Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
J Transl Med ; 21(1): 24, 2023 01 13.
Article in English | MEDLINE | ID: mdl-36635651

ABSTRACT

BACKGROUND: Idiopathic pulmonary fibrosis is a chronic progressive, lethal disease in which ectopic lung fibroblast (LF) activation plays a vital part. We have previously shown that alamandine (ALA) exerts anti-fibrosis effects via the MAS-related G-protein coupled receptor D (MrgD). Here, we further investigate how it moderates transforming growth factor ß1 (TGF-ß1)-induced LF activation by regulating glucose metabolism and mitochondria autophagy (mitophagy). METHODS: In vitro, we examined glycolysis-related protein hexokinase 2 (HK2), 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3), and lactic acid in cells treated with TGF-ß1. The oxygen consumption rate and the extracellular acidification rate were detected using Seahorse assays. Then, mitophagy was evaluated using transmission electron microscopy, mt-Keima, and the co-localization of Parkin and COX IV with LC3 and LAMP1, respectively. The autophagic degradation of HK2 and PFKFB3 was detected by 3MA and bafilomycin A1 and assessed by their co-localization with LC3 and LAMP1, respectively. The effects of ALA on LF activation markers collagen I and α-SMA were detected. The effects of ALA on glucose metabolism, mitophagy, and the activation of LF were also investigated in vivo. RESULTS: We found that the ALA/MrgD axis improved TGF-ß1-mediated LF activation by repressing glycolysis by downregulating HK2 and PFKFB3 expression. Lactic acid sustained positive feedback between glycolysis and LF activation by maintaining the expression of HK2 and PFKFB3. We also showed that glycolysis enhancement resulted from blocking the autophagic degradation of HK2 and PFKFB3 while upregulated mRNA levels by TGF-ß1, while all of those improved by ALA adding. Importantly, we determined that moderation of Parkin/LC3-mediated mitophagy by TGF-ß1 also promotes glycolysis but is reversed by ALA. Furthermore, we proved that ALA counteracts the effects of bleomycin on HK2, PFKFB3, LC3, Parkin, and LF activation in vivo. CONCLUSION: In this study, we show that the ALA/MrgD axis prevents TGF-ß1-mediated fibroblast activation via regulation of aerobic glycolysis and mitophagy.


Subject(s)
Mitophagy , Transforming Growth Factor beta1 , Transforming Growth Factor beta1/metabolism , Fibroblasts/metabolism , Glycolysis , Ubiquitin-Protein Ligases/metabolism , Glucose/metabolism
2.
J Immunol Res ; 2022: 2291877, 2022.
Article in English | MEDLINE | ID: mdl-36033388

ABSTRACT

Endoplasmic reticulum stress (ER stress) contributes to the development of pulmonary fibrosis, especially in type II alveolar epithelial cells (AECs) apoptosis. ER stress also promotes NLRP3 inflammasome activation which is inhibited by upregulation of cAMP/PKA pathway. However, it is confused whether ER stress-induced NLRP3 inflammasome activation and pyroptosis in type II alveolar epithelial cells which exacerbates pulmonary fibrosis via a mechanism that is suppressed by cAMP/PKA pathway. In our research, we explored that potential links among NLRP3 inflammasome, ER stress, and cAMP/PKA pathway in type II AECs to explain the new mechanisms of pulmonary fibrosis. We found that in vivo, ER stress, NLRP3 inflammasome, and PKA upregulated in the alveolar epithelial area in animal models of pulmonary fibrosis. In addition, immunofluorescence staining further confirmed that ER stress, NLRP3 inflammasome, and cAMP/PKA had potential links on type II AECs in BLM group. In vitro, ER stress stimulated NLRP3 inflammasome activation, promoted pyroptosis, and also upregulated cAMP/PKA pathway. Upregulation of cAMP/PKA pathway inhibited ER stress-induced pyroptosis of A549 cells and vice versa. These results initially supported conclusion that ER stress may stimulate NLRP3 inflammasome activation and pyroptosis in type II AECs, which exacerbated pulmonary fibrosis, and cAMP/PKA pathway may act as a feedback regulator.


Subject(s)
Pulmonary Fibrosis , Pyroptosis , Alveolar Epithelial Cells , Animals , Endoplasmic Reticulum Stress , Feedback , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein
3.
J Inflamm Res ; 15: 2121-2139, 2022.
Article in English | MEDLINE | ID: mdl-35386222

ABSTRACT

Purpose: Acute lung injury (ALI) is a life-threatening condition with limited therapeutic options. Macrophage inflammation plays a key role in the development of ALI. Abnormal glycolysis of macrophages contributes to the inflammatory response. However, the role of macrophage glycolysis in ALI still requires investigation. Apelin-13 has been shown to protect against ALI, whereas the underlying mechanisms remain unclear. In this study, we explored the effect of apelin-13 on lipopolysaccharide (LPS)-induced inflammation and ALI via regulation of glycolysis by modulating redox homeostasis in macrophages. Methods: Serums from 34 patients with sepsis and 13 healthy volunteers were analyzed. In vivo, the protective effect of apelin-13 against LPS-induced ALI was evaluated using a mouse model of LPS-induced ALI. In vitro, mouse bone marrow macrophages (BMDMs) were pretreated with the antioxidant, NADPH oxidase (NOX) 4 (NOX4) small-interfering RNA (siRNA), the 6-phosphofructo-2 -kinase/fructose- 2,6-biphosphatase 3 (PFKFB3) siRNA, or the PFKFB3 overexpression plasmid before exposure to LPS. Results: Serum apelin-13 levels were significantly elevated in patients with sepsis and sepsis-associated acute respiratory distress syndrome (ARDS) (P<0.0001). In vivo, apelin-13 suppressed LPS-induced ALI and inflammatory cytokine production (P<0.05). Furthermore, apelin-13 reduced hydrogen peroxide (H2O2) content, NOX4 protein levels, and glycolysis. In vitro, LPS stimulation elevated NOX4 protein levels and reactive oxygen species (ROS) production (P<0.05). These changes resulted in the accumulation of glycolysis in BMDMs. Treatment with antioxidant or NOX4 siRNA inhibited LPS-induced glycolysis and inflammatory cytokine production (P<0.05). Moreover, in vitro experiments revealed that PFKFB3 regulates the release of pro-inflammatory cytokines by modulating glycolysis. In contrast, the action of apelin-13 opposed the effects of LPS. Conclusion: In conclusion, apelin-13 protects against LPS-induced inflammatory responses and ALI by regulating PFKFB3-driven glycolysis induced by NOX4-dependent ROS.

4.
Free Radic Biol Med ; 166: 116-127, 2021 04.
Article in English | MEDLINE | ID: mdl-33609723

ABSTRACT

AIMS: The senescence of alveolar epithelial type 2 (AT2) cells is implicated in the pathogenesis of idiopathic pulmonary fibrosis (IPF). Cigarette smoke (CS) is a strong risk factor for IPF and it is also a pro-senescent factor. Here we aimed to investigate whether and how CS induces AT2 cells senescence via a SIRT1/autophagy dependent pathway. Our results showed that CS extract (CSE) reduced autophagy and mitophagy and increased mitochondrial reactive oxygen species (mitoROS) in MLE-12 cells, an AT2 cell line. The autophagy inducer rapamycin (RAPA) and the mitochondria-targeted antioxidant mitoquinone (mitoQ) inhibited CSE-related senescence and decreased mitoROS. Next, we found that CSE promoted DNA damage, downregulated the nicotinamide adenine dinucleotide (NAD+)/nicotinamide adenine dinucleotide (NADH) ratio and suppressed SIRT1 activity. Activating SIRT1 with its activator SRT1720 attenuated senescence through an autophagy-dependent pathway. The NAD+ precursor nicotinamide mononucleotide and the poly ADP-ribose polymerase (PARP1) inhibitor olaparib also exerted anti-senescent effects by activating SIRT1. Moreover, the results showed that mitoQ and RAPA, in turn, elevated SIRT1 activity by inhibiting DNA damage. Consistent with these results, SRT1720 and mitoQ mitigated CS-induced AT2 cells senescence and lung fibrosis in vivo. Moreover, autophagy in AT2 cells was rescued by SRT1720. Taken together, our results suggested that CS-induced senescence of AT2 cells was due to decreased autophagy mediated by SIRT1 inactivation, which was attributed to competitive consumption of NAD+ caused by DNA damage-induced PARP1 activation. The reduction in autophagy, in turn, decreased SIRT1 activity by promoting mitochondrial oxidative stress-related DNA damage, thereby establishing a positive feedback loop between SIRT1 and autophagy in CS-induced AT2 cells senescence. Consequently, CS-inactivated SIRT1 promoted autophagy-dependent senescence of AT2 cells to induce pulmonary fibrosis.


Subject(s)
Pulmonary Fibrosis , Sirtuin 1 , Autophagy , Cellular Senescence , Epithelial Cells , Humans , Sirtuin 1/genetics , Smoke , Smoking
5.
Can J Physiol Pharmacol ; 99(9): 885-893, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33517849

ABSTRACT

Alamandine (ALA) and its receptor MrgD were recently identified as components of the renin-angiotensin system, which confer protection against cardio-fibrosis and renal-fibrosis; however, the effects of ALA on pulmonary fibrosis are unknown. This study was designed to serve two goals: (i) to evaluate the ALA/MrgD axis ability in the prevention of angiotensin II (Ang II) - induced pulmonary fibrosis in fibroblasts, and (ii) to determine the effect of ALA in bleomycin (BLM) - treated C57B/6 mice. In vivo experiments revealed that the treatment of C57B/6 mice with ALA prevented BLM-induced fibrosis, and these findings were similar to those reported for pirfenidone. The antifibrosis actions of ALA were mediated via alleviation of oxidative injury and autophagy induction. In addition, in vitro studies revealed that ALA treatment attenuated Ang II-induced α-collagen I, CTGF, and α-SMA production in fibroblast which was blocked by D-Pro7-Ang-(1-7), a MrgD antagonist. This led to alleviation of oxidative injury and induction of autophagy similar to that reported for rapamycin. This study demonstrated that ALA via MrgD receptor reduced pulmonary fibrosis through attenuation of oxidative injury and induction of autophagy.


Subject(s)
Autophagy/physiology , NADPH Oxidase 4/physiology , Oligopeptides/therapeutic use , Pulmonary Fibrosis/drug therapy , Receptors, G-Protein-Coupled/physiology , Angiotensin II/pharmacology , Animals , Bleomycin/toxicity , Cells, Cultured , Collagen/biosynthesis , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Oligopeptides/pharmacology , Pulmonary Fibrosis/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...