZIP8 expression is under the transcriptional control of the NF-κB pathway. On the basis of this, we hypothesized that cigarette-smoke induced inflammation would increase ZIP8 expression in lung epithelia, thereby enhancing Cd uptake and cell toxicity. Herein we report that ZIP8 is a central mediator of Cd-mediated toxicity. TNF-α treatment of primary human lung epithelia and A549 cells induced ZIP8 expression, resulting in significantly higher cell death attributable to both apoptosis and necrosis following Cd exposure. Inhibition of the NF-κB pathway and ZIP8 expression significantly reduced cell toxicity. Zinc (Zn), a known cytoprotectant, prevented Cd-mediated cell toxicity via ZIP8 uptake. Consistent with cell culture findings, a significant increase in ZIP8 mRNA and protein expression was observed in the lung of chronic smokers compared with nonsmokers. From these studies, we conclude that ZIP8 expression is induced in lung epithelia in an NF-κB-dependent manner, thereby resulting in increased cell death in the presence of Cd. From this we contend that ZIP8 plays a critical role at the interface between micronutrient (Zn) metabolism and toxic metal exposure (Cd) in the lung microenvironment following cigarette smoke exposure. Furthermore, dietary Zn intake, or a lack thereof, may be a contributing factor in smoking-induced lung disease.


Asunto(s)
Cadmio/toxicidad , Proteínas de Transporte de Catión/genética , Células Epiteliales/efectos de los fármacos , Pulmón/patología , FN-kappa B/metabolismo , Activación Transcripcional , Apoptosis/efectos de los fármacos , Cadmio/metabolismo , Proteínas de Transporte de Catión/metabolismo , Línea Celular , Polaridad Celular , Citoprotección , Células Epiteliales/metabolismo , Humanos , FN-kappa B/antagonistas & inhibidores , Necrosis/inducido químicamente , Nitrilos/farmacología , Cultivo Primario de Células , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/patología , Fumar/metabolismo , Sulfonas/farmacología , Factor de Necrosis Tumoral alfa/fisiología , Regulación hacia Arriba , Zinc/metabolismo , Zinc/farmacología
16.
Front Cell Dev Biol ; 10: 924820, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35832795

RESUMEN

Manganese (Mn) and Zinc (Zn) are essential micronutrients whose concentration and location within cells are tightly regulated at the onset of infection. Two families of Zn transporters (ZIPs and ZnTs) are largely responsible for regulation of cytosolic Zn levels and to a certain extent, Mn levels, although much less is known regarding Mn. The capacity of pathogens to persevere also depends on access to micronutrients, yet a fundamental gap in knowledge remains regarding the importance of metal exchange at the host interface, often referred to as nutritional immunity. ZIP8, one of 14 ZIPs, is a pivotal importer of both Zn and Mn, yet much remains to be known. Dietary Zn deficiency is common and commonly occurring polymorphic variants of ZIP8 that decrease cellular metal uptake (Zn and Mn), are associated with increased susceptibility to infection. Strikingly, ZIP8 is the only Zn transporter that is highly induced following bacterial exposure in key immune cells involved with host defense against leading pathogens. We postulate that mobilization of Zn and Mn into key cells orchestrates the innate immune response through regulation of fundamental defense mechanisms that include phagocytosis, signal transduction, and production of soluble host defense factors including cytokines and chemokines. New evidence also suggests that host metal uptake may have long-term consequences by influencing the adaptive immune response. Given that activation of ZIP8 expression by pathogens has been shown to influence parenchymal, myeloid, and lymphoid cells, the impact applies to all mucosal surfaces and tissue compartments that are vulnerable to infection. We also predict that perturbations in metal homeostasis, either genetic- or dietary-induced, has the potential to impact bacterial communities in the host thereby adversely impacting microbiome composition. This review will focus on Zn and Mn transport via ZIP8, and how this vital metal transporter serves as a "go to" conductor of metal uptake that bolsters host defense against pathogens. We will also leverage past studies to underscore areas for future research to better understand the Zn-, Mn- and ZIP8-dependent host response to infection to foster new micronutrient-based intervention strategies to improve our ability to prevent or treat commonly occurring infectious disease.

17.
Adv Sci (Weinh) ; 9(8): e2103676, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34994102

RESUMEN

Local pulmonary administration of therapeutic siRNA represents a promising approach to the treatment of lung fibrosis, which is currently hampered by inefficient delivery. Development of perfluorooctylbromide (PFOB) nanoemulsions as a way of improving the efficiency of pulmonary polycation-based delivery of siRNA is reported. The results show that the polycation/siRNA/PFOB nanoemulsions are capable of efficiently silencing the expression of STAT3 and inhibiting chemokine receptor CXCR4-two validated targets in pulmonary fibrosis. Both in vitro and in vivo results demonstrate that the nanoemulsions improve mucus penetration and facilitate effective cellular delivery of siRNA. Pulmonary treatment of mice with bleomycin-induced pulmonary fibrosis shows strong inhibition of the progression of the disease and significant prolongation of animal survival. Overall, the study points to a promising local treatment strategy of pulmonary fibrosis.


Asunto(s)
Fluorocarburos , Fibrosis Pulmonar , Animales , Bleomicina/efectos adversos , Bleomicina/metabolismo , Fluorocarburos/efectos adversos , Fluorocarburos/metabolismo , Pulmón/metabolismo , Pulmón/patología , Ratones , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/terapia , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , ARN Interferente Pequeño/farmacología
18.
Am J Respir Cell Mol Biol ; 44(3): 264-9, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20889801

RESUMEN

Mitochondrial antigens released from damaged cells act as "danger signals" capable of promoting innate immune cell migration and activation via formyl peptide receptors (FPRs). Lung epithelial cells are equipped to migrate and mount innate immune responses in the context of acute lung injury. The goal of this study was to determine whether lung epithelial cells express FPRs, which are capable of responding to mitochondrial antigens to promote wound closure and inflammation. Using human Beas2B lung epithelial cells grown to confluency and subjected to linear scratch injury, it was found that mitochondrial antigens enhanced epithelial wound closure, and this phenomenon was inhibited by cyclosporin H, a selective inhibitor of FPR. Although mitochondrial antigens also promoted IL-8 release, this release was not FPR dependent and was unrelated to FPR-induced lung epithelial cell wound closure. The expression of functional FPR was confirmed in Beas2B and primary human tracheobronchial epithelial cells, particularly in lamellipodia at the leading edge of the closing wound. The expression of FPR was increased in response to TNF-α, LPS, scratch injury, and mitochondrial antigen treatment. Considered together, these data confirm that human lung epithelial cells express functional FPRs, which are capable of responding to endogenous mitochondrial danger signals, to promote wound closure.


Asunto(s)
Células Epiteliales/citología , Pulmón/patología , Receptores de Formil Péptido/metabolismo , Antígenos/química , Línea Celular , Movimiento Celular , Humanos , Interleucina-8/metabolismo , Ligandos , Microscopía Fluorescente/métodos , Mitocondrias/patología , Necrosis/patología , Fracciones Subcelulares/metabolismo , Cicatrización de Heridas
19.
Am J Respir Cell Mol Biol ; 44(4): 483-90, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20525806

RESUMEN

An integral membrane protein, Claudin 5 (CLDN5), is a critical component of endothelial tight junctions that control pericellular permeability. Breaching of endothelial barriers is a key event in the development of pulmonary edema during acute lung injury (ALI). A major irritant in smoke, acrolein can induce ALI possibly by altering CLDN5 expression. This study sought to determine the cell signaling mechanism controlling endothelial CLDN5 expression during ALI. To assess susceptibility, 12 mouse strains were exposed to acrolein (10 ppm, 24 h), and survival monitored. Histology, lavage protein, and CLDN5 transcripts were measured in the lung of the most sensitive and resistant strains. CLDN5 transcripts and phosphorylation status of forkhead box O1 (FOXO1) and catenin (cadherin-associated protein) beta 1 (CTNNB1) proteins were determined in control and acrolein-treated human endothelial cells. Mean survival time (MST) varied more than 2-fold among strains with the susceptible (BALB/cByJ) and resistant (129X1/SvJ) strains (MST, 17.3 ± 1.9 h vs. 41.4 ± 5.1 h, respectively). Histological analysis revealed earlier perivascular enlargement in the BALB/cByJ than in 129X1/SvJ mouse lung. Lung CLDN5 transcript and protein increased more in the resistant strain than in the susceptible strain. In human endothelial cells, 30 nM acrolein increased CLDN5 transcripts and increased p-FOXO1 protein levels. The phosphatidylinositol 3-kinase inhibitor LY294002 diminished the acrolein-induced increased CLDN5 transcript. Acrolein (300 nM) decreased CLDN5 transcripts, which were accompanied by increased FOXO1 and CTNNB1. The phosphorylation status of these transcription factors was consistent with the observed CLDN5 alteration. Preservation of endothelial CLDN5 may be a novel clinical approach for ALI therapy.


Asunto(s)
Endotelio/fisiopatología , Lesión Pulmonar/fisiopatología , Proteínas de la Membrana/metabolismo , Acroleína , Animales , Línea Celular , Claudina-5 , Células Endoteliales/efectos de los fármacos , Células Endoteliales/enzimología , Endotelio/efectos de los fármacos , Endotelio/metabolismo , Endotelio/patología , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Células Híbridas/efectos de los fármacos , Células Híbridas/metabolismo , Pulmón/irrigación sanguínea , Pulmón/efectos de los fármacos , Pulmón/patología , Pulmón/fisiopatología , Lesión Pulmonar/genética , Lesión Pulmonar/patología , Proteínas de la Membrana/genética , Ratones , Microvasos/citología , Fosfatidilinositol 3-Quinasa/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Análisis de Supervivencia , beta Catenina/metabolismo
20.
Int J Exp Pathol ; 92(5): 366-76, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21838744

RESUMEN

Mitochondrial morphology and function are altered in intestinal epithelia during endotoxemia. However, it is unclear whether mitochondrial abnormalities occur in lung epithelial cells during acute sepsis or whether mitochondrial dysfunction corresponds with altered epithelial barrier function. Thus, we hypothesized that the intestinal epithelium is more susceptible to mitochondrial injury than the lung epithelium during acute sepsis and that mitochondrial dysfunction precedes impaired barrier function. Using a resuscitated feline model of Escherichia coli-induced sepsis, lung and ileal tissues were harvested after 6 h for histological and mitochondrial ultrastructural analyses in septic (n = 6) and time-matched controls (n = 6). Human lung epithelial cells (HLEC) and Caco-2 monolayers (n = 5) were exposed to 'cytomix' (TNFα: 40 ng/ml, IL-1ß: 20 ng/ml, IFNγ: 10 ng/ml) for 24-72 h, and measurements of transepithelial electrical resistance (TER), epithelial permeability and mitochondrial membrane potential (ΔΨ) were taken. Lung epithelial morphology, mitochondrial ultrastructure and pulmonary gas exchange were unaltered in septic animals compared to matching controls. While histologically intact, ileal epithelia demonstrated marked mitochondrial ultrastructural damage during sepsis. Caco-2 monolayers treated with cytomix showed a significant decrease in mitochondrial ΔΨ within 24 h, which was associated with a progressive reduction in TER and increased epithelial permeability over the subsequent 48 h. In contrast, mitochondrial ΔΨ and epithelial barrier functions were preserved in HLEC following cytomix. These findings indicate that intestinal epithelium is more susceptible to mitochondrial damage and dysfunction than the lung epithelium in the context of sepsis. Early alterations in mitochondrial function portend subsequent epithelial barrier dysfunction.


Asunto(s)
Permeabilidad de la Membrana Celular/fisiología , Infecciones por Escherichia coli/fisiopatología , Escherichia coli/aislamiento & purificación , Mucosa Intestinal/fisiopatología , Mucosa Respiratoria/fisiopatología , Sepsis/fisiopatología , Enfermedad Aguda , Animales , Apoptosis/fisiología , Células CACO-2 , Gatos , Células Cultivadas , Colon/microbiología , Colon/patología , Colon/fisiopatología , Modelos Animales de Enfermedad , Infecciones por Escherichia coli/microbiología , Infecciones por Escherichia coli/patología , Humanos , Íleon/microbiología , Íleon/patología , Íleon/fisiopatología , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Pulmón/microbiología , Pulmón/patología , Pulmón/fisiopatología , Masculino , Potencial de la Membrana Mitocondrial/fisiología , Mucosa Respiratoria/microbiología , Mucosa Respiratoria/patología , Sepsis/microbiología , Sepsis/patología , Factor de Necrosis Tumoral alfa/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA