Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
BMC Genomics ; 22(1): 759, 2021 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-34689749

RESUMEN

BACKGROUND: Archival formalin-fixed, paraffin-embedded (FFPE) tissue samples with clinical and histological data are a singularly valuable resource for developing new molecular biomarkers. However, transcriptome analysis remains challenging with standard mRNA-seq methods as FFPE derived-RNA samples are often highly modified and fragmented. The recently developed 3' mRNA-seq method sequences the 3' region of mRNA using unique molecular identifiers (UMI), thus generating gene expression data with minimal PCR bias. In this study, we evaluated the performance of 3' mRNA-Seq using Lexogen QuantSeq 3' mRNA-Seq Library Prep Kit FWD with UMI, comparing with TruSeq Stranded mRNA-Seq and RNA Exome Capture kit. The fresh-frozen (FF) and FFPE tissues yielded nucleotide sizes range from 13 to > 70% of DV200 values; input amounts ranged from 1 ng to 100 ng for validation. RESULTS: The total mapped reads of QuantSeq 3' mRNA-Seq to the reference genome ranged from 99 to 74% across all samples. After PCR bias correction, 3 to 56% of total sequenced reads were retained. QuantSeq 3' mRNA-Seq data showed highly reproducible data across replicates in Universal Human Reference RNA (UHR, R > 0.94) at input amounts from 1 ng to 100 ng, and FF and FFPE paired samples (R = 0.92) at 10 ng. Severely degraded FFPE RNA with ≤30% of DV200 value showed good concordance (R > 0.87) with 100 ng input. A moderate correlation was observed when directly comparing QuantSeq 3' mRNA-Seq data with TruSeq Stranded mRNA-Seq (R = 0.78) and RNA Exome Capture data (R > 0.67). CONCLUSION: In this study, QuantSeq 3' mRNA-Seq with PCR bias correction using UMI is shown to be a suitable method for gene quantification in both FF and FFPE RNAs. 3' mRNA-Seq with UMI may be applied to severely degraded RNA from FFPE tissues generating high-quality sequencing data.


Asunto(s)
Perfilación de la Expresión Génica , ARN , Formaldehído , Humanos , Adhesión en Parafina , ARN Mensajero/genética , Análisis de Secuencia de ARN , Fijación del Tejido
2.
BMC Genomics ; 21(1): 890, 2020 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-33308163

RESUMEN

BACKGROUND: There are challenges in generating mRNA-Seq data from whole-blood derived RNA as globin gene and rRNA are frequent contaminants. Given the abundance of erythrocytes in whole blood, globin genes comprise some 80% or more of the total RNA. Therefore, depletion of globin gene RNA and rRNA are critical steps required to have adequate coverage of reads mapping to the reference transcripts and thus reduce the total cost of sequencing. In this study, we directly compared the performance of probe hybridization (GLOBINClear Kit and Globin-Zero Gold rRNA Removal Kit) and RNAse-H enzymatic depletion (NEBNext® Globin & rRNA Depletion Kit and Ribo-Zero Plus rRNA Depletion Kit) methods from 1 µg of whole blood-derived RNA on mRNA-Seq profiling. All RNA samples were treated with DNaseI for additional cleanup before the depletion step and were processed for poly-A selection for library generation. RESULTS: Probe hybridization revealed a better overall performance than the RNAse-H enzymatic depletion method, detecting a higher number of genes and transcripts without 3' region bias. After depletion, samples treated with probe hybridization showed globin genes at 0.5% (±0.6%) of the total mapped reads; the RNAse-H enzymatic depletion had 3.2% (±3.8%). Probe hybridization showed more junction reads and transcripts compared with RNAse-H enzymatic depletion and also had a higher correlation (R > 0.9) than RNAse-H enzymatic depletion (R > 0.85). CONCLUSION: In this study, our results showed that 1 µg of high-quality RNA from whole blood could be routinely used for transcriptional profiling analysis studies with globin gene and rRNA depletion pre-processing. We also demonstrated that the probe hybridization depletion method is better suited to mRNA sequencing analysis with minimal effect on RNA quality during depletion procedures.


Asunto(s)
Poli A , ARN , Perfilación de la Expresión Génica , Globinas/genética , ARN Mensajero/genética , Análisis de Secuencia de ARN
3.
Biochim Biophys Acta ; 1843(7): 1259-71, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24637330

RESUMEN

HMG-CoA reductase, the proximal rate-limiting enzyme in the mevalonate pathway, is inhibited by statins. Beyond their cholesterol lowering impact, statins have pleiotropic effects and their use is linked to improved lung health. We have shown that mevalonate cascade inhibition induces apoptosis and autophagy in cultured human airway mesenchymal cells. Here, we show that simvastatin also induces endoplasmic reticulum (ER) stress and the unfolded protein response (UPR) in these cells. We tested whether coordination of ER stress, autophagy and apoptosis determines survival or demise of human lung mesenchymal cells exposed to statin. We observed that simvastatin exposure activates UPR (activated transcription factor 4, activated transcription factor 6 and IRE1α) and caspase-4 in primary human airway fibroblasts and smooth muscle cells. Exogenous mevalonate inhibited apoptosis, autophagy and UPR, but exogenous cholesterol was without impact, indicating that sterol intermediates are involved with mechanisms mediating statin effects. Caspase-4 inhibition decreased simvastatin-induced apoptosis, whereas inhibition of autophagy by ATG7 or ATG3 knockdown significantly increased cell death. In BAX(-/-)/BAK(-/-) murine embryonic fibroblasts, simvastatin-triggered apoptotic and UPR events were abrogated, but autophagy flux was increased leading to cell death via necrosis. Our data indicate that mevalonate cascade inhibition, likely associated with depletion of sterol intermediates, can lead to cell death via coordinated apoptosis, autophagy, and ER stress. The interplay between these pathways appears to be principally regulated by autophagy and Bcl-2-family pro-apoptotic proteins. These findings uncover multiple mechanisms of action of statins that could contribute to refining the use of such agent in treatment of lung disease.


Asunto(s)
Autofagia/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Ácido Mevalónico/farmacología , Miocitos del Músculo Liso/efectos de los fármacos , Respuesta de Proteína Desplegada/efectos de los fármacos , Proteína Destructora del Antagonista Homólogo bcl-2/genética , Proteína X Asociada a bcl-2/genética , Animales , Apoptosis/efectos de los fármacos , Proteína 7 Relacionada con la Autofagia , Proteínas Relacionadas con la Autofagia , Bronquios/citología , Bronquios/efectos de los fármacos , Bronquios/metabolismo , Caspasas Iniciadoras/genética , Caspasas Iniciadoras/metabolismo , Supervivencia Celular , Fibroblastos/citología , Fibroblastos/metabolismo , Regulación de la Expresión Génica , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Ratones , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Cultivo Primario de Células , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Simvastatina/farmacología , Enzimas Activadoras de Ubiquitina/antagonistas & inhibidores , Enzimas Activadoras de Ubiquitina/genética , Enzimas Activadoras de Ubiquitina/metabolismo , Enzimas Ubiquitina-Conjugadoras/antagonistas & inhibidores , Enzimas Ubiquitina-Conjugadoras/genética , Enzimas Ubiquitina-Conjugadoras/metabolismo , Respuesta de Proteína Desplegada/genética , Proteína Destructora del Antagonista Homólogo bcl-2/deficiencia , Proteína X Asociada a bcl-2/deficiencia
4.
mSphere ; 8(6): e0023223, 2023 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-37882516

RESUMEN

IMPORTANCE: We analyzed over 22,000 severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) genomes of patient samples tested at Mayo Clinic Laboratories during a 2-year period in the COVID-19 pandemic, which included Alpha, Delta, and Omicron variants of concern to examine the roles and relationships of Minnesota virus transmission. We found that Hennepin County, the most populous county, drove the transmission of SARS-CoV-2 viruses in the state after including the formation of earlier clades including 20A, 20C, and 20G, as well as variants of concern Alpha and Delta. We also found that Hennepin County was the source for most of the county-to-county introductions after an initial predicted introduction with the virus in early 2020 from an international source, while other counties acted as transmission "sinks." In addition, major policies, such as the end of the lockdown period in 2020 or the end of all restrictions in 2021, did not appear to have an impact on virus diversity across individual counties.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , SARS-CoV-2/genética , Minnesota/epidemiología , Pandemias , COVID-19/epidemiología , Control de Enfermedades Transmisibles , Genómica
5.
medRxiv ; 2023 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-35923324

RESUMEN

SARS-CoV-2 has had an unprecedented impact on human health and highlights the need for genomic epidemiology studies to increase our understanding of virus evolution and spread, and to inform policy decisions. We sequenced viral genomes from over 22,000 patient samples tested at Mayo Clinic Laboratories between 2020-2022 and use Bayesian phylodynamics to describe county and regional spread in Minnesota. The earliest introduction into Minnesota was to Hennepin County from a domestic source around January 22, 2020; six weeks before the first confirmed case in the state. This led to the virus spreading to Northern Minnesota, and eventually, the rest of the state. International introductions were most abundant in Hennepin (home to the Minneapolis/St. Paul International (MSP) airport) totaling 45 (out of 107) over the two-year period. Southern Minnesota counties were most common for domestic introductions with 19 (out of 64), potentially driven by bordering states such as Iowa and Wisconsin as well as Illinois which is nearby. Hennepin also was, by far, the most dominant source of in-state transmissions to other Minnesota locations (n=772) over the two-year period. We also analyzed the diversity of the location source of SARS-CoV-2 viruses in each county and noted the timing of state-wide policies as well as trends in clinical cases. Neither the number of clinical cases or major policy decisions, such as the end of the lockdown period in 2020 or the end of all restrictions in 2021, appeared to have impact on virus diversity across each individual county.

6.
Am J Physiol Lung Cell Mol Physiol ; 302(4): L420-8, 2012 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-22160308

RESUMEN

Geranylgeranyl transferase 1 (GGT1) is involved in the posttranslational prenylation of signaling proteins, such as small GTPases. We have shown that blocking the formation of isoprenoids with statins regulates survival of human lung mesenchymal cells; thus, we tested the hypothesis that GGT1 may specifically modulate programmed cell death pathways in these cells. To this end, human airway smooth muscle (HASM) cells were treated with the selective GGT1 inhibitor GGTi-298. Apoptosis was seen using assays for cellular DNA content and caspase activation. Induction of autophagy was observed using transmission electron microscopy, immunoblotting for LC3 lipidation and Atg5-12 complex content, and confocal microscopy to detect formation of lysosome-localized LC3 punctae. Notably, GGT1 inhibition induced expression of p53-dependent proteins, p53 upregulated modulator of apoptosis (Noxa), and damage-regulated autophagy modulator (DRAM), this was inhibited by the p53 transcriptional activation inhibitor cyclic-pifithrin-α. Inhibition of autophagy with bafilomycin-A1 or short-hairpin RNA silencing of Atg7 substantially augmented GGTi-298-induced apoptosis. Overall, we demonstrate for the first time that pharmacological inhibition of GGT1 induces simultaneous p53-dependent apoptosis and autophagy in HASM. Moreover, autophagy regulates apoptosis induction. Thus, our findings identify GGT1 as a key regulator of HASM cell viability.


Asunto(s)
Transferasas Alquil y Aril/metabolismo , Apoptosis , Autofagia , Bronquios/citología , Farnesiltransferasa/metabolismo , Miocitos del Músculo Liso/enzimología , Transferasas Alquil y Aril/antagonistas & inhibidores , Transferasas Alquil y Aril/genética , Benzamidas/farmacología , Benzotiazoles/farmacología , Supervivencia Celular , Células Cultivadas , Farnesiltransferasa/antagonistas & inhibidores , Farnesiltransferasa/genética , Humanos , Miocitos del Músculo Liso/fisiología , Cultivo Primario de Células , Transducción de Señal , Tolueno/análogos & derivados , Tolueno/farmacología , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
7.
J Cell Sci ; 123(Pt 18): 3061-70, 2010 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-20736308

RESUMEN

The dystrophin-glycoprotein complex (DGC) links the extracellular matrix and actin cytoskeleton. Caveolae form membrane arrays on smooth muscle cells; we investigated the mechanism for this organization. Caveolin-1 and beta-dystroglycan, the core transmembrane DGC subunit, colocalize in airway smooth muscle. Immunoprecipitation revealed the association of caveolin-1 with beta-dystroglycan. Disruption of actin filaments disordered caveolae arrays, reduced association of beta-dystroglycan and caveolin-1 to lipid rafts, and suppressed the sensitivity and responsiveness of methacholine-induced intracellular Ca2+ release. We generated novel human airway smooth muscle cell lines expressing shRNA to stably silence beta-dystroglycan expression. In these myocytes, caveolae arrays were disorganized, caveolae structural proteins caveolin-1 and PTRF/cavin were displaced, the signaling proteins PLCbeta1 and G(alphaq), which are required for receptor-mediated Ca2+ release, were absent from caveolae, and the sensitivity and responsiveness of methacholine-induced intracellular Ca2+ release, was diminished. These data reveal an interaction between caveolin-1 and beta-dystroglycan and demonstrate that this association, in concert with anchorage to the actin cytoskeleton, underpins the spatial organization and functional role of caveolae in receptor-mediated Ca2+ release, which is an essential initiator step in smooth muscle contraction.


Asunto(s)
Calcio/metabolismo , Caveolas/metabolismo , Caveolina 1/metabolismo , Distroglicanos/metabolismo , Músculo Liso/metabolismo , Animales , Caveolina 1/genética , Células Cultivadas , Citoesqueleto/genética , Citoesqueleto/metabolismo , Perros , Distroglicanos/genética , Humanos , Células Musculares/metabolismo , Unión Proteica
8.
Am J Respir Cell Mol Biol ; 44(3): 394-403, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20463291

RESUMEN

Smooth muscle cells promote fibroproliferative airway remodeling in asthma, and transforming growth factor ß1 (TGFß1) is a key inductive signal. Statins are widely used to treat hyperlipidemia. Growing evidence indicates they also exert a positive impact on lung health, but the underlying mechanisms are unclear. We assessed the effects of 3-hydroxy-3-methlyglutaryl-coenzyme A (HMG-CoA) reductase inhibition with simvastatin on the fibrotic function of primary cultured human airway smooth muscle cells. Simvastatin blocked de novo cholesterol synthesis, but total myocyte cholesterol content was unaffected. Simvastatin also abrogated TGFß1-induced collagen I and fibronectin expression, and prevented collagen I secretion. The depletion of mevalonate cascade intermediates downstream from HMG-CoA underpinned the effects of simvastatin, because co-incubation with mevalonate, geranylgeranylpyrophosphate, or farnesylpyrophosphate prevented the inhibition of matrix protein expression. We also showed that human airway myocytes express both geranylgeranyl transferase 1 (GGT1) and farnesyltransferase (FT), and the inhibition of GGT1 (GGTI inhibitor-286, 10 µM), but not FT (FTI inhibitor-277, 10 µM), mirrored the suppressive effects of simvastatin on collagen I and fibronectin expression and collagen I secretion. Moreover, simvastatin and GGTI-286 both prevented TGFß1-induced membrane association of RhoA, a downstream target of GGT1. Our findings suggest that simvastatin and GGTI-286 inhibit synthesis and secretion of extracellular matrix proteins by human airway smooth muscle cells by suppressing GGT1-mediated posttranslational modification of signaling molecules such as RhoA. These findings reveal mechanisms related to evidence for the positive impact of statins on pulmonary health.


Asunto(s)
Matriz Extracelular/metabolismo , Regulación de la Expresión Génica , Ácido Mevalónico/metabolismo , Tráquea/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Transferasas Alquil y Aril/metabolismo , Colágeno/metabolismo , Colágeno Tipo I/metabolismo , Farnesiltransferasa/metabolismo , Humanos , Leucina/análogos & derivados , Leucina/farmacología , Modelos Biológicos , Procesamiento Proteico-Postraduccional , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Simvastatina/farmacología
9.
J Cell Mol Med ; 15(11): 2430-42, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21199324

RESUMEN

Airway smooth muscle cells exhibit phenotype plasticity that underpins their ability to contribute both to acute bronchospasm and to the features of airway remodelling in chronic asthma. A feature of mature, contractile smooth muscle cells is the presence of abundant caveolae, plasma membrane invaginations that develop from the association of lipid rafts with caveolin-1, but the functional role of caveolae and caveolin-1 in smooth muscle phenotype plasticity is unknown. Here, we report a key role for caveolin-1 in promoting phenotype maturation of differentiated airway smooth muscle induced by transforming growth factor (TGF)-ß(1). As assessed by Western analysis and laser scanning cytometry, caveolin-1 protein expression was selectively enriched in contractile phenotype airway myocytes. Treatment with TGF-ß(1) induced profound increases in the contractile phenotype markers sm-α-actin and calponin in cells that also accumulated abundant caveolin-1; however, siRNA or shRNAi inhibition of caveolin-1 expression largely prevented the induction of these contractile phenotype marker proteins by TGF-ß(1). The failure by TGF-ß(1) to adequately induce the expression of these smooth muscle specific proteins was accompanied by a strongly impaired induction of eukaryotic initiation factor-4E binding protein(4E-BP)1 phosphorylation with caveolin-1 knockdown, indicating that caveolin-1 expression promotes TGF-ß(1) signalling associated with myocyte maturation and hypertrophy. Furthermore, we observed increased expression of caveolin-1 within the airway smooth muscle bundle of guinea pigs repeatedly challenged with allergen, which was associated with increased contractile protein expression, thus providing in vivo evidence linking caveolin-1 expression with accumulation of contractile phenotype myocytes. Collectively, we identify a new function for caveolin-1 in controlling smooth muscle phenotype; this mechanism could contribute to allergic asthma.


Asunto(s)
Caveolina 1/metabolismo , Contracción Muscular , Miocitos del Músculo Liso/metabolismo , Sistema Respiratorio/metabolismo , Actinas/biosíntesis , Remodelación de las Vías Aéreas (Respiratorias) , Animales , Asma/fisiopatología , Proteínas de Unión al Calcio , Caveolas/metabolismo , Caveolas/fisiología , Caveolina 1/genética , Células Cultivadas , Perros , Factor 4E Eucariótico de Iniciación/metabolismo , Cobayas , Humanos , Proteínas de Microfilamentos , Células Musculares , Miocitos del Músculo Liso/fisiología , Fenotipo , Interferencia de ARN , ARN Interferente Pequeño , Transducción de Señal , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Crecimiento Transformador beta1/farmacología , Calponinas
10.
Biochim Biophys Acta ; 1803(4): 452-67, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20045437

RESUMEN

Statins inhibit 3-hydroxy-3-methyl-glutarylcoenzyme CoA (HMG-CoA) reductase, the proximal enzyme for cholesterol biosynthesis. They exhibit pleiotropic effects and are linked to health benefits for diseases including cancer and lung disease. Understanding their mechanism of action could point to new therapies, thus we investigated the response of primary cultured human airway mesenchymal cells, which play an effector role in asthma and chronic obstructive lung disease (COPD), to simvastatin exposure. Simvastatin induced apoptosis involving caspase-9, -3 and -7, but not caspase-8 in airway smooth muscle cells and fibroblasts. HMG-CoA inhibition did not alter cellular cholesterol content but did abrogate de novo cholesterol synthesis. Pro-apoptotic effects were prevented by exogenous mevalonate, geranylgeranyl pyrophosphate and farnesyl pyrophosphate, downstream products of HMG-CoA. Simvastatin increased expression of Bax, oligomerization of Bax and Bak, and expression of BH3-only p53-dependent genes, PUMA and NOXA. Inhibition of p53 and silencing of p53 unregulated modulator of apoptosis (PUMA) expression partly counteracted simvastatin-induced cell death, suggesting a role for p53-independent mechanisms. Simvastatin did not induce mitochondrial release of cytochrome c, but did promote release of inhibitor of apoptosis (IAP) proteins, Smac and Omi. Simvastatin also inhibited mitochondrial fission with the loss of mitochondrial Drp1, an essential component of mitochondrial fission machinery. Thus, simvastatin activates novel apoptosis pathways in lung mesenchymal cells involving p53, IAP inhibitor release, and disruption of mitochondrial fission.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Apoptosis/efectos de los fármacos , Citocromos c/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Pulmón/patología , Proteínas Mitocondriales/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Serina Endopeptidasas/metabolismo , Simvastatina/farmacología , Proteína p53 Supresora de Tumor/metabolismo , Western Blotting , Caspasa 8/metabolismo , Caspasa 9/metabolismo , Colesterol/metabolismo , Fibroblastos/efectos de los fármacos , Serina Peptidasa A2 que Requiere Temperaturas Altas , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Pulmón/metabolismo , Mesodermo/citología , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Músculo Liso Vascular/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Proteína X Asociada a bcl-2/metabolismo
11.
Respir Res ; 12: 113, 2011 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-21864337

RESUMEN

BACKGROUND: Bronchial fibroblasts contribute to airway remodelling, including airway wall fibrosis. Transforming growth factor (TGF)-ß1 plays a major role in this process. We previously revealed the importance of the mevalonate cascade in the fibrotic response of human airway smooth muscle cells. We now investigate mevalonate cascade-associated signaling in TGFß1-induced fibronectin expression by bronchial fibroblasts from non-asthmatic and asthmatic subjects. METHODS: We used simvastatin (1-15 µM) to inhibit 3-hydroxy-3-methlyglutaryl-coenzyme A (HMG-CoA) reductase which converts HMG-CoA to mevalonate. Selective inhibitors of geranylgeranyl transferase-1 (GGT1; GGTI-286, 10 µM) and farnesyl transferase (FT; FTI-277, 10 µM) were used to determine whether GGT1 and FT contribute to TGFß1-induced fibronectin expression. In addition, we studied the effects of co-incubation with simvastatin and mevalonate (1 mM), geranylgeranylpyrophosphate (30 µM) or farnesylpyrophosphate (30 µM). RESULTS: Immunoblotting revealed concentration-dependent simvastatin inhibition of TGFß1 (2.5 ng/ml, 48 h)-induced fibronectin. This was prevented by exogenous mevalonate, or isoprenoids (geranylgeranylpyrophosphate or farnesylpyrophosphate). The effects of simvastatin were mimicked by GGTI-286, but not FTI-277, suggesting fundamental involvement of GGT1 in TGFß1-induced signaling. Asthmatic fibroblasts exhibited greater TGFß1-induced fibronectin expression compared to non-asthmatic cells; this enhanced response was effectively reduced by simvastatin. CONCLUSIONS: We conclude that TGFß1-induced fibronectin expression in airway fibroblasts relies on activity of GGT1 and availability of isoprenoids. Our results suggest that targeting regulators of isoprenoid-dependent signaling holds promise for treating airway wall fibrosis.


Asunto(s)
Remodelación de las Vías Aéreas (Respiratorias)/efectos de los fármacos , Asma/metabolismo , Bronquios/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Simvastatina/farmacología , Factor de Crecimiento Transformador beta1/metabolismo , Adulto , Transferasas Alquil y Aril/antagonistas & inhibidores , Transferasas Alquil y Aril/metabolismo , Asma/patología , Bronquios/metabolismo , Bronquios/patología , Estudios de Casos y Controles , Células Cultivadas , Relación Dosis-Respuesta a Droga , Farnesiltransferasa/antagonistas & inhibidores , Farnesiltransferasa/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patología , Fibronectinas/metabolismo , Humanos , Leucina/análogos & derivados , Leucina/farmacología , Metionina/análogos & derivados , Metionina/farmacología , Ácido Mevalónico/metabolismo , Fosfatos de Poliisoprenilo/metabolismo , Sesquiterpenos/metabolismo , Factores de Tiempo , Adulto Joven
12.
Biochim Biophys Acta ; 1793(3): 546-60, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19321129

RESUMEN

Tumor necrosis factor alpha (TNF) is a cytokine that induces caspase-dependent (apoptotic) and caspase-independent (necrosis-like) cell death in different cells. We used the murine fibrosarcoma cell line model L929 and a stable L929 transfectant over-expressing a mutated dominant-negative form of BNIP3 lacking the C-terminal transmembrane (TM) domain (L929-DeltaTM-BNIP3) to test if TNF-induced cell death involved pro-apoptotic Bcl2 protein BNIP3. Treatment of cells with TNF in the absence of actinomycin D caused a rapid fall in the mitochondrial membrane potential (DeltaPsim) and a prompt increase in reactive oxygen species (ROS) production, which was significantly less pronounced in L929-DeltaTM-BNIP3. TNF did not cause the mitochondrial release of apoptosis inducing factor (AIF) and Endonuclease G (Endo-G) but provoked the release of cytochrome c, Smac/Diablo, and Omi/HtrA2 at similar levels in both L929 and in L929-DeltaTM-BNIP3 cells. We observed TNF-associated increase in the expression of BNIP3 in L929 that was mediated by nitric oxide and significantly inhibited by nitric oxide synthase inhibitor N5-(methylamidino)-L-ornithine acetate. In L929, lysosomal swelling and activation were markedly increased as compared to L929-DeltaTM-BNIP3 and could be inhibited by treatment with inhibitors to vacuolar H+-ATPase and cathepsins -B/-L. Together, these data indicate that TNF-induced cell death involves BNIP3, ROS production, and activation of the lysosomal death pathway.


Asunto(s)
Proteínas de la Membrana/genética , Proteínas Mitocondriales/genética , Factor de Necrosis Tumoral alfa/farmacología , Regulación hacia Arriba , Animales , Caspasa 9/metabolismo , Muerte Celular , Células Cultivadas , Citocromos c/metabolismo , Lisosomas/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Proteínas Mitocondriales/metabolismo , Óxido Nítrico Sintasa/metabolismo , Especies Reactivas de Oxígeno/metabolismo
13.
Curr Mol Med ; 8(8): 741-53, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19075672

RESUMEN

Caveolae are flask-shaped invaginations of the plasma membrane that are present in most structural cells. They owe their characteristic Omega-shape to complexes of unique proteins, the caveolins, which indirectly tether cholesterol and sphingolipid-enriched membrane microdomains to the cytoskeleton. Caveolins possess a unique scaffolding domain that anchors receptors, ion channels, second messenger producing enzymes, and effector kinases, thereby sequestering them to caveolae, and modulating cellular signaling and vesicular transport. The lungs express numerous caveolae and high levels of caveolins; therefore they likely play an important role in lung physiology. Indeed, recent and ongoing studies indicate important roles for caveolae and caveolins in the airway epithelium, airway smooth muscle, airway fibroblasts, airway inflammatory cells and the pulmonary vasculature. We review the role of caveolae and caveolins in lung cells and discuss their involvement in cellular signaling associated with asthma, COPD, lung cancer, idiopathic pulmonary fibrosis and pulmonary vascular defects.


Asunto(s)
Caveolas/fisiología , Caveolinas/fisiología , Fenómenos Fisiológicos Respiratorios , Animales , Secuencia de Bases , Caveolinas/genética , Matriz Extracelular/fisiología , Humanos , Neoplasias Pulmonares/etiología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/fisiopatología , Familia de Multigenes , Óxido Nítrico/metabolismo , Fibrosis Pulmonar/etiología , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/fisiopatología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Fenómenos Fisiológicos Respiratorios/genética
14.
Br J Pharmacol ; 167(3): 548-60, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22551156

RESUMEN

BACKGROUND AND PURPOSE: Acute silencing of caveolin-1 (Cav-1) modulates receptor-mediated contraction of airway smooth muscle. Moreover, COX-2- and 5-lipoxygenase (5-LO)-derived prostaglandin and leukotriene biosynthesis can influence smooth muscle reactivity. COX-2 half-life can be prolonged through association with Cav-1. We suggested that lack of Cav-1 modulated levels of COX-2 which in turn modulated tracheal contraction, when arachidonic acid signalling was disturbed by inhibition of COX-2. EXPERIMENTAL APPROACH: Using tracheal rings from Cav-1 knockout (KO) and wild-type mice (B6129SF2/J), we measured isometric contractions to methacholine and used PCR, immunoblotting and immunohistology to monitor expression of relevant proteins. KEY RESULTS: Tracheal rings from Cav-1 KO and wild-type mice exhibited similar responses, but the COX-2 inhibitor, indomethacin, increased responses of tracheal rings from Cav-1 KO mice to methacholine. The phospholipase A2 inhibitor, eicosatetraynoic acid, which inhibits formation of both COX-2 and 5-LO metabolites, had no effect on wild-type or Cav-1 KO tissues. Indomethacin-mediated hyperreactivity was ablated by the LTD4 receptor antagonist (montelukast) and 5-LO inhibitor (zileuton). The potentiating effect of indomethacin on Cav-1 KO responses to methacholine was blocked by epithelial denudation. Immunoprecipitation showed that COX-2 binds Cav-1 in wild-type lungs. Immunoblotting and qPCR revealed elevated levels of COX-2 and 5-LO protein, but not COX-1, in Cav-1 KO tracheas, a feature that was prevented by removal of the epithelium. CONCLUSION AND IMPLICATIONS: The indomethacin-induced hypercontractility observed in Cav-1 KO tracheas was linked to increased expression of COX-2 and 5-LO, which probably enhanced arachidonic acid shunting and generation of pro-contractile leukotrienes when COX-2 was inhibited.


Asunto(s)
Araquidonato 5-Lipooxigenasa/metabolismo , Caveolina 1/genética , Ciclooxigenasa 2/metabolismo , Indometacina/farmacología , Animales , Araquidonato 5-Lipooxigenasa/efectos de los fármacos , Araquidonato 5-Lipooxigenasa/genética , Ácido Araquidónico/metabolismo , Hiperreactividad Bronquial/patología , Broncoconstricción/efectos de los fármacos , Ciclooxigenasa 2/efectos de los fármacos , Ciclooxigenasa 2/genética , Inhibidores de la Ciclooxigenasa/farmacología , Células Epiteliales/metabolismo , Femenino , Regulación de la Expresión Génica , Inmunoprecipitación , Cloruro de Metacolina/farmacología , Ratones , Ratones Noqueados , Músculo Liso/efectos de los fármacos , Músculo Liso/metabolismo , Tráquea/efectos de los fármacos , Tráquea/metabolismo
15.
PLoS One ; 6(1): e16523, 2011 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-21304979

RESUMEN

Statins inhibit the proximal steps of cholesterol biosynthesis, and are linked to health benefits in various conditions, including cancer and lung disease. We have previously investigated apoptotic pathways triggered by statins in airway mesenchymal cells, and identified reduced prenylation of small GTPases as a primary effector mechanism leading to p53-mediated cell death. Here, we extend our studies of statin-induced cell death by assessing endpoints of both apoptosis and autophagy, and investigating their interplay and coincident regulation. Using primary cultured human airway smooth muscle (HASM) and human airway fibroblasts (HAF), autophagy, and autophagosome formation and flux were assessed by transmission electron microscopy, cytochemistry (lysosome number and co-localization with LC3) and immunoblotting (LC3 lipidation and Atg12-5 complex formation). Chemical inhibition of autophagy increased simvastatin-induced caspase activation and cell death. Similarly, Atg5 silencing with shRNA, thus preventing Atg5-12 complex formation, increased pro-apoptotic effects of simvastatin. Simvastatin concomitantly increased p53-dependent expression of p53 up-regulated modulator of apoptosis (PUMA), NOXA, and damage-regulated autophagy modulator (DRAM). Notably both mevalonate cascade inhibition-induced autophagy and apoptosis were p53 dependent: simvastatin increased nuclear p53 accumulation, and both cyclic pifithrin-α and p53 shRNAi partially inhibited NOXA, PUMA expression and caspase-3/7 cleavage (apoptosis) and DRAM expression, Atg5-12 complex formation, LC3 lipidation, and autophagosome formation (autophagy). Furthermore, the autophagy response is induced rapidly, significantly delaying apoptosis, suggesting the existence of a temporally coordinated p53 regulation network. These findings are relevant for the development of statin-based therapeutic approaches in obstructive airway disease.


Asunto(s)
Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Mesodermo/citología , Ácido Mevalónico/farmacología , Sistema Respiratorio/citología , Proteína p53 Supresora de Tumor/fisiología , Células Cultivadas , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Mesodermo/efectos de los fármacos , Sistema Respiratorio/efectos de los fármacos , Simvastatina/farmacología
16.
Am J Physiol Lung Cell Mol Physiol ; 294(1): L57-68, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17993586

RESUMEN

Airway smooth muscle (ASM) cells may contribute to asthma pathogenesis through their capacity to switch between a synthetic/proliferative and a contractile phenotype. The multimeric dystrophin-glycoprotein complex (DGC) spans the sarcolemma, linking the actin cytoskeleton and extracellular matrix. The DGC is expressed in smooth muscle tissue, but its functional role is not fully established. We tested whether contractile phenotype maturation of human ASM is associated with accumulation of DGC proteins. We compared subconfluent, serum-fed cultures and confluent cultures subjected to serum deprivation, which express a contractile phenotype. Western blotting confirmed that beta-dystroglycan, beta-, delta-, and epsilon-sarcoglycan, and dystrophin abundance increased six- to eightfold in association with smooth muscle myosin heavy chain (smMHC) and calponin accumulation during 4-day serum deprivation. Immunocytochemistry showed that the accumulation of DGC subunits was specifically localized to a subset of cells that exhibit robust staining for smMHC. Laminin competing peptide (YIGSR, 1 microM) and phosphatidylinositol 3-kinase (PI3K) inhibitors (20 microM LY-294002 or 100 nM wortmannin) abrogated the accumulation of smMHC, calponin, and DGC proteins. These studies demonstrate that the accumulation of DGC is an integral feature for phenotype maturation of human ASM cells. This provides a strong rationale for future studies investigating the role of the DGC in ASM smooth muscle physiology in health and disease.


Asunto(s)
Distrofina/genética , Glicoproteínas/genética , Músculo Liso/fisiología , Fenómenos Fisiológicos Respiratorios , Línea Celular , Senescencia Celular , Distroglicanos/análisis , Marcadores Genéticos , Humanos , Inmunohistoquímica , Músculo Liso/citología , Fenotipo , Subunidades de Proteína/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sarcoglicanos/análisis , Telomerasa/análisis
17.
Am J Physiol Lung Cell Mol Physiol ; 293(6): L1406-18, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17890325

RESUMEN

Contractile responses of airway smooth muscle (ASM) determine airway resistance in health and disease. Caveolae microdomains in the plasma membrane are marked by caveolin proteins and are abundant in contractile smooth muscle in association with nanospaces involved in Ca(2+) homeostasis. Caveolin-1 can modulate localization and activity of signaling proteins, including trimeric G proteins, via a scaffolding domain. We investigated the role of caveolae in contraction and intracellular Ca(2+) ([Ca(2+)](i)) mobilization of ASM induced by the physiological muscarinic receptor agonist, acetylcholine (ACh). Human and canine ASM tissues and cells predominantly express caveolin-1. Muscarinic M(3) receptors (M(3)R) and Galpha(q/11) cofractionate with caveolin-1-rich membranes of ASM tissue. Caveolae disruption with beta-cyclodextrin in canine tracheal strips reduced sensitivity but not maximum isometric force induced by ACh. In fura-2-loaded canine and human ASM cells, exposure to methyl-beta-cyclodextrin (mbetaCD) reduced sensitivity but not maximum [Ca(2+)](i) induced by ACh. In contrast, both parameters were reduced for the partial muscarinic agonist, pilocarpine. Fluorescence microscopy revealed that mbetaCD disrupted the colocalization of caveolae-1 and M(3)R, but [N-methyl-(3)H]scopolamine receptor-binding assay revealed no effect on muscarinic receptor availability or affinity. To dissect the role of caveolin-1 in ACh-induced [Ca(2+)](i) flux, we disrupted its binding to signaling proteins using either a cell-permeable caveolin-1 scaffolding domain peptide mimetic or by small interfering RNA knockdown. Similar to the effects of mbetaCD, direct targeting of caveolin-1 reduced sensitivity to ACh, but maximum [Ca(2+)](i) mobilization was unaffected. These results indicate caveolae and caveolin-1 facilitate [Ca(2+)](i) mobilization leading to ASM contraction induced by submaximal concentrations of ACh.


Asunto(s)
Señalización del Calcio , Caveolas/metabolismo , Espacio Intracelular/metabolismo , Contracción Muscular/fisiología , Músculo Liso/metabolismo , Receptor Muscarínico M3/metabolismo , Sistema Respiratorio/metabolismo , Acetilcolina/farmacología , Animales , Señalización del Calcio/efectos de los fármacos , Caveolas/efectos de los fármacos , Caveolina 1/química , Células Cultivadas , Citosol/efectos de los fármacos , Citosol/metabolismo , Perros , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Humanos , Espacio Intracelular/efectos de los fármacos , Células Musculares/citología , Células Musculares/efectos de los fármacos , Contracción Muscular/efectos de los fármacos , Músculo Liso/citología , Músculo Liso/efectos de los fármacos , Músculo Liso/ultraestructura , N-Metilescopolamina/metabolismo , Estructura Terciaria de Proteína , ARN Interferente Pequeño/metabolismo , Sistema Respiratorio/citología , Sistema Respiratorio/efectos de los fármacos , Sistema Respiratorio/ultraestructura , Tráquea/citología , Tráquea/efectos de los fármacos , Tráquea/metabolismo , Tritio/metabolismo , beta-Ciclodextrinas/farmacología
18.
Can J Physiol Pharmacol ; 81(3): 220-33, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12733821

RESUMEN

We investigated the functional interdependence of sarco-endoplasmic reticulum Ca2+ ATPase isoform 1 and ryanodine receptor isoform 1 in heavy sarcoplasmic reticulum membranes by synchronous fluorescence determination of extravesicular Ca2+ transients and catalytic activity. Under conditions of dynamic Ca2+ exchange ATPase catalytic activity was well coordinated to ryanodine receptor activation/inactivation states. Ryanodine-induced activation of Ca2+ release channel leaks also produced marked ATPase activation in the absence of measurable increases in bulk free extravesicular Ca2+. This suggested that Ca2+ pumps are highly sensitive to Ca2+ release channel leak status and potently buffer Ca2+ ions exiting cytoplasmic openings of ryanodine receptors. Conversely, ryanodine receptor activation was dependent on Ca2+-ATPase pump activity. Ryanodine receptor activation by cytosolic Ca2+ was (i) inversely proportional to luminal Ca2+ load and (ii) dependent upon the rate of presentation of cytosolic Ca2+. Progressive Ca2+ filling coincided with progressive loss of Ca2+ sequestration rates and at a threshold loading, ryanodine-induced Ca2+ release produced small transient reversals of catalytic activity. These data indicate that attainment of threshold luminal Ca2+ loads coordinates sensitization of Ca2+ release channels with autogenic inhibition of Ca2+ pumping. This suggests that Ca2+-dependent control of Ca2+ release in intact heavy sarcoplasmic reticulum membranes involves a Ca2+-mediated "cross-talk" between sarco-endoplasmic reticulum Ca2+ ATPase isoform 1 and ryanodine receptor isoform 1.


Asunto(s)
ATPasas Transportadoras de Calcio/metabolismo , Calcio/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Retículo Sarcoplasmático/metabolismo , Animales , Transporte Biológico , Western Blotting , Canales de Calcio/efectos de los fármacos , Canales de Calcio/fisiología , Señalización del Calcio , Catálisis , Electroforesis en Gel de Poliacrilamida , Colorantes Fluorescentes , Técnicas In Vitro , Membranas Intracelulares/metabolismo , Isoformas de Proteínas/metabolismo , Conejos , Rianodina/farmacología , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA