Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Sci Adv ; 10(38): eado4274, 2024 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-39303038

RESUMO

Recurrent somatic mutations in spliceosome factor 3b subunit 1 (SF3B1) are identified in hematopoietic malignancies, with SF3B1-K700E being the most common one. Here, we show that regulatory T cell (Treg)-specific expression of SF3B1-K700E (Sf3b1K700Efl/+/Foxp3YFP-Cre) results in spontaneous autoimmune phenotypes. CD4+ T cells from Sf3b1K700Efl/+/Foxp3YFP-Cre mice display defective Treg differentiation and inhibitory function, which is demonstrated by failed prevention of adoptive transfer colitis by Sf3b1K700Efl/+/Foxp3YFP-Cre Tregs. Mechanically, SF3B1-K700E induces an aberrant splicing event that results in reduced expression of a cell proliferation regulator Anapc13 due to the insertion of a 231-base pair DNA fragment to the 5' untranslated region. Forced expression of the Anapc13 gene restores the differentiation and ability of Sf3b1K700Efl/+/Foxp3YFP-Cre Tregs to prevent adoptive transfer colitis. In addition, acute myeloid leukemia grows faster in aged, but not young, Sf3b1K700Efl/+/Foxp3YFP-Cre mice compared to Foxp3YFP-Cre mice. Our results highlight the impact of cancer-associated SF3B1 mutation on immune responses, which affect cancer development.


Assuntos
Mutação , Fatores de Processamento de RNA , Linfócitos T Reguladores , Animais , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Fatores de Processamento de RNA/genética , Fatores de Processamento de RNA/metabolismo , Camundongos , Splicing de RNA , Humanos , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Neoplasias/genética , Neoplasias/imunologia , Diferenciação Celular , Colite/genética , Colite/imunologia
2.
Crit Rev Immunol ; 44(6): 111-126, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38848298

RESUMO

Steroid receptor coactivator (SRC) family members (SRC1, SRC2 and SRC3) are transcriptional co-regulators. SRCs orchestrate gene transcription by inducing transactivation of nuclear receptors and other transcription factors. Overexpression of SRCs is widely implicated in a range of cancers, especially hormone-related cancers. As coactivators, SRCs regulate multiple metabolic pathways involved in tumor growth, invasion, metastasis, and chemo-resistance. Emerging evidence in recent years suggest that SRCs also regulate maturation, differentiation, and cytotoxicity of T cells by controlling metabolic activities. In this review, we summarize the current understanding of the function of SRCs in T cells as well as cancer cells. Importantly, the controversies of targeting SRCs for cancer immunotherapy as well as possible reconciliation strategies are also discussed.


Assuntos
Imunoterapia , Neoplasias , Linfócitos T , Humanos , Neoplasias/terapia , Neoplasias/imunologia , Neoplasias/metabolismo , Imunoterapia/métodos , Animais , Linfócitos T/imunologia , Linfócitos T/metabolismo , Coativadores de Receptor Nuclear/metabolismo , Coativadores de Receptor Nuclear/imunologia
3.
Cancer Immunol Res ; 11(10): 1414-1431, 2023 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-37540802

RESUMO

Nuclear receptor coactivator 2 (Ncoa2) is a member of the Ncoa family of coactivators, and we previously showed that Ncoa2 regulates the differentiation of induced regulatory T cells. However, it remains unknown if Ncoa2 plays a role in CD8+ T-cell function. Here, we show that Ncoa2 promotes CD8+ T cell-mediated immune responses against tumors by stimulating T-cell activation via upregulating PGC-1α expression to enhance mitochondrial function. Mice deficient in Ncoa2 in T cells (Ncoa2fl/fl/CD4Cre) displayed defective immune responses against implanted MC38 tumors, which associated with significantly reduced tumor-infiltrating CD8+ T cells and decreased IFNγ production. Consistently, CD8+ T cells from Ncoa2fl/fl/CD4Cre mice failed to reject tumors after adoptive transfer into Rag1-/- mice. Further, in response to TCR stimulation, Ncoa2fl/fl/CD4Cre CD8+ T cells failed to increase mitochondrial mass, showed impaired oxidative phosphorylation, and had lower expression of PGC-1α, a master regulator of mitochondrial biogenesis and function. Mechanically, T-cell activation-induced phosphorylation of CREB triggered the recruitment of Ncoa2 to bind to enhancers, thus, stimulating PGC-1α expression. Forced expression of PGC-1α in Ncoa2fl/fl/CD4Cre CD8+ T cells restored mitochondrial function, T-cell activation, IFNγ production, and antitumor immunity. This work informs the development of Ncoa2-based therapies that modulate CD8+ T cell-mediated antitumor immune responses.


Assuntos
Mitocôndrias , Neoplasias , Animais , Camundongos , Linfócitos T CD8-Positivos/metabolismo , Mitocôndrias/metabolismo , Neoplasias/metabolismo , Coativador 2 de Receptor Nuclear/metabolismo , Regulação para Cima
4.
Biomed Pharmacother ; 150: 113071, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35658237

RESUMO

BACKGROUND: Nicotinamide adenine dinucleotide (NAD+), a coenzyme that plays crucial roles in many cellular processes, is a potential therapeutic target for various diseases. Dihydronicotinamide riboside (NRH), a novel reduced form of nicotinamide riboside, has emerged as a potent NAD+ precursor. Here, we studied the protective effects and underlying mechanism of NRH on aminoglycoside-induced ototoxicity. METHODS: Auditory function and hair-cell (HC) morphology were examined to assess the effects of NRH on kanamycin-induced hearing loss. The pharmacokinetic parameters of NRH were measured in plasma and the cochlea using liquid chromatography tandem mass spectrometry. NAD+ levels in organ explant cultures were assessed to compare NRH with known NAD+ precursors. Immunofluorescence analysis was performed to detect reactive oxygen species (ROS) and apoptosis. We analyzed SIRT1 and 14-3-3 protein expression. EX527 and resveratrol were used to investigate the role of SIRT1 in the protective effect of NRH against kanamycin-induced ototoxicity. RESULTS: NRH alleviated kanamycin-induced HC damage and attenuated hearing loss in mice. NRH reduced gentamicin-induced vestibular HC loss. Compared with NAD and NR, NRH produced more NAD+ in cochlear HCs and significantly ameliorated kanamycin-induced oxidative stress and apoptosis. NRH rescued the aminoglycoside-induced decreases in SIRT1 and 14-3-3 protein expression. Moreover, EX527 antagonized the protective effect of NRH on kanamycin-induced HC loss by inhibition of SIRT1, while resveratrol alleviated HC damage caused by EX527. CONCLUSIONS: NRH ameliorates aminoglycoside-induced ototoxicity by inhibiting HC apoptosis by activating SIRT1 and decreasing ROS. NRH is an effective therapeutic option for aminoglycoside-induced ototoxicity.


Assuntos
Perda Auditiva , Ototoxicidade , Proteínas 14-3-3/metabolismo , Aminoglicosídeos/metabolismo , Aminoglicosídeos/toxicidade , Animais , Antibacterianos/farmacologia , Cóclea , Perda Auditiva/induzido quimicamente , Perda Auditiva/prevenção & controle , Canamicina/farmacologia , Camundongos , NAD/metabolismo , Niacinamida/análogos & derivados , Ototoxicidade/prevenção & controle , Compostos de Piridínio , Espécies Reativas de Oxigênio/metabolismo , Resveratrol/farmacologia , Sirtuína 1/metabolismo
5.
Stem Cell Res ; 61: 102783, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35413565

RESUMO

Otosclerosis is caused by abnormal bone remodeling in the middle ear, resulting in progressive hearing loss, dizziness, balance problems, and tinnitus. Previous infection, stress fractures of the bony tissue surrounding the inner ear, immune disorders, and genetic factors are believed to contribute to this disease. Currently, no effective drug treatment for otosclerosis is known. Herein, we generated an induced pluripotent stem cell line from the peripheral blood mononuclear cells of an otosclerosis patient. The cell line exhibited normal morphology, karyotype, and pluripotency marker expression. A teratoma assay revealed successful differentiation into all three germ layers.


Assuntos
Células-Tronco Pluripotentes Induzidas , Otosclerose , Teratoma , Linhagem Celular , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Leucócitos Mononucleares , Otosclerose/metabolismo , Teratoma/metabolismo
6.
Exp Ther Med ; 22(4): 1057, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34434271

RESUMO

p16INK4a (p16) inhibits the vital G1 to S phase transition during cell cycle progression through the p16/cyclin D1/CDK4/retinoblastoma(Rb)/E2F1 pathway. Hyperoxia can suppress the G1/S checkpoint and induce more lung fibroblasts (LFs) to transition from the G1 phase to the S phase and undergo cell proliferation. The present study investigated the rate of p16 gene promoter methylation and the protein expression levels of p16, cyclin D1, CDK4, Rb and E2F1 in LFs from the lungs of rats exposed to hyperoxia and normoxia on postnatal days 3, 7 and 14. In the hyperoxia-exposed group, the methylation rate was 50 and 80% on days 7 and 14, respectively. Cyclin D1 and CDK4 overexpression was associated with p16 loss and Rb inactivation by phosphorylation. Rb phosphorylation induced E2F1 release in the G1 phase, which promoted cell proliferation. No methylation was observed in the normoxia-exposed group. These observations suggested that p16 loss may stimulate aberrant LF proliferation via the p16/cyclin D1/CDK4/Rb/E2F1 pathway.

7.
Mol Med Rep ; 20(6): 4867-4874, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31702808

RESUMO

Hyperoxia may cause pulmonary fibrosis in neonates and is characterized by the epithelial­to­mesenchymal transition (EMT) of alveolar epithelial cells. The placental growth factor (PLGF) gene is a member of the vascular endothelial growth factor family and is highly expressed in lung tissues that have been exposed to hyperoxia. The aim of the present study was to assess the role of PLGF in the EMT of lung tissue. Lung tissue exhibiting low PLGF expression was obtained by injecting rats exposed to hyperoxia with a PLGF­silencing lentiviral plasmid. Western blot analysis and immunohistochemistry revealed that expression levels of the EMT­related protein epithelial­cadherin were increased, whereas its inhibitor protein zinc­finger E­box binding homeobox 2 was decreased in these rats. These data demonstrated that PLGF silencing may significantly mitigate hyperoxia­induced EMT in rat lung tissue. Additionally, an increase in phosphorylated­p38 MAPK protein expression indicated that PLGF may be able to regulate hyperoxia­induced lung injury in rats via the p38 MAPK pathway.


Assuntos
Transição Epitelial-Mesenquimal/fisiologia , Inativação Gênica , Hiperóxia/complicações , Lesão Pulmonar/metabolismo , Fator de Crescimento Placentário/genética , Fator de Crescimento Placentário/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Células Epiteliais Alveolares/metabolismo , Animais , Caderinas/metabolismo , Modelos Animais de Doenças , Feminino , Pulmão/metabolismo , Pulmão/patologia , Lesão Pulmonar/patologia , Masculino , Gravidez , Ratos , Ratos Sprague-Dawley , Homeobox 2 de Ligação a E-box com Dedos de Zinco/metabolismo
8.
J Biomed Sci ; 26(1): 62, 2019 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-31470850

RESUMO

BACKGROUND: Loss of ovarian function, as in menopause or after ovariectomy (OVX), is closely associated with obesity and white adipose tissue (WAT) inflammation. Estrogen replacement protects against postmenopausal obesity but increases the risks of carcinogenesis. In the present study, we investigated the effects of long-term treatment of raloxifene (RAL), a selective estrogen receptor modulator, on the features of estrogen deficiency-induced obesity and explored the involvement of canonical and non-canonical Wnt regulation in vivo and in vitro. METHODS: Adult female rats received bilateral OVX and divided into 5 groups: (1) Sham, (2) OVX, (3) OVX + E2: OVX rats were administered with E2 (50 µg/kg, s.c., 3 times/week), (4) OVX + RAL: OVX rats were treated with RAL (gavage, 1 mg/kg/day) suspended in 0.8% carboxymethylcellulose (CMC), (5) OVX + CMC: 0.8% CMC as vehicle control. All treatments were given for 8 weeks beginning at 1 week after OVX. In 3 T3-L1 cells, the effects of RAL on adipogenesis and lipopolysaccharide (LPS)-induced inflammation were evaluated. RESULTS: Treatment with RAL significantly decreased body weight, visceral fat pad mass, adipocyte size and plasma levels of glucose but increased plasma adiponectin. RAL reduced the elevation of HIF-1α, VEGF-A and proinflammatory cytokines (MCP-1 and TNF-α) expression by inhibition of NF-κB p65 and JNK cascades in retroperitoneal WAT. This anti-inflammatory capacity of RAL may result from upregulation of secreted frizzle-related protein 5 (SFRP5), an adipokine that repressed Wnt5a signaling. Furthermore, RAL inhibited adipogenic factors such as PPAR-γ, C/EBP-α, and FABP4, and preserved canonical Wnt10b/ß-catenin protein expression. In 3 T3-L1 adipocytes, RAL (20 µM) diminished lipid accumulation and inhibited adipogenic factors accompanied with the induction of ß-catenin, which were effectively reversed by the ß-catenin inhibitor IWR-1-endo. In addition, RAL reduced LPS-induced NF-κB p65 and p-IκB expression as well as TNF-α secretion. Suppression of SFRP5 by small interfering RNA significantly abrogated the anti-inflammatory effects of RAL. CONCLUSIONS: Distinct activation of canonical ß-catenin on inhibition of adipogenesis and non-canonical SFRP5 on suppression of WAT inflammation may contribute to the beneficial effects of RAL. Therefore, this study provides a rationale for the therapeutic potential of RAL for postmenopausal obesity.


Assuntos
Adipogenia/efeitos dos fármacos , Tecido Adiposo/efeitos dos fármacos , Inflamação/induzido quimicamente , Cloridrato de Raloxifeno/farmacologia , Receptores de Estrogênio/antagonistas & inibidores , Moduladores Seletivos de Receptor Estrogênico/farmacologia , Proteínas Wnt/genética , Células 3T3-L1 , Tecido Adiposo/imunologia , Animais , Feminino , Regulação da Expressão Gênica , Lipopolissacarídeos/farmacologia , Camundongos , Ovariectomia , Ratos , Ratos Wistar , Proteínas Wnt/metabolismo , Proteína Wnt1
9.
Biotechnol Lett ; 41(6-7): 763-777, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31025146

RESUMO

OBJECTIVES: An efficient bacterial surface display system based on the anchoring motif derived from Escherichia coli (E. coli) outer membrane protease OmpT was developed in this study. RESULTS: Referring to the classical Lpp-OmpA (LOA) display system, the signal peptide and nine amino acids of mature Lpp were fused to the transmembrane domain comprising five ß-strands of truncated OmpT to generate a novel Lpp-OmpT (LOT) display system. The C-terminal fusion strategy was used to fuse a small peptide (His tag) and red fluorescent protein (mCherry) to the C-terminus of LOT. Cell surface exposure of His tag and mCherry were compared between the LOA and LOT display systems. E. coli expressing LOT-His tag adsorbed more Cu2+ than E. coli expressing LOA-His tag. E. coli expressing both LOT-mCherry-His tag and LOA-mCherry-His tag adhered to Cu2+ chelating sepharose beads, and adhered cells could be dissociated from the beads after excess Cu2+ treatment. More importantly, compared with the LOA system, a higher amount of LOT-mCherry-His tag hybrid protein was demonstrated to be localized at the outer membrane by both fluorescence spectrophotometric determination of cell fractions and cell-surface immunofluorescence assay. CONCLUSIONS: These results suggest that genetically modified OmpT can be used as a potential anchoring motif to efficiently and stably display polypeptides and proteins, and that the LOT system could be used in a variety of biotechnological and industrial processes.


Assuntos
Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/metabolismo , Técnicas de Visualização da Superfície Celular/métodos , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Peptídeo Hidrolases/genética , Peptídeo Hidrolases/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Engenharia Metabólica/métodos
10.
Cytotechnology ; 70(2): 751-760, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29340836

RESUMO

Lung fibrosis is an ultimate consequence of bronchopulmonary dysplasia (BPD) which shows the excessive proliferation of lung fibroblasts (LFs). To find a better model for studying the role of LFs in hyperoxia-induced lung fibrosis at the cellular level, we isolated LFs from the lung tissue of hyperoxia- and normoxia-exposed rat lungs on postnatal days 7, 14 and 21 for primary culture to study their proliferative behavior. In the present study, the LF predominance was > 95% in our culture method. The LFs isolated from rats exposed to hyperoxia in vivo showed significantly greater proliferation than that from normoxia-exposed rats. Flow cytometry revealed that percentage of LFs in S and G2/M stage increased, and proportion in the G0/G1 stage declined at the same time. A greater presence of myofibroblasts in LFs isolated from rats exposed to hyperoxia compared with those exposed to normoxia. In addition, elevated collagen level, transforming growth factor-ß and connective tissue growth factor protein expression in conditioned medium were also found in hyperoxia LFs. These data demonstrate that hyperoxia promotes LFs proliferation, myofibroblast transdifferentiation and collagen synthesis in a time-dependent manner. The primary culture of LFs from hyperoxia-exposed rats is a feasible method for studying the pathogenesis and treatment of lung fibrosis caused by BPD at the cellular level.

11.
Pediatr Res ; 83(3): 723-730, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29166374

RESUMO

BackgroundP16 methylation plays an important role in the pathogenesis of hyperoxia-induced lung fibrosis. 5-aza-2'-deoxycytidine (5-aza-CdR) is a major methyltransferase-specific inhibitor. In this study, the effects of 5-aza-CdR on a hyperoxia-induced lung fibrosis in neonatal rats were investigated.MethodsRat pups were exposed to 85% O2 for 21 days of and received intraperitoneal injections of 5-aza-CdR or normal saline (NS) once every other day. Survival rates and lung coefficients were calculated. Hematoxylin-eosin staining was performed to analyze the degree of lung fibrosis. Collagen content and TGF-ß1 levels were determined. A methylation-specific polymerase chain reaction and western blotting were performed to determine P16 methylation status and P16, cyclin D1, and E2F1 protein expression.Results5-aza-CdR treatment during hyperoxia significantly improved the survival rate and weight gain, while it decreases the degree of lung fibrosis and levels of hydroxyproline and TGF-ß1. Hyperoxia induced abnormal P16 methylation and 5-aza-CdR effectively reversed the hypermethylation of P16. Expression of the P16 protein in lung tissues was enhanced, while cyclin D1 and E2F1 protein were reduced by 5-aza-CdR treatment during hyperoxia.ConclusionThese data show that 5-aza-CdR attenuated lung fibrosis in neonatal rats exposed to hyperoxia by lowering hydroxyproline and TGF-ß1 expression and via re-expression of P16 in neonatal rats.


Assuntos
Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Decitabina/farmacologia , Regulação da Expressão Gênica , Hiperóxia/tratamento farmacológico , Pulmão/efeitos dos fármacos , Fator de Crescimento Transformador beta1/metabolismo , Animais , Animais Recém-Nascidos , Metilação de DNA/efeitos dos fármacos , Feminino , Fibrose/tratamento farmacológico , Hidroxiprolina/metabolismo , Pulmão/patologia , Ratos , Ratos Sprague-Dawley
12.
Pediatr Neonatol ; 58(2): 122-127, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-27663361

RESUMO

BACKGROUND: A persistent increase in the number of lung fibroblasts (LFs) is found in the interstitium of the lungs of infants with bronchopulmonary dysplasia (BPD), which leads to lung fibrosis. P16 methylation plays an important role in the pathogenesis of BPD. 5-aza-2'-deoxycytidine (5-aza-CdR) is a major methyltransferase-specific inhibitor. This study investigated the effects of 5-aza-CdR on LFs in vitro from a hyperoxia-induced lung fibrosis model in newborn rats. METHODS: Methylation-specific polymerase chain reaction (PCR) and Western blotting were performed to determine P16 gene methylation status and protein expression after LFs were treated with 0 µmol/L, 0.5 µmol/L, 1.0 µmol/L, and 5.0 µmol/L 5-aza-CdR for 120 hours. Proliferation was assessed by an MTT assay after LFs were treated with 0 µmol/L, 0.5 µmol/L, 1.0 µmol/L, and 5.0 µmol/L 5-aza-CdR for 24 hours, 48 hours, 72 hours, 96 hours, and 120 hours. At the final time point, cells were also analyzed by flow cytometry to identify any change in their cell cycle profiles. RESULTS: A methylated P16 gene promoter was detected in hyperoxia LFs. Following treatment with 5-aza-CdR, partial methylation and demethylation was detected. The expression protein's level of the P16 gene was significantly higher in the 5.0 µmol/L 5-aza-CdR-treated group compared with that in the control group (p < 0.01). The cell growth rate at each tested time point was lower in the 5-aza-CdR-treated group compared with that in the control group after 72 hours (p < 0.01). Flow cytometry revealed that the cells in the 1.0 µmol/L and 5.0 µmol/L 5-aza-CdR-treated groups were apparently arrested in the G0/G1 phase and that the number of cells in the S phase was significantly lower than the control group (p < 0.01). CONCLUSION: 5-aza-CdR inhibits the growth of the LFs in hyperoxia-induced neonatal BPD rats in vitro by demethylating the P16 gene.


Assuntos
Azacitidina/análogos & derivados , Proliferação de Células/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Fibroblastos/efeitos dos fármacos , Pulmão/citologia , Animais , Animais Recém-Nascidos , Azacitidina/farmacologia , Displasia Broncopulmonar/metabolismo , Decitabina , Modelos Animais de Doenças , Hiperóxia/complicações , Ratos , Ratos Sprague-Dawley
13.
Int Immunopharmacol ; 38: 167-74, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27280587

RESUMO

Although supplemental high-level oxygen treatment can promote the survival of premature infants, hyperoxia may adversely induce acute lung injury (ALI) in newborns. Our prior work illustrated that hyperoxic exposure could enhance the release of placental growth factor (PLGF) in the lungs of neonatal rats. We therefore postulated that PLGF contributed to hyperoxic ALI in newborns and evaluated the anti-PLGF treatment mediated by systematic delivery of lentivirus in hyperoxic ALI in this study. Lentivirus particles containing PLGF specific shRNA were injected into neonatal rats prior to hyperoxic exposure (90% oxygen for 72h) to inhibit PLGF expression. Hyperoxia induced oxidative damages in lung tissues as evidenced by the increased malondialdehyde and myeloperoxidase, and the decreased antioxidant superoxide dismutase. Also, hyperoxia caused excessive infiltration of inflammatory cells and overproduction of proinflammatory cytokines (tumor necrosis factor-α, interleukin-1ß and interleukin-6) in rat lung tissue. These pathological alterations were partly reversed by PLGF shRNA delivery. The expression levels and activities of metalloproteinase (MMP)-2 and MMP9 were up-regulated in response to hyperoxia, whereas down-regulated when PLGF was inhibited. Moreover, PLGF shRNA inhibited nuclear factor kappa B (NFκB) signaling delivery in hyperoxic rat lungs. Additionally, exogenous PLGF-induced activation of MMPs in rat RLE-6TN alveolar epithelial cells was suppressed by NFκB inhibitor pyrrolidine dithiocarbamate. These results suggest that therapy targeting PLGF may be beneficial for infants with hyperoxic ALI.


Assuntos
Lesão Pulmonar Aguda/metabolismo , Células Epiteliais/fisiologia , Hiperóxia/metabolismo , Pulmão/patologia , Fator de Crescimento Placentário/metabolismo , Animais , Animais Recém-Nascidos , Linhagem Celular , Citocinas/metabolismo , Células Epiteliais/efeitos dos fármacos , Feminino , Masculino , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , NF-kappa B/antagonistas & inibidores , Fator de Crescimento Placentário/genética , Pirrolidinas/farmacologia , RNA Interferente Pequeno/genética , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Superóxido Dismutase/metabolismo , Tiocarbamatos/farmacologia
14.
Clin Exp Pharmacol Physiol ; 43(9): 834-43, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27187184

RESUMO

Life-saving mechanical ventilation can also cause lung injury through the overproduction of reactive oxygen species (ROS), leading to bronchopulmonary dysplasia (BPD)-like symptoms in preterm infants. It is reported that the autophagic protein microtubule-associated protein-1 light chain (LC)-3B can confer protection against hyperoxia-induced DNA damage in lung alveolar epithelium. However, its role in the transdifferentiation of type II alveolar epithelial cells (AECIIs) to type I cells (AECIs) is unclear and requires further investigation. In this study, newborn Sprague-Dawley rats were exposed to 90% oxygen for up to 14 days to mimic BPD in human infants, with neonatal pups exposed to room air (21% oxygen) as controls. Primary rat AECIIs were cultured under hyperoxic conditions for up to 24 hours to further investigate the underlying mechanisms. This study found that hyperoxia promoted a significant and time-dependent increase of AECII marker surfactant protein (SP)-C in the lung. The increase of AECI marker T1α was repressed by hyperoxia during lung development. These results indicated an impaired AECII transdifferentiation. Pulmonary ROS concentration and expression of autophagic protein LC-3B were increased gradually in response to hyperoxia exposure. Furthermore, AECIIs produced more ROS when cultured under hyperoxic conditions in vitro. Both the LC3B expression and the conversion from LC3BI to LC3BII were enhanced in hyperoxic AECs. Interestingly, inhibition of LC3B either by ROS inhibitor N-acetyl-l-cysteine (NAC) or adenovirus-mediated LC3B shRNA could partly restore AECII transdifferentiation under hyperoxia condition. In summary, the current study reveals a novel role of activated LC3B induced by hyperoxia in AECII transdifferentiation.


Assuntos
Células Epiteliais Alveolares/citologia , Células Epiteliais Alveolares/efeitos dos fármacos , Transdiferenciação Celular/efeitos dos fármacos , Proteínas Associadas aos Microtúbulos/metabolismo , Oxigênio/farmacologia , Células Epiteliais Alveolares/metabolismo , Animais , Animais Recém-Nascidos , Autofagia/efeitos dos fármacos , Relação Dose-Resposta a Droga , Feminino , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Masculino , Proteínas Associadas aos Microtúbulos/antagonistas & inibidores , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo
15.
Iran J Public Health ; 45(2): 146-57, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27114979

RESUMO

BACKGROUND: Depression and smoking decrease health status in Chronic Obstructive Pulmonary Disease (COPD), but the combined effect of the two factors is unknown. This study aimed to assess the interactive effects of depression and smoking on the severity of patients with COPD. METHODS: A prospective cohort study including 2,268 patients with COPD was conducted in seven rural communities from May 2008 to May 2012. The relationships between the BODE (body mass index, airflow obstruction, dyspnea, and exercise capacity) index and depression and nicotine addiction were assessed. The product of depression and smoking was added to the logistic regression model to evaluate the multiplicative interaction and relative excess risk of interaction (RERI). The Attributable Proportion (AP) of interaction and the synergy index (S) was applied to evaluate the additive interaction of two factors. RESULTS: The severity of COPD in patients with depressive symptoms who never smoked was increased 1.74-fold and in smoking patients it increased by 6.08-fold. Highly addicted smokers with depressive symptoms had a nearly 40-fold increase in severity (all P<0.001). The increased values of the BODE index, HADS-D and Fagerström score also correlated positively (P<0.001). The co-presence of depressive symptoms and smoking significantly increased the BODE index by 11.99-fold with significant biological interactions, relative excess risk of interaction (RERI) was 12.12, the biological interactions were increased with increasing nicotine addiction. CONCLUSIONS: Patients with depressive symptoms who also smoke have an increased risk of severity from COPD interaction effects. The risk for severity of COPD and depressive symptoms increased with increasing the severity of nicotine addiction.

16.
Mol Cell Biochem ; 408(1-2): 181-9, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26122393

RESUMO

Surfactant protein C (SP-C) deficiency is a risk factor for hyperoxia-induced bronchopulmonary dysplasia in newborn infants. However, the role of SP-C deficiency in the process is unclear. Here, using neonatal rat BPD model and MLE-12, mouse alveolar epithelial type II cell, we examined the changes of SP-C levels during hyperoxia. Immunohistochemistry, immunofluorescence, and ELISA analysis showed SP-C accumulation in alveolar epithelial type II cells. Electron microscopy further demonstrated the accumulation of lamellar bodies and the co-localization of lamellar bodies with autophagosomes in the cytoplasm of alveolar epithelial type II cells. The inhibition of autophagy with 3-Methyladenine and knockdown of Atg7 abolished hyperoxia-induced SP-C accumulation in the cytoplasm. Furthermore, inhibition of JNK signaling with SP600125 suppressed hyperoxia-induced Atg7 expression and SP-C accumulation. These findings suggest that hyperoxia triggers autophagy via JNK signaling-mediated Atg7 expression, which promotes the accumulation of SP-C within alveolar epithelial type II cells. Our data provide a potential approach for hyperoxic lung injury therapy by targeted pharmacological inhibition of autophagic pathway.


Assuntos
Células Epiteliais/citologia , Sistema de Sinalização das MAP Quinases , Alvéolos Pulmonares/metabolismo , Proteína C Associada a Surfactante Pulmonar/metabolismo , Enzimas Ativadoras de Ubiquitina/metabolismo , Animais , Animais Recém-Nascidos , Autofagia , Hipóxia Celular , Linhagem Celular , Camundongos , Alvéolos Pulmonares/citologia , Ratos
17.
Cell Biochem Funct ; 33(1): 44-9, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25515701

RESUMO

Prolonged exposure to hyperoxia leads to acute lung injury. Alveolar type II cells are main target of hyperoxia-induced lung injury. However, the cellular and molecular mechanisms remain unknown. Here, we aimed to investigate the role of placental growth factor (PLGF) in hyperoxia-induced lung injury. Using experimental hyperoxia-induced lung injury model of neonatal rat and mouse lung epithelial type II cells (MLE-12), we examined the levels of PLGF in bronchoalveolar lavage fluid and in the supernatants of MLE-12 cells. Our results revealed that exogenous PLGF induced hyperoxia-induced lung injury. Furthermore, PLGF triggered a shift of vinculin from insoluble to soluble cell fraction, similar to the observation under hyperoxia stimulation. Moreover, we observed significantly reduced phosphorylation of focal adhesion kinase and increased permeability in MLE-12 cells treated with PLGF. These results suggest that PLGF triggers focal adhesion disassembly in alveolar type II cells via inhibiting the activation of focal adhesion kinase. Our findings reveal a novel role of PLGF in hyperoxia-induced lung injury and provide a potential target for the management of hyperoxia-induced acute lung injury.


Assuntos
Lesão Pulmonar Aguda/metabolismo , Modelos Animais de Doenças , Hiperóxia/metabolismo , Pulmão/metabolismo , Proteínas da Gravidez/metabolismo , Animais , Animais Recém-Nascidos , Líquido da Lavagem Broncoalveolar , Células Epiteliais , Quinase 1 de Adesão Focal/metabolismo , Adesões Focais/metabolismo , Pulmão/patologia , Camundongos , Permeabilidade , Fator de Crescimento Placentário , Ratos
18.
Respir Care ; 60(1): 102-12, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25371402

RESUMO

BACKGROUND: A number of effective strategies have been developed to improve the quality of life in patients with COPD. However, few have been implemented in patients with COPD at all stages in a community setting. This study evaluated the efficacy of a complex COPD health management intervention in rural communities in China. METHODS: A randomized controlled trial including 8,217 subjects with COPD was implemented from May 2008 to May 2012 in 14 community health-care centers. The control group of subjects received usual care, whereas the management group of subjects participated in a health management program that included assessing the subjects' health status, giving regular health lectures, smoking cessation counseling, encouraging regular exercise, providing rehabilitative training and psychological counseling, and regular follow-up. As a primary outcome, we examined the changes in the BODE index. Secondary outcomes included depression and anxiety rates, current smoking rate, awareness of COPD, mortality, risk factors, respiratory medication use, hospital admissions, and emergency department visits. RESULTS: After 4 y, the mean cumulative value of the BODE index increased by 0.7 per subject in the control group and decreased by 0.4 per subject in the health management group (difference of 1.1, 95% CI 0.2-2.0, P < .001). Health management reduced anxiety symptoms by 4.8%, depression symptoms by 6.6%, current smoking by 14.3%, mortality due to all causes by 9.0%, hospitalizations due to all causes by 16.1%, and emergency department visits due to all causes by 18.1% (P < .05 for all). However, it increased immunomodulator use by 58.9%, respiratory medication use by 14.1-21.9%, and awareness scores for COPD by 57.2% (P < .05 for all). CONCLUSIONS: The health management program is an effective community-based strategy for the prevention and management of COPD in China, increasing awareness and knowledge among patients and practitioners and improving management within the limitations of access to pharmacotherapy.


Assuntos
Serviços de Saúde Comunitária , Aconselhamento Diretivo , Educação de Pacientes como Assunto , Atenção Primária à Saúde/métodos , Doença Pulmonar Obstrutiva Crônica/terapia , Serviços de Saúde Rural , Idoso , Ansiedade/etiologia , Índice de Massa Corporal , Exercícios Respiratórios , China/epidemiologia , Depressão/etiologia , Dieta , Dispneia/etiologia , Serviço Hospitalar de Emergência/estatística & dados numéricos , Exercício Físico , Tolerância ao Exercício , Feminino , Volume Expiratório Forçado , Conhecimentos, Atitudes e Prática em Saúde , Hospitalização/estatística & dados numéricos , Humanos , Fatores Imunológicos/uso terapêutico , Masculino , Pessoa de Meia-Idade , Cooperação do Paciente , Padrões de Prática Médica , Doença Pulmonar Obstrutiva Crônica/mortalidade , Doença Pulmonar Obstrutiva Crônica/fisiopatologia , Doença Pulmonar Obstrutiva Crônica/psicologia , Índice de Gravidade de Doença , Abandono do Hábito de Fumar
19.
Eur J Pharmacol ; 727: 115-24, 2014 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-24486702

RESUMO

Sirtuins maintain energy balance. Particularly, sirtuin 1 (SIRT1) activation mimics calorie restriction and nutrient utilization. However, no medications are available for the up-regulation of SIRT1. Methylene blue (MB) had been in clinical trials for the treatment of neurological diseases. This study investigated the effect of MB on sirtuin expression in association with the treatment of steatosis and steatohepatitis, and explored the underlying basis. The effects of MB on mitochondrial function, molecular markers, pharmacokinetics, and histopathology were assessed using hepatocyte and/or mouse models. Immunoblotting, PCR and reporter assays were done for molecular experiments. After oral administration, MB was well distributed in the liver. MB treatment increased NAD(+)/NADH ratio in hepatocytes. Of the major forms, MB treatment up-regulated SIRT1, and thereby decreased PGC-1α acetylation. Consistently, hepatic mitochondrial DNA contents and oxygen consumption rates were enhanced. MB treatment also notably activated AMPK, CPT-1 and PPARα: the AMPK activation relied on SIRT1. Activation of LXRα and the induction of SREBP-1c and its target genes by T0901317 were diminished by MB. In addition, MB treatment antagonized the ability of palmitate to acetylate PGC-1α, and increase SERBP-1c, FAS, and ACC levels. In mice fed on a high-fat diet for 8 weeks, MB treatment inhibited excessive hepatic fat accumulation and steatohepatitis. The ability of MB to activate SIRT1 promotes mitochondrial biogenesis and oxygen consumption and activates AMPK, contributing to anti-lipogenesis in the liver. Our results provide new information on the potential use of MB for the treatment of steatosis and steatohepatitis.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Reposicionamento de Medicamentos , Ativadores de Enzimas/farmacologia , Fígado Gorduroso/prevenção & controle , Fígado/efeitos dos fármacos , Azul de Metileno/farmacologia , Sirtuína 1/metabolismo , Administração Oral , Animais , DNA Mitocondrial/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Metabolismo Energético/efeitos dos fármacos , Ativação Enzimática , Ativadores de Enzimas/farmacocinética , Fígado Gorduroso/enzimologia , Fígado Gorduroso/patologia , Células Hep G2 , Humanos , Metabolismo dos Lipídeos/efeitos dos fármacos , Fígado/enzimologia , Fígado/patologia , Masculino , Azul de Metileno/farmacocinética , Camundongos Endogâmicos C57BL , Mitocôndrias Hepáticas/efeitos dos fármacos , Mitocôndrias Hepáticas/enzimologia , Renovação Mitocondrial/efeitos dos fármacos , NAD/metabolismo , Interferência de RNA , Ratos , Sirtuína 1/genética , Fatores de Tempo , Transfecção
20.
Respir Care ; 59(1): 54-61, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23737545

RESUMO

BACKGROUND: Smoking, depression, and anxiety increase the risk of death in patients with COPD, but the combined effect of these factors is unknown. We assessed the interactive effects of smoking, depression, and anxiety on mortality in patients with COPD. METHODS: We collected and analyzed data from 7,787 subjects with COPD, in 14 rural communities, from May 2008 to May 2012, and used logistic regression to evaluate the interactions and relative excess risk due to interaction (RERI). We applied the attributable proportion of interaction and the synergy index to evaluate the additive interactions of the factors. RESULTS: In our COPD subjects the interaction of current smoking and depression symptoms increased the death risk by 3.8-fold (odds ratio 3.78, 95% CI 2.51-5.05), with significant biological interactions (RERI 1.74, 95% CI 0.51-2.99, attributable proportion 0.48, 95% CI 0.13-0.85, synergy index 2.98, 95% CI 1.44-4.56). The biological interactions increased with increasing years or pack-years of smoking: for subjects with ≥ 30 years of smoking: RERI 1.80, 95% CI 1.05-2.75, attributable proportion 0.48, 95% CI 0.15-0.82, synergy index 2.85, 95% CI 1.75-3.96; for subjects with ≥ 40 pack-years of smoking: RERI 3.11, 95% CI 1.54-4.71, attributable proportion 0.60, 95% CI 0.31-0.91, synergy index 4.00, 95% CI 2.84-5.26. Similarly, the combined effect of current smoking and anxiety symptoms increased the death risk by 4.3-fold (odds ratio 4.27, 95% CI 95% CI 2.96-5.59), with significant biological interactions (RERI 1.51, 95% CI 0.31-2.74, attributable proportion 0.46, 95% CI 0.11-0.87, synergy index 2.89, 95% CI 1.31-4.51). The biological interactions also increased with increasing years or pack-years of smoking: for subjects with ≥ 30 years of smoking: RERI 1.41, 95% CI 0.45-2.43, attributable proportion 0.45, 95% CI 0.12-0.81, synergy index 2.88, 95% CI 1.24-5.98; for subjects with ≥ 40 pack-years of smoking: RERI 3.15, 95% CI 2.07-4.61, attributable proportion 0.55, 95% CI 0.21-0.94, synergy index 3.00, 95% CI 1.45-4.75. CONCLUSIONS: Smoking, depression, and anxiety are associated with higher risk of death in patients with COPD. The risk of death, depression, and anxiety increases with increasing duration of smoking (years) and cigarette pack-years. Chinese Clinical Trials Registration ChiCTR-TRC-12001958.


Assuntos
Ansiedade/epidemiologia , Depressão/epidemiologia , Doença Pulmonar Obstrutiva Crônica/mortalidade , Fumar/epidemiologia , Idoso , Idoso de 80 Anos ou mais , China/epidemiologia , Comorbidade , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Escalas de Graduação Psiquiátrica , Fatores de Risco , Índice de Gravidade de Doença
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA