Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Environ Int ; 177: 108027, 2023 07.
Article in English | MEDLINE | ID: mdl-37321070

ABSTRACT

Over 400,000 people are estimated to have been exposed to World Trade Center particulate matter (WTCPM) since the attack on the Twin Towers in Lower Manhattan on September 11, 2001. Epidemiological studies have found that exposure to dust may cause respiratory ailments and cardiovascular diseases. However, limited studies have performed a systematic analysis of transcriptomic data to elucidate the biological responses to WTCPM exposure and the therapeutic options. Here, we developed an in vivo mouse exposure model of WTCPM and administered two drugs (i.e., rosoxacin and dexamethasone) to generate transcriptomic data from lung samples. WTCPM exposure increased the inflammation index, and this index was significantly reduced by both drugs. We analyzed the transcriptomics derived omics data using a hierarchical systems biology model (HiSBiM) with four levels, including system, subsystem, pathway, and gene analyses. Based on the selected differentially expressed genes (DEGs) from each group, WTCPM and the two drugs commonly affected the inflammatory responses, consistent with the inflammation index. Among these DEGs, the expression of 31 genes was affected by WTCPM exposure and consistently reversed by the two drugs, and these genes included Psme2, Cldn18, and Prkcd, which are involved in immune- and endocrine-related subsystems and pathways such as thyroid hormone synthesis, antigen processing and presentation, and leukocyte transendothelial migration. Furthermore, the two drugs reduced the inflammatory effects of WTCPM through distinct pathways, e.g., vascular-associated signaling by rosoxacin, whereas mTOR-dependent inflammatory signaling was found to be regulated by dexamethasone. To the best of our knowledge, this study constitutes the first investigation of transcriptomics data of WTCPM and an exploration of potential therapies. We believe that these findings provide strategies for the development of promising optional interventions and therapies for airborne particle exposure.


Subject(s)
Particulate Matter , Pneumonia , Mice , Animals , Particulate Matter/toxicity , Transcriptome , Dust/analysis , Inflammation , Dexamethasone/toxicity , Proteasome Endopeptidase Complex
3.
J Mol Med (Berl) ; 94(7): 809-19, 2016 07.
Article in English | MEDLINE | ID: mdl-26864306

ABSTRACT

UNLABELLED: Understanding the underlying molecular mechanisms in burn wound progression is crucial to providing appropriate diagnoses and designing therapeutic regimens for burn patients. When inflammation becomes unregulated, recurrent, or excessive, it interferes with burn wound healing. Autophagy, which is a homeostatic and catabolic degradation process, was found to protect against ischemic injury, inflammatory diseases, and apoptosis in some cases. In the present study, we investigated whether far-infrared (FIR) could ameliorate burn wound progression and promote wound healing both in vitro and in a rat model of deep second-degree burn. We found that FIR induced autophagy in differentiated THP-1 cells (human monocytic cells differentiated to macrophages). Furthermore, FIR inhibited both the NLRP3 inflammasome and the production of IL-1ß in lipopolysaccharide-activated THP-1 macrophages. In addition, FIR induced the ubiquitination of ASC, which is the adaptor protein of the inflammasome, by increasing tumor necrosis factor receptor-associated factor 6 (TRAF6), which is a ubiquitin E3 ligase. Furthermore, the exposure to FIR then promoted the delivery of inflammasome to autophagosomes for degradation. In a rat burn model, FIR ameliorated burn-induced epidermal thickening, inflammatory cell infiltration, and loss of distinct collagen fibers. Moreover, FIR enhanced autophagy and suppressed the activity of the NLRP3 inflammasome in the rat skin tissue of the burn model. Based on these results, we suggest that FIR-regulated autophagy and inflammasomes will be important for the discovery of novel therapeutics to promote the healing of burn wounds. KEY MESSAGES: Far-infrared (FIR) induced autophagy in THP-1 macrophages. FIR suppressed the NLRP3 inflammasome through the activation of autophagy. FIR induced the ubiquitination of ASC by increasing TRAF6. FIR ameliorated burn wound progression and promoted wound healing in a rat burn model.


Subject(s)
Autophagy/radiation effects , Burns/therapy , Inflammasomes/radiation effects , Infrared Rays/therapeutic use , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Wound Healing/radiation effects , Animals , Autophagy/drug effects , Autophagy/genetics , Burns/genetics , Burns/immunology , Burns/pathology , CARD Signaling Adaptor Proteins/genetics , CARD Signaling Adaptor Proteins/metabolism , Cell Line , Disease Models, Animal , Gene Expression Regulation , Humans , Inflammasomes/drug effects , Inflammasomes/genetics , Inflammasomes/immunology , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Intracellular Signaling Peptides and Proteins , Lipopolysaccharides/pharmacology , Macrophage Activation/drug effects , Macrophages/cytology , Macrophages/drug effects , Macrophages/immunology , Macrophages/radiation effects , Male , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Rats , Rats, Wistar , TNF Receptor-Associated Factor 6/agonists , TNF Receptor-Associated Factor 6/genetics , TNF Receptor-Associated Factor 6/immunology , Ubiquitination/radiation effects , Wound Healing/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...