Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Int J Mol Sci ; 21(8)2020 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-32344623

RESUMEN

Circadian oscillations are regulated at both central and peripheral levels to maintain physiological homeostasis. The central circadian clock consists of a central pacemaker in the suprachiasmatic nucleus that is entrained by light dark cycles and this, in turn, synchronizes the peripheral clock inherent in other organs. Circadian dysregulation has been attributed to dysregulation of peripheral clock and also associated with several diseases. Components of the molecular clock are disrupted in lung diseases like chronic obstructive pulmonary disease (COPD), asthma and IPF. Airway epithelial cells play an important role in temporally organizing magnitude of immune response, DNA damage response and acute airway inflammation. Non-coding RNAs play an important role in regulation of molecular clock and in turn are also regulated by clock components. Dysregulation of these non-coding RNAs have been shown to impact the expression of core clock genes as well as clock output genes in many organs. However, no studies have currently looked at the potential impact of these non-coding RNAs on lung molecular clock. This review focuses on the ways how these non-coding RNAs regulate and in turn are regulated by the lung molecular clock and its potential impact on lung diseases.


Asunto(s)
Relojes Circadianos/genética , Susceptibilidad a Enfermedades , Enfermedades Pulmonares/etiología , ARN no Traducido/genética , Animales , Biomarcadores , Ritmo Circadiano/genética , Regulación de la Expresión Génica , Humanos , MicroARNs
2.
Am J Respir Cell Mol Biol ; 61(6): 678-688, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31486667

RESUMEN

Chronic airway inflammation from recurring exposures to noxious environmental stimuli results in a progressive and irreversible airflow limitation and the lung parenchymal damage that characterizes chronic obstructive pulmonary disease (COPD). The large variability observed in the onset and progression of COPD is primarily driven by complex gene-environment interactions. The transcriptomic and epigenetic memory potential of lung epithelial and innate immune cells drive responses, such as mucus hyperreactivity and airway remodeling, that are tightly regulated by various molecular mechanisms, for which several candidate susceptibility genes have been described. However, the recently described noncoding RNA species, in particular the long noncoding RNAs, may also have an important role in modulating pulmonary responses to chronic inhalation of toxic substances and the development of COPD. This review outlines the features of long noncoding RNAs that have been implicated in regulating the airway inflammatory responses to cigarette smoke exposure and their possible association with COPD pathogenesis. As COPD continues to debilitate the increasingly aging population and contribute to higher morbidity and mortality rates worldwide, the search for better biomarkers and alternative therapeutic options is pivotal.


Asunto(s)
Enfermedad Pulmonar Obstructiva Crónica/genética , ARN Largo no Codificante/fisiología , Transcriptoma , Envejecimiento/genética , Envejecimiento/metabolismo , Contaminantes Atmosféricos/efectos adversos , Animales , Biomarcadores , Senescencia Celular , Células Epiteliales/metabolismo , Regulación de la Expresión Génica , Interacción Gen-Ambiente , Humanos , Inmunidad Innata/genética , Inflamación/genética , Inflamación/metabolismo , Pulmón/efectos de los fármacos , Pulmón/crecimiento & desarrollo , Pulmón/metabolismo , Ratones , Mitocondrias/patología , Modelos Animales , Enfermedad Pulmonar Obstructiva Crónica/etiología , Enfermedad Pulmonar Obstructiva Crónica/patología , ARN Largo no Codificante/genética , Humo/efectos adversos , Lesión por Inhalación de Humo/complicaciones , Fumar/efectos adversos , Fumar/genética , Nicotiana
3.
Front Mol Neurosci ; 17: 1365752, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38476461

RESUMEN

The leucine-rich repeat-containing protein 25 (LRRC25) is relatively a novel protein with no information on its role in neuronal or brain function. A recent study suggested LRRC25 is a potential risk factor for Alzheimer's disease (AD). As a first step to understanding LRRC25's role in the brain and AD, we found LRRC25 is expressed in both cell membranes and cytoplasm in a punctuate appearance in astrocytes, microglia, and neurons in cell lines as well as mouse brain. We also found that LRRC25 expression is both age- and brain region-dependent and that 1-day-old (1D) pups expressed the least amount of LRRC25 protein compared to adult ages. In the APΔE9 mice, immunoblot quantified LRRC25 protein levels were increased by 166% (**p < 0.01) in the cortex (CX) and by 215% (***p < 0.001) in the hippocampus (HP) relative to wild-type (WT) controls. Both the brainstem (BS) and cerebellum (CB) showed no significant alterations. In the 3xTg mice, only CX showed an increase of LRRC25 protein by 91% (*p < 0.05) when compared to WT controls although the increased trend was noted in the other brain regions. In the AD patient brains also LRRC25 protein levels were increased by 153% (***p < 0.001) when compared to normal control (NC) subjects. Finally, LRRC25 expression in the iPSC-derived neurons quantified by immunofluorescence was increased by 181% (**p < 0.01) in AD-derived neurons when compared to NC-derived neurons. Thus increased LRRC25 protein in multiple models of AD suggests that LRRC25 may play a pathogenic role in either Aß or tau pathology in AD. The mechanism for the increased levels of LRRC25 in AD is unknown at present, but a previous study showed that LRRC25 levels also increase during neonatal hypoxic-ischemia neuronal damage. Based on the evidence that autophagy is highly dysregulated in AD, the increased LRRC25 levels may be due to decreased autophagic degradation of LRRC25. Increased LRRC25 in turn may regulate the stability or activity of key enzymes involved in either Aß or hyperphosphorylated tau generation and thus may contribute to increased plaques and neurofibrillary tangles.

5.
Sci Rep ; 13(1): 10980, 2023 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-37414804

RESUMEN

Portable air purifiers help improve indoor air quality by neutralizing allergens, including animal dander proteins. However, there are limited in-vivo models to assess the efficacy of these devices. Here, we developed a novel animal model of experimental asthma using aerosolized cat dander extract (CDE) exposure and compared the efficacy of select air purification technologies. Mice were exposed to CDE aerosols for 6 weeks in separate custom-built whole-body exposure chambers equipped with either a photoelectrochemical oxidative (PECO) Molekule filtration device (PFD) or a HEPA-assisted air filtration device (HFD) along with positive (a device with no filtration capability) and negative controls. Compared to the positive control group, the CDE-induced airway resistance, and plasma IgE and IL-13 levels were significantly reduced in both air purifier groups. However, PFD mice showed a better attenuation of lung tissue mucous hyperplasia and eosinophilia than HFD and positive control mice, indicating a better efficacy in managing CDE-induced allergic responses. Cat dander protein destruction was evaluated by LCMS proteomic analysis, which revealed the degradation of 2731 unique peptides on PECO media in 1 h. Thus, allergen protein destruction on filtration media enhances air purifier efficacy that could provide relief from allergy responses compared to traditional HEPA-based filtration alone.


Asunto(s)
Contaminación del Aire Interior , Asma , Hipersensibilidad , Ratones , Animales , Modelos Animales de Enfermedad , Alérgenos Animales/metabolismo , Proteómica , Hipersensibilidad/metabolismo , Alérgenos
6.
Toxicol Rep ; 9: 1823-1830, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36518432

RESUMEN

Electronic cigarette (e-cig) aerosol exposures are strongly associated with pulmonary dysfunctions, and the airway epithelial cells (AECs) of respiratory passages play a pivotal role in understanding this association. However, not much is known about the effect of synthetic cooling agents such as WS-23 on AECs. WS-23 is a synthetic menthol-like cooling agent widely used to enhance the appeal of e-cigs and to suppress the harshness and bitterness of other e-cig constituents. Using primary human AECs, we compared the effects of aerosolized WS-23 with propylene glycol/vegetable glycerin (PG/VG) vehicle control and nicotine aerosol exposures. AECs treated with 3 % WS-23 aerosols showed a significant increase in viable cell numbers compared to PG/VG-vehicle aerosol exposed cells and cell growth was comparable following 2.5 % nicotine aerosol exposure. AEC inflammatory factors, IL-6 and ICAM-1 levels were significantly suppressed by WS-23 aerosols compared to PG/VG-controls. When differentiated AECs were challenged with WS-23 aerosols, there was a significant increase in secretory mucin MUC5AC expression with no discernible change in airway inflammatory SCGB1A1 expression. Compared to PG/VG-controls, WS-23 or nicotine aerosols presented with increased MUC5AC expression, but there was no synergistic effect of WS-23 + nicotine combination exposure. Thus, WS-23 and nicotine aerosols modulate the AEC responses and induce goblet cell hyperplasia, which could impact the airway physiology and susceptibility to respiratory diseases.

7.
Front Cell Neurosci ; 16: 954071, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35928571

RESUMEN

Alzheimer's disease (AD) is complex and highly heterogeneous. Less than 10% of AD cases are early-onset (EOAD) caused by autosomal dominantly inherited mutations in amyloid precursor protein (APP), presenilin 1 (PS1), or presenilin 2 (PS2), each of which can increase Aß generation and, thus, amyloid plaques. The remaining 90% of cases of AD are late-onset (LOAD) or sporadic. Intense research efforts have led to identification of many genes that increase the risk of AD. An IQ motif containing protein K (IQCK) was recently identified by several investigators as an Alzheimer's disease risk gene. However, how IQCK increases AD risk is completely unknown. Since IQCK is a novel gene, there is limited information on its physiological characterization. To understand its role in AD, it is first important to determine its subcellular localization, whether and where it is expressed in the brain, and what type of brain cells express the IQCK protein. Therefore, in this study, we show by immunocytochemical (ICC) staining that IQCK is expressed in both the nucleus and the cytoplasm of SH-SY5Y neuroblastoma cells as well as HeLa cells but not in either HMC3 microglial or CHO cells. By immunohistochemistry (IHC), we also show that IQCK is expressed in both mouse and human neurons, including neuronal processes in vivo in the mouse brain. IHC data also show that the IQCK protein is widely expressed throughout the mouse brain, although regional differences were noted. IQCK expression was highest in the brainstem (BS), followed by the cerebellum (CB) and the cortex (CX), and it was lowest in the hippocampus (HP). This finding was consistent with data from an immunoblot analysis of brain tissue homogenates. Interestingly, we found IQCK expression in neurons, astrocytes, and oligodendrocytes using cell-specific antibodies, but IQCK was not detected in microglial cells, consistent with negative in vitro results in HMC3 cells. Most importantly, we found that actin-normalized IQCK protein levels were increased by 2 folds in AD brains relative to normal control (NC) brains. Furthermore, the IQCK protein was found in amyloid plaques, suggesting that IQCK may play a pathogenic role in either Aß generation or amyloid plaque deposition in AD.

8.
iScience ; 25(8): 104685, 2022 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-35789750

RESUMEN

Noncoding RNAs are important regulators of mucoinflammatory response, but little is known about the contribution of airway long noncoding RNAs (lncRNAs) in COVID-19. RNA-seq analysis showed a more than 4-fold increased expression of IL-6, ICAM-1, CXCL-8, and SCGB1A1 inflammatory factors; MUC5AC and MUC5B mucins; and SPDEF, FOXA3, and FOXJ1 transcription factors in COVID-19 patient nasal samples compared with uninfected controls. A lncRNA on antisense strand to ICAM-1 or LASI was induced 2-fold in COVID-19 patients, and its expression was directly correlated with viral loads. A SARS-CoV-2-infected 3D-airway model largely recapitulated these clinical findings. RNA microscopy and molecular modeling indicated a possible interaction between viral RNA and LASI lncRNA. Notably, blocking LASI lncRNA reduced the SARS-CoV-2 replication and suppressed MUC5AC mucin levels and associated inflammation, and select LASI-dependent miRNAs (e.g., let-7b-5p and miR-200a-5p) were implicated. Thus, LASI lncRNA represents an essential facilitator of SARS-CoV-2 infection and associated airway mucoinflammatory response.

9.
Front Immunol ; 13: 803362, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35774797

RESUMEN

Research Impact: Cigarette smoke (CS) exposure is strongly associated with chronic obstructive pulmonary disease (COPD). In respiratory airways, CS exposure disrupts airway barrier functions, mucous/phlegm production, and basic immune responses of airway epithelial cells. Based on our recent identification of a specific immunomodulatory long noncoding RNA (lncRNA), we investigated its role in CS-induced responses in bronchial airways of cynomolgus macaque model of CS-induced COPD and in former smokers with and without COPD. The lncRNA was significantly upregulated in CS-induced macaque airways and in COPD airways that exhibited higher mucus expression and goblet cell hyperplasia. Experimental models of cells derived from COPD subjects recapitulated the augmented inflammation and mucus expression following the smoke challenge. Blocking of lncRNA expression in cell culture setting suppressed the smoke-induced and COPD-associated dysregulated mucoinflammatory response suggesting that this airway specific immunomodulatory lncRNA may represent a novel target to mitigate the smoke-mediated inflammation and mucus hyperexpression. Rationale: In conducting airways, CS disrupts airway epithelial functions, mucociliary clearances, and innate immune responses that are primarily orchestrated by human bronchial epithelial cells (HBECs). Mucus hypersecretion and dysregulated immune response are the hallmarks of chronic bronchitis (CB) that is often exacerbated by CS. Notably, we recently identified a long noncoding RNA (lncRNA) antisense to ICAM-1 (LASI) that mediates airway epithelial responses. Objective: To investigate the role of LASI lncRNA in CS-induced airway inflammation and mucin hyperexpression in an animal model of COPD, and in HBECs and lung tissues from former smokers with and without COPD. To interrogate LASI lncRNA role in CS-mediated airway mucoinflammatory responses by targeted gene editing. Methods: Small airway tissue sections from cynomolgus macaques exposed to long-term mainstream CS, and those from former smokers with and without COPD were analyzed. The structured-illumination imaging, RNA fluorescence in-situ hybridization (FISH), and qRT-PCR were used to characterize lncRNA expression and the expression of inflammatory factors and airway mucins in a cell culture model of CS extract (CSE) exposure using HBECs from COPD (CHBEs) in comparison with cells from normal control (NHBEs) subjects. The protein levels of mucin MUC5AC, and inflammatory factors ICAM-1, and IL-6 were determined using specific ELISAs. RNA silencing was used to block LASI lncRNA expression and lentivirus encoding LASI lncRNA was used to achieve LASI overexpression (LASI-OE). Results: Compared to controls, LASI lncRNA was upregulated in CS-exposed macaques and in COPD smoker airways, correlating with mucus hyperexpression and mucus cell hyperplasia in severe COPD airways. At baseline, the unstimulated CHBEs showed increased LASI lncRNA expression with higher expression of secretory mucin MUC5AC, and inflammatory factors, ICAM-1, and IL-6 compared to NHBEs. CSE exposure of CHBEs resulted in augmented inflammation and mucus expression compared to controls. While RNA silencing-mediated LASI knockdown suppressed the mucoinflammatory response, cells overexpressing LASI lncRNA showed elevated mRNA levels of inflammatory factors. Conclusions: Altogether, LASI lncRNA may represent a novel target to control the smoke-mediated dysregulation in airway responses and COPD exacerbations.


Asunto(s)
Fumar Cigarrillos , Enfermedad Pulmonar Obstructiva Crónica , ARN Largo no Codificante , Animales , Fumar Cigarrillos/efectos adversos , Células Caliciformes/metabolismo , Humanos , Hiperplasia , Inflamación , Molécula 1 de Adhesión Intercelular/genética , Interleucina-6 , Enfermedad Pulmonar Obstructiva Crónica/genética , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , ARN Largo no Codificante/genética , Nicotiana/efectos adversos
11.
Mol Neurobiol ; 58(6): 2465-2480, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33439437

RESUMEN

The recent outbreak of SARS-CoV-2 infections that causes coronavirus-induced disease of 2019 (COVID-19) is the defining and unprecedented global health crisis of our time in both the scale and magnitude. Although the respiratory tract is the primary target of SARS-CoV-2, accumulating evidence suggests that the virus may also invade both the central nervous system (CNS) and the peripheral nervous system (PNS) leading to numerous neurological issues including some serious complications such as seizures, encephalitis, and loss of consciousness. Here, we present a comprehensive review of the currently known role of SARS-CoV-2 and identify all the neurological problems reported among the COVID-19 case reports throughout the world. The virus might gain entry into the CNS either through the trans-synaptic route via the olfactory neurons or through the damaged endothelium in the brain microvasculature using the ACE2 receptor potentiated by neuropilin-1 (NRP-1). The most critical of all symptoms appear to be the spontaneous loss of breathing in some COVID-19 patients. This might be indicative of a dysfunction within the cardiopulmonary regulatory centers in the brainstem. These pioneering studies, thus, lay a strong foundation for more in-depth basic and clinical research required to confirm the role of SARS-CoV-2 infection in neurodegeneration of critical brain regulatory centers.


Asunto(s)
COVID-19/complicaciones , Enfermedades del Sistema Nervioso Central/etiología , Enfermedades del Sistema Nervioso Periférico/etiología , SARS-CoV-2 , Adulto , Factores de Edad , Enzima Convertidora de Angiotensina 2/metabolismo , Encéfalo/virología , COVID-19/epidemiología , COVID-19/fisiopatología , Enfermedades Cardiovasculares/epidemiología , Enfermedades del Sistema Nervioso Central/diagnóstico por imagen , Enfermedades del Sistema Nervioso Central/fisiopatología , Niño , Comorbilidad , Diabetes Mellitus/epidemiología , Células Endoteliales/patología , Femenino , Humanos , Enfermedades Renales/etiología , Hepatopatías/etiología , Masculino , Proteínas del Tejido Nervioso/metabolismo , Neuroimagen , Neuropilina-1/fisiología , Obesidad/epidemiología , Especificidad de Órganos , Enfermedades del Sistema Nervioso Periférico/fisiopatología , Receptores Virales/metabolismo , Glicoproteína de la Espiga del Coronavirus/metabolismo
12.
Stem Cell Rev Rep ; 17(3): 900-910, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33389682

RESUMEN

Cardiac mesenchymal cells (CMCs), a newly-discovered and promising type of progenitor cells, are effective in improving cardiac function in rodents after myocardial infarction. Stem/progenitor cells are usually cultured at atmospheric O2 tension (21%); however, the physiologic O2 tension in the heart is ~5%, raising the concern that 21% O2 may cause toxicity due to oxidative stress. Thus, we compared mouse CMCs cultured at 21% or 5% O2 beginning at passage 2. At passage 5, CMCs underwent severe hypoxic stress (1% O2 for 24 h). Compared with CMCs cultured at 21% O2, culture at 5% O2 consistently improved cell morphology throughout 5 passages, markedly decreased cell size, increased cell number, shortened cell doubling time, and dramatically reduced lactate dehydrogenase release from CMCs into culture media after hypoxic stress. Furthermore, culture at 5% O2 increased telomerase activity and telomere length, implying that 21% O2 tension impairs telomerase activity, resulting in telomere shortening and decreased cell proliferation. Thus far, almost all preclinical and clinical studies of cell therapy for the heart disease have used atmospheric (21%) O2 to culture cells. Our data challenge this paradigm. Our results demonstrate that, compared with 21% O2, 5% O2 tension greatly enhances the competence and functional properties of CMCs. The increased proliferation rate at 5% O2 means that target numbers of CMCs can be achieved with much less time and cost. Furthermore, since this increased proliferation may continue in vivo after CMC transplantation, and since cells grown at 5% O2 are markedly resistant to severe hypoxic stress, and thus may be better able to survive after transplantation into scarred regions of the heart where O2 is very low, culture at 5% O2 may enhance the reparative properties of CMCs (and possibly other cell types). In conclusion, our data support a change in the methods used to culture CMCs and possibly other progenitor cells.


Asunto(s)
Malformaciones Vasculares , Animales , Corazón , Ratones , Oxígeno , Células Madre , Telomerasa/genética
13.
medRxiv ; 2021 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-34031668

RESUMEN

Respiratory epithelial cells are the primary target for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). We investigated the 3D human airway tissue model to evaluate innate epithelial cell responses to SARS-CoV-2 infection. A SARS-CoV-2 clinical isolate productively infected the 3D-airway model with a time-dependent increase in viral load (VL) and concurrent upregulation of airway immunomodulatory factors ( IL-6, ICAM-1 , and SCGB1A1 ) and respiratory mucins ( MUC5AC, MUC5B, MUC2 , and MUC4) , and differential modulation of select long noncoding RNAs (lncRNAs i.e., LASI, TOSL, NEAT1 , and MALAT1 ). Next, we examined these immunomodulators in the COVID-19 patient nasopharyngeal swab samples collected from subjects with high- or low-VLs (∻100-fold difference). As compared to low-VL, high-VL patients had prominent mucoinflammatory signature with elevated expression of IL-6, ICAM-1, SCGB1A1, SPDEF, MUC5AC, MUC5B , and MUC4 . Interestingly, LASI, TOSL , and NEAT1 lncRNA expressions were also markedly elevated in high-VL patients with no change in MALAT1 expression. In addition, dual-staining of LASI and SARS-CoV-2 nucleocapsid N1 RNA showed predominantly nuclear/perinuclear localization at 24 hpi in 3D-airway model as well as in high-VL COVID-19 patient nasopharyngeal cells, which exhibited high MUC5AC immunopositivity. Collectively, these findings suggest SARS-CoV-2 induced lncRNAs may play a role in acute mucoinflammatory response observed in symptomatic COVID-19 patients.

14.
Neurobiol Aging ; 106: 26-36, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34229273

RESUMEN

Identification of molecules and molecular pathways that can ameliorate aging-associated decline in cognitive function is crucial. Here we report that the protein levels of transcription factor EB (TFEB) were markedly reduced in both the cytosolic and nuclear fractions of the frontal cortex and hippocampus at 18-months of age relative to 6 months in the normal male wild-type mice. In the transgenic mice with ectopic expression of flag-TFEB in neurons, we observed that the levels of actin-normalized PGC1α and mtTFA were significantly increased in both the cortex and the hippocampus. Additionally, we confirmed increased mitochondria numbers in the flag-TFEB mice by transmission electron microscopy. Most importantly, TFEB expression in the 18-month-old transgenic mice mitigated markers of senescence including P16INK4a, γ-H2AX, and lamin B1, and improved memory skills implying that TFEB may exert an anti-aging effect by modulating neuronal senescence. Taken together these data strongly support that TFEB can be a useful therapeutic target for brain senescent cells to help overcome the age-related issues in cognition and possibly, achieve healthy aging.


Asunto(s)
Envejecimiento/genética , Envejecimiento/metabolismo , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Biomarcadores/metabolismo , Encéfalo/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Expresión Génica/genética , Trastornos de la Memoria/genética , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/fisiología , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Histonas/metabolismo , Trastornos de la Memoria/terapia , Ratones Transgénicos , Terapia Molecular Dirigida
15.
Mucosal Immunol ; 14(3): 630-639, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33122732

RESUMEN

Epithelial cells of the conducting airways are a pivotal first line of defense against airborne pathogens and allergens that orchestrate inflammatory responses and mucociliary clearance. Nonetheless, the molecular mechanisms responsible for epithelial hyperreactivity associated with allergic asthma are not completely understood. Transcriptomic analysis of human airway epithelial cells (HAECs), differentiated in-vitro at air-liquid interface (ALI), showed 725 differentially expressed immediate-early transcripts, including putative long noncoding RNAs (lncRNAs). A novel lncRNA on the antisense strand of ICAM-1 or LASI was identified, which was induced in LPS-primed HAECs along with mucin MUC5AC and its transcriptional regulator SPDEF. LPS-primed expression of LASI, MUC5AC, and SPDEF transcripts were higher in ex-vivo cultured asthmatic HAECs that were further augmented by LPS treatment. Airway sections from asthmatics with increased mucus load showed higher LASI expression in MUC5AC+ goblet cells following multi-fluorescent in-situ hybridization and immunostaining. LPS- or IL-13-induced LASI transcripts were mostly enriched in the nuclear/perinuclear region and were associated with increased ICAM-1, IL-6, and CXCL-8 expression. Blocking LASI expression reduced the LPS or IL-13-induced epithelial inflammatory factors and MUC5AC expression, suggesting that the novel lncRNA LASI could play a key role in LPS-primed trained airway epithelial responses that are dysregulated in allergic asthma.


Asunto(s)
Asma/genética , Hipersensibilidad/genética , Molécula 1 de Adhesión Intercelular/genética , ARN sin Sentido/genética , Mucosa Respiratoria/fisiología , Diferenciación Celular , Línea Celular , Células Cultivadas , Citocinas/metabolismo , Perfilación de la Expresión Génica , Humanos , Interleucina-8/metabolismo , Lipopolisacáridos/inmunología , Mucina 5AC/genética , Mucina 5AC/metabolismo , Proteínas Proto-Oncogénicas c-ets/genética , Proteínas Proto-Oncogénicas c-ets/metabolismo , ARN Largo no Codificante , Hipersensibilidad Respiratoria , Regulación hacia Arriba
16.
Crit Rev Biomed Eng ; 48(3): 137-152, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33389892

RESUMEN

Mitochondria are among the most dynamic organelles regulating a wide array of cellular processes. They are the cellular hub for oxidative phosphorylation, energy production, and cellular metabolism, and they are important determinants of cell fate, as they control cell death/survival pathways. The mitochondrial network plays a critical role in cellular inflammatory responses, and mitochondria are central in many pathologic conditions such as chronic inflammatory and aging-associated degenerative diseases. Recent advancements in our understanding of the pathogenic pathways and the role of mitochondria therein have identified highly specific therapeutic targets in order to develop personalized nanomedicine approaches for treatment. A wide array of nanoparticle-based formulations has been employed for potential usage in both diagnosing and treating chronic and fatal conditions, with gold nanoparticles and liposomal encapsulation being of particular interest. In this review, we highlight and summarize the advantages and challenges of developing these nanoformulations for targeted and spatiotemporally controlled drug delivery. We discuss the potential of nanotherapy in neoplasms to target the mitochondrial regulated cell death pathways and recent seminal developments in liposomal nanotherapy against chronic inflammatory lung diseases. The need for further development of nanoparticle-based treatment options for neuroinflammatory and neurodegenerative conditions, such as Alzheimer's disease (AD), is also discussed.


Asunto(s)
Nanopartículas del Metal , Enfermedades Neurodegenerativas , Encéfalo , Muerte Celular , Oro , Humanos , Pulmón , Mitocondrias , Enfermedades Neurodegenerativas/tratamiento farmacológico
17.
Front Immunol ; 11: 1628, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32849552

RESUMEN

Rationale: Gestational cigarette smoke (CS) impairs lung angiogenesis and alveolarization, promoting transgenerational development of asthma and bronchopulmonary dysplasia (BPD). Hydrogen sulfide (H2S), a proangiogenic, pro-alveolarization, and anti-asthmatic gasotransmitter is synthesized by cystathionine-γ-lyase (CSE), cystathionine-ß-synthase (CBS), and 3-mercaptopyruvate sulfur transferase (3MST). Objective: Determine if gestational CS exposure affected the expression of H2S synthesizing enzymes in the mouse lung and human placenta. Methods: Mice were exposed throughout gestational period to secondhand CS (SS) at approximating the dose of CS received by a pregnant woman sitting in a smoking bar for 3 h/days during pregnancy. Lungs from 7-days old control and SS-exposed pups and human placenta from mothers who were either non-smokers or smokers during pregnancy were analyzed for expression of the enzymes. Measurements: Mouse lungs and human placentas were examined for the expression of CSE, CBS, and 3MST by immunohistochemical staining, qRT-PCR and/or Western blot (WB) analyses. Results: Compared to controls, mouse lung exposed gestationally to SS had significantly lower levels of CSE, CBS, and 3MST. Moreover, the SS-induced suppression of CSE and CBS in F1 lungs was transmitted to the F2 generation without significant change in the magnitude of the suppression. These changes were associated with impaired epithelial-mesenchymal transition (EMT)-a process required for normal lung angiogenesis and alveolarization. Additionally, the placentas from mothers who smoked during pregnancy, expressed significantly lower levels of CSE, CBS, and 3MST, and the effects were partially moderated by quitting smoking during the first trimester. Conclusions: Lung H2S synthesizing enzymes are downregulated by gestational CS and the effects are transmitted to F2 progeny. Smoking during pregnancy decreases H2S synthesizing enzymes is human placentas, which may correlate with the increased risk of asthma/BPD in children.


Asunto(s)
Gasotransmisores/biosíntesis , Sulfuro de Hidrógeno/metabolismo , Exposición Materna/efectos adversos , Efectos Tardíos de la Exposición Prenatal , Fumar Tabaco/efectos adversos , Animales , Modelos Animales de Enfermedad , Transición Epitelial-Mesenquimal , Femenino , Técnica del Anticuerpo Fluorescente , Regulación Enzimológica de la Expresión Génica , Humanos , Sulfuro de Hidrógeno/efectos adversos , Inmunohistoquímica , Pulmón/metabolismo , Pulmón/patología , Intercambio Materno-Fetal , Ratones , Modelos Biológicos , Placenta/metabolismo , Embarazo
18.
Redox Biol ; 33: 101443, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32037306

RESUMEN

The abnormal inflammatory responses due to the lung tissue damage and ineffective repair/resolution in response to the inhaled toxicants result in the pathological changes associated with chronic respiratory diseases. Investigation of such pathophysiological mechanisms provides the opportunity to develop the molecular phenotype-specific diagnostic assays and could help in designing the personalized medicine-based therapeutic approaches against these prevalent diseases. As the central hubs of cell metabolism and energetics, mitochondria integrate cellular responses and interorganellar signaling pathways to maintain cellular and extracellular redox status and the cellular senescence that dictate the lung tissue responses. Specifically, as observed in chronic obstructive pulmonary disease (COPD) and pulmonary fibrosis, the mitochondria-endoplasmic reticulum (ER) crosstalk is disrupted by the inhaled toxicants such as the combustible and emerging electronic nicotine-delivery system (ENDS) tobacco products. Thus, the recent research efforts have focused on understanding how the mitochondria-ER dysfunctions and oxidative stress responses can be targeted to improve inflammatory and cellular dysfunctions associated with these pathologic illnesses that are exacerbated by viral infections. The present review assesses the importance of these redox signaling and cellular senescence pathways that describe the role of mitochondria and ER on the development and function of lung epithelial responses, highlighting the cause and effect associations that reflect the disease pathogenesis and possible intervention strategies.


Asunto(s)
Senescencia Celular , Enfermedad Pulmonar Obstructiva Crónica , Humanos , Pulmón , Mitocondrias/metabolismo , Estrés Oxidativo , Enfermedad Pulmonar Obstructiva Crónica/metabolismo
19.
Front Cell Infect Microbiol ; 10: 612360, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33614527

RESUMEN

Background: The role of lung epithelial cells in HIV-1-related lung comorbidities remains unclear, and the major hurdle in curing HIV is the persistence of latent HIV reservoirs in people living with HIV (PLWH). The advent of combined antiretroviral therapy has considerably increased the life span; however, the incidence of chronic lung diseases is significantly higher among PLWH. Lung epithelial cells orchestrate the respiratory immune responses and whether these cells are productively infected by HIV-1 is debatable. Methods: Normal human bronchial epithelial cells (NHBEs) grown on air-liquid interface were infected with X4-tropic HIV-1LAV and examined for latency using latency-reversing agents (LRAs). The role of CD4 and CXCR4 HIV coreceptors in NHBEs were tested, and DNA sequencing analysis was used to analyze the genomic integration of HIV proviral genes, Alu-HIVgag-pol, HIV-nef, and HIV-LTR. Lung epithelial sections from HIV-infected humans and SHIV-infected macaques were analyzed by FISH for HIV-gag-pol RNA and epithelial cell-specific immunostaining. Results and Discussion: NHBEs express CD4 and CXCR4 at higher levels than A549 cells. NHBEs are infected with HIV-1 basolaterally, but not apically, by X4-tropic HIV-1LAV in a CXCR4/CD4-dependent manner leading to HIV-p24 antigen production; however, NHBEs are induced to express CCR5 by IL-13 treatment. In the presence of cART, HIV-1 induces latency and integration of HIV provirus in the cellular DNA, which is rescued by the LRAs (endotoxin/vorinostat). Furthermore, lung epithelial cells from HIV-infected humans and SHIV-infected macaques contain HIV-specific RNA transcripts. Thus, lung epithelial cells are targeted by HIV-1 and could serve as potential HIV reservoirs that may contribute to the respiratory comorbidities in PLWH.


Asunto(s)
Infecciones por VIH , VIH-1 , Antirretrovirales , Linfocitos T CD4-Positivos , Células Epiteliales , Infecciones por VIH/tratamiento farmacológico , VIH-1/genética , Humanos , Latencia del Virus
20.
Nanoscale ; 11(46): 22387-22397, 2019 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-31730144

RESUMEN

The blood-brain barrier (BBB) is a main obstacle for drug delivery targeting the central nervous system (CNS) and treating Alzheimer's disease (AD). In order to enhance the efficiency of drug delivery without harming the BBB integrity, nanoparticle-mediated drug delivery has become a popular therapeutic strategy. Carbon dots (CDs) are one of the most promising and novel nanocarriers. In this study, amphiphilic yellow-emissive CDs (Y-CDs) were synthesized with an ultrasonication-mediated methodology using citric acid and o-phenylenediamine with a size of 3 nm that emit an excitation-independent yellow photoluminescence (PL). The content of primary amine and carboxyl groups on CDs was measured as 6.12 × 10-5 and 8.13 × 10-3 mmol mg-1, respectively, indicating the potential for small-molecule drug loading through bioconjugation. Confocal image analyses revealed that Y-CDs crossed the BBB of 5-day old wild-type zebrafish, most probably by passive diffusion due to the amphiphilicity of Y-CDs. And the amphiphilicity and BBB penetration ability didn't change when Y-CDs were coated with different hydrophilic molecules. Furthermore, Y-CDs were observed to enter cells to inhibit the overexpression of human amyloid precursor protein (APP) and ß-amyloid (Aß) which is a major factor responsible for AD pathology. Therefore, data suggest that Y-CDs have a great potential as nontoxic nanocarriers for drug delivery towards the CNS as well as a promising inhibiting agent of Aß-related pathology of the AD.


Asunto(s)
Carbono/química , Portadores de Fármacos/química , Puntos Cuánticos/química , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/antagonistas & inhibidores , Péptidos beta-Amiloides/metabolismo , Animales , Barrera Hematoencefálica/metabolismo , Línea Celular , Supervivencia Celular/efectos de los fármacos , Humanos , Microscopía Confocal , Puntos Cuánticos/metabolismo , Puntos Cuánticos/uso terapéutico , Puntos Cuánticos/toxicidad , Pez Cebra
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA