1
|
Hu X, Zhang N, Zhong Y, Liu T, Zhu X. Mechanisms of Apoptosis and Pulmonary Fibrosis Resulting From Sulfur Mustard-Induced Acute Pulmonary Injury in Rats. Int J Toxicol 2025:10915818251315907. [PMID: 39888856 DOI: 10.1177/10915818251315907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2025]
Abstract
Sulfur mustard (SM) is a highly toxic bifunctional alkylating agent that inflicts severe damage on the respiratory tract. Although numerous studies have examined the mechanisms underlying SM-induced pulmonary injury, the exact pathways involved remain unclear. This study aims to investigate an acute pulmonary injury model, with SM administered as a single intraperitoneal injection (8 mg/kg) or single intratracheal instillation (2 mg/kg) at equal toxicity doses (1LD50). The results revealed that epithelial cells in the alveolar septa of the intraperitoneal SM group exhibited a significantly higher expression of apoptotic markers, including pro-apoptotic protein Bax, caspase-3, and caspase-9 proteins, than those in the tracheal SM group. Conversely, the expression of the anti-apoptotic protein Bcl-2 was significantly lower in the intraperitoneal SM group than in the tracheal SM group, as confirmed by TUNEL staining and immunohistochemical staining. The intraperitoneal SM group exhibited markedly higher expression of fibrosis-related proteins, including MMP-2, MMP-9, TIMP-1, TIMP-2, collagen type I, collagen type III, TGF-β1, and Smad7, than the tracheal SM group. These markers, detected through immunohistochemical immunolabeling, indicate a more significant fibrotic response in the intraperitoneal group. In summary, this study demonstrates that intraperitoneal exposure to SM results in increased apoptosis, elevated expression of pro-apoptotic proteins, and fibrosis-related proteins in the alveolar epithelial cells compared with intratracheal exposure, even at equivalent toxicity levels. Our findings highlight the suitability of the intraperitoneal route for further investigation and identify apoptotic and fibrosis-related proteins as potential targets for intervention in SM-induced pulmonary injury.
Collapse
Affiliation(s)
- Xiaoxuan Hu
- Weifang No. 2 People's Hospital, Weifang Respiratory Disease Hospital, Weifang, China
- State Key Laboratory of Antitoxic Drugs and Toxicology, Institute of Toxicology and Pharmacology, Academy of Military Medical Sciences, Beijing, China
| | - Na Zhang
- Department of Respiration, The 80th Group Army Hospital of People's Liberation Army, Weifang, China
| | - Yuxu Zhong
- State Key Laboratory of Antitoxic Drugs and Toxicology, Institute of Toxicology and Pharmacology, Academy of Military Medical Sciences, Beijing, China
| | - Tao Liu
- Department of Respiration, The 80th Group Army Hospital of People's Liberation Army, Weifang, China
| | - Xiaoji Zhu
- Department of Respiration, Jiaozhou Branch of Shanghai East Hospital, Tongji University, Qingdao, China
| |
Collapse
|
2
|
Jatczak-Pawlik I, Jurewicz A, Domowicz M, Ewiak-Paszyńska A, Stasiołek M. CHI3L1 in Multiple Sclerosis-From Bench to Clinic. Cells 2024; 13:2086. [PMID: 39768177 PMCID: PMC11674340 DOI: 10.3390/cells13242086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 12/05/2024] [Accepted: 12/14/2024] [Indexed: 01/11/2025] Open
Abstract
Multiple sclerosis (MS) is a chronic demyelinating disease of the central nervous system (CNS) with a complex and not fully understood etiopathological background involving inflammatory and neurodegenerative processes. CHI3L1 has been implicated in pathological conditions such as inflammation, injury, and neurodegeneration, and is likely to play a role in the physiological development of the CNS. CHI3L1 is primarily produced by CNS macrophages, microglia, and activated astrocytes. The CHI3L1 expression pattern in MS lesions might support the important role of astrocytes in modulating inflammatory processes in this disease. The potential applications of CHI3L1 as a biomarker in MS are multifactorial. The measurement of CHI3L1 in body fluids might find its role in the early diagnosis of MS. In further stages, the monitoring of CHI3L1 levels might provide information on disease severity and progression, enabling a better adjustment of therapeutic strategies. Importantly, CHI3L1 might potentially serve as a marker of ongoing glial activation, reflecting the dynamic response of the CNS cells to the inflammatory processes in MS. Although preliminary findings have been promising, further research is needed to validate the utility of CHI3L1 measurements in the diagnosis and prediction of the progression of MS. Additionally, comparisons with other biomarkers might be useful in clinical practice.
Collapse
Affiliation(s)
| | | | | | | | - Mariusz Stasiołek
- Department of Neurology, Medical University of Lodz, Kosciuszki Street 4, 90-419 Lodz, Poland; (I.J.-P.); (A.J.); (M.D.); (A.E.-P.)
| |
Collapse
|
3
|
Delrue C, Speeckaert R, Delanghe JR, Speeckaert MM. Breath of fresh air: Investigating the link between AGEs, sRAGE, and lung diseases. VITAMINS AND HORMONES 2024; 125:311-365. [PMID: 38997169 DOI: 10.1016/bs.vh.2024.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/14/2024]
Abstract
Advanced glycation end products (AGEs) are compounds formed via non-enzymatic reactions between reducing sugars and amino acids or proteins. AGEs can accumulate in various tissues and organs and have been implicated in the development and progression of various diseases, including lung diseases. The receptor of advanced glycation end products (RAGE) is a receptor that can bind to advanced AGEs and induce several cellular processes such as inflammation and oxidative stress. Several studies have shown that both AGEs and RAGE play a role in the pathogenesis of lung diseases, such as chronic obstructive pulmonary disease, asthma, idiopathic pulmonary fibrosis, cystic fibrosis, and acute lung injury. Moreover, the soluble form of the receptor for advanced glycation end products (sRAGE) has demonstrated its ability to function as a decoy receptor, possessing beneficial characteristics such as anti-inflammatory, antioxidant, and anti-fibrotic properties. These qualities make it an encouraging focus for therapeutic intervention in managing pulmonary disorders. This review highlights the current understanding of the roles of AGEs and (s)RAGE in pulmonary diseases and their potential as biomarkers and therapeutic targets for preventing and treating these pathologies.
Collapse
Affiliation(s)
- Charlotte Delrue
- Department of Nephrology, Ghent University Hospital, Ghent, Belgium
| | | | - Joris R Delanghe
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Marijn M Speeckaert
- Department of Nephrology, Ghent University Hospital, Ghent, Belgium; Research Foundation-Flanders (FWO), Brussels, Belgium.
| |
Collapse
|
4
|
Wu W, Wang Z, Zhang H, Zhang X, Tian H. circGRHPR inhibits aberrant epithelial-mesenchymal transformation progression of lung epithelial cells associated with idiopathic pulmonary fibrosis. Cell Biol Toxicol 2024; 40:7. [PMID: 38267743 PMCID: PMC10808371 DOI: 10.1007/s10565-024-09839-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 12/05/2023] [Indexed: 01/26/2024]
Abstract
Air pollution has greatly increased the risk of idiopathic pulmonary fibrosis (IPF). Circular RNAs (circRNAs) have been found to play a significant role in the advancement of IPF, but there is limited evidence of correlation between circRNAs and lung epithelial cells (LECs) in IPF. This research aimed to explore the influence of circRNAs on the regulation of EMT progression in LECs, with the objective of elucidating its mechanism and establishing its association with IPF. Our results suggested that the downregulation of circGRHPR in peripheral blood of clinical cases was associated with the diagnosis of IPF. Meanwhile, we found that circGRHPR was downregulated in transforming growth factor-beta1 (TGF-β1)-induced A549 and Beas-2b cells. It is a valid model to study the abnormal EMT progression of IPF-associated LECs in vitro. The overexpression of circGRHPR inhibited the abnormal EMT progression of TGF-β1-induced LECs. Furthermore, as the sponge of miR-665, circGRHPR released the expression of E3 ubiquitin-protein ligase NEDD4-like (NEDD4L), thus promoting its downstream transforming growth factor beta receptor 2 (TGFBR2) ubiquitination. It is helpful to reduce the response of LECs to TGF-β1 signaling. In summary, circGRHPR/miR-665/NEDD4L axis inhibited the abnormal EMT progression of TGF-β1-induced LECs by promoting TGFBR2 ubiquitination, which provides new ideas and potential targets for the treatment of IPF.
Collapse
Affiliation(s)
- Wensi Wu
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, No. 107, Wenhua West Road, Lixia District, Jinan, 250012, People's Republic of China
| | - Zhi Wang
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, No. 107, Yanjiang West Road, Yuexiu District, Guangzhou, 510120, People's Republic of China
| | - Huiying Zhang
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, No. 107, Wenhua West Road, Lixia District, Jinan, 250012, People's Republic of China
| | - Xiaojun Zhang
- Department of Anesthesiology, Qilu Hospital of Shandong University, No. 107, Wenhua West Road, Lixia District, Jinan, 250012, People's Republic of China.
| | - Hui Tian
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, No. 107, Wenhua West Road, Lixia District, Jinan, 250012, People's Republic of China.
| |
Collapse
|
5
|
Zhao X, Zhang Y, Wu F, Li X, Guo S, Li X. MeCP2-Induced Alternations of Transcript Levels and m6A Methylation in Human Retinal Pigment Epithelium Cells. ACS OMEGA 2023; 8:47964-47973. [PMID: 38144074 PMCID: PMC10734004 DOI: 10.1021/acsomega.3c06610] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 11/05/2023] [Accepted: 11/20/2023] [Indexed: 12/26/2023]
Abstract
MeCP2 is a transcriptional regulator that is involved in epithelial-mesenchymal transition (EMT) and is highly expressed in proliferative vitreoretinopathy. m6A methylation is a critical post-transcriptional regulation in eukaryotic cells. However, the connection between MeCP2 and m6A methylation has not been revealed in retinal pigment epithelium (RPE), and the regulatory role of MeCP2 at the post-transcriptional level in an m6A-dependent manner is rarely investigated. In this study, we used sequencing to reveal differences in transcript levels and m6A abundance of individual genes in RPE cells after treatment with human recombinant protein MeCP2. The biological functions and processes of differential genes were further analyzed by bioinformatics. The results exhibited that after MeCP2 treatment, 65 genes were up-regulated and 43 genes were down-regulated at the transcription level, and 4 peaks were hypermethylated and 9,041 peaks were hypomethylated at the m6A modification level. Enrichment analysis found that differentially expressed genes were associated with organic acid metabolism, melanogenesis, and vascular smooth muscle contraction. In addition, differentially methylated genes were related to cell junction, RNA processing and metabolism, cell activity, actin cytoskeleton, and several signaling pathways associated with EMT. Further conjoint analysis indicated that the transcription and m6A levels of the EGR1, ELOVL2, and SFR1 genes were altered, and EGR1 is an essential transcription factor in the EMT process. The RNA levels and m6A levels of the three genes were verified by qPCR and m6A-IP-qPCR, respectively. Overall, this study preliminarily revealed the differential mapping of MeCP2-induced m6A modifications, which contributes to the study of the epigenetic and EMT mechanism in RPE cells.
Collapse
Affiliation(s)
- Xueru Zhao
- Henan
Eye Hospital, Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People’s Hospital, Zhengzhou 450003, China
- Zhengzhou
University People’s Hospital, 450000 Zhengzhou, China
- People’s
Hospital of Henan University, 450003 Zhengzhou, China
- Eye
Institute, Henan Academy of Innovations
in Medical Science, 450000 Zhengzhou, China
| | - Yongya Zhang
- Henan
Eye Hospital, Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People’s Hospital, Zhengzhou 450003, China
- Zhengzhou
University People’s Hospital, 450000 Zhengzhou, China
| | - Fei Wu
- Henan
Eye Hospital, Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People’s Hospital, Zhengzhou 450003, China
- Zhengzhou
University People’s Hospital, 450000 Zhengzhou, China
| | - Xue Li
- Henan
Eye Hospital, Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People’s Hospital, Zhengzhou 450003, China
- Zhengzhou
University People’s Hospital, 450000 Zhengzhou, China
- People’s
Hospital of Henan University, 450003 Zhengzhou, China
- Eye
Institute, Henan Academy of Innovations
in Medical Science, 450000 Zhengzhou, China
| | - Sibei Guo
- Henan
Eye Hospital, Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People’s Hospital, Zhengzhou 450003, China
- Xinxiang
Medical University Henan Provincial People’s Hospital, 453003 Xinxiang, China
| | - Xiaohua Li
- Henan
Eye Hospital, Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People’s Hospital, Zhengzhou 450003, China
- Zhengzhou
University People’s Hospital, 450000 Zhengzhou, China
- People’s
Hospital of Henan University, 450003 Zhengzhou, China
- Eye
Institute, Henan Academy of Innovations
in Medical Science, 450000 Zhengzhou, China
| |
Collapse
|
6
|
Lei L, Wan G, Geng X, Sun J, Zhang Y, Wang J, Yang C, Pan Z. The total iridoid glycoside extract of Lamiophlomis rotata Kudo induces M2 macrophage polarization to accelerate wound healing by RAS/ p38 MAPK/NF-κB pathway. JOURNAL OF ETHNOPHARMACOLOGY 2023; 307:116193. [PMID: 36746295 DOI: 10.1016/j.jep.2023.116193] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 01/16/2023] [Accepted: 01/17/2023] [Indexed: 06/18/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Lamiophlomis rotata (Benth.) Kudo (L. rotata), a Tibetan medicinal plant, is used to treat "yellow-water diseases", such as skin disease, jaundice and rheumatism. Our previous study showed that the iridoid glycoside extract of L. rotata (IGLR) is the major constituent of skin wound healing. However, the role of IGLR in the biological process of trauma repair and the probable mechanism of the action remain largely unknown. AIM OF THE STUDY To investigate the role of IGLR in the biological process of trauma repair and the probable mechanism of the action. MATERIALS AND METHODS The role of IGLR in wound healing was investigated by overall skin wound in mice with Hematoxylin and Eosin (H&E) and Masson trichrome staining. The anti-inflammatory, angiogenesis-promoting and fibril formation effects of IGLR were visualized in wound skin tissue by immunofluorescence staining, and the proinflammatory factors and growth factors were assayed by real-time polymerase chain reaction (RT-PCR) and enzyme-linked immunosorbent assay (ELISA). Macrophages, dermal fibroblasts, and endothelial cells were cultured to measure the direct/indirect interaction effects of IGLR on the proliferation and migration of cells, and flow cytometry was employed to assess the role of IGLR on macrophage phenotype. Network pharmacology combined with Western blot experiments were conducted to explore possible mechanisms of the actions. RESULTS IGLR increased the expression of CD206 (M2 markers) through the RAS/p38 MAPK/NF-κB signaling pathway during wound injury in vivo and in vitro. IGLR suppressed the inflammatory cytokines iNOS, IL-1β and TNF-α in the early stage of wound healing. During the proliferation step of wound repair, IGLR promoted angiogenesis and fibril formation by increasing the expression of VEGF, CD31, TGF-β and α-SMA in wound tissue, and similar results were verified by RT-PCR and ELISA. In a paracrine mechanism, the extract promoted the proliferation of dermal fibroblasts, and endothelial cells were founded by the conditioned medium (CM). CONCLUSION IGLR induced M2 macrophage polarization in the early stage of wound healing; in turn, IGLR played a key role in the transition from inflammation to cell proliferation during the biological process of wound healing.
Collapse
Affiliation(s)
- Lei Lei
- Chongqing Medical University, Chongqing, China
| | - Guoguo Wan
- Chongqing Medical University, Chongqing, China
| | - Xiaoyu Geng
- Chongqing Medical University, Chongqing, China
| | - Jianguo Sun
- Chongqing Medical University, Chongqing, China
| | - Yi Zhang
- Chengdu University of Traditional Chinese Medicine, Chengdu 611130, China
| | | | | | - Zheng Pan
- Chongqing Medical University, Chongqing, China.
| |
Collapse
|
7
|
Dong H, Zhang Y, Huang Y, Deng H. Pathophysiology of RAGE in inflammatory diseases. Front Immunol 2022; 13:931473. [PMID: 35967420 PMCID: PMC9373849 DOI: 10.3389/fimmu.2022.931473] [Citation(s) in RCA: 95] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 07/06/2022] [Indexed: 12/24/2022] Open
Abstract
The receptor for advanced glycation end products (RAGE) is a non-specific multi-ligand pattern recognition receptor capable of binding to a range of structurally diverse ligands, expressed on a variety of cell types, and performing different functions. The ligand-RAGE axis can trigger a range of signaling events that are associated with diabetes and its complications, neurological disorders, cancer, inflammation and other diseases. Since RAGE is involved in the pathophysiological processes of many diseases, targeting RAGE may be an effective strategy to block RAGE signaling.
Collapse
|
8
|
Kinscherf NA, Pehar M. Role and Therapeutic Potential of RAGE Signaling in Neurodegeneration. Curr Drug Targets 2022; 23:1191-1209. [PMID: 35702767 PMCID: PMC9589927 DOI: 10.2174/1389450123666220610171005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/29/2022] [Accepted: 04/29/2022] [Indexed: 01/03/2023]
Abstract
Activation of the receptor for advanced glycation end products (RAGE) has been shown to play an active role in the development of multiple neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, and Amyotrophic Lateral Sclerosis. Although originally identified as a receptor for advanced glycation end products, RAGE is a pattern recognition receptor able to bind multiple ligands. The final outcome of RAGE signaling is defined in a context and cell type specific manner and can exert both neurotoxic and neuroprotective functions. Contributing to the complexity of the RAGE signaling network, different RAGE isoforms with distinctive signaling capabilities have been described. Moreover, multiple RAGE ligands bind other receptors and RAGE antagonism can significantly affect their signaling. Here, we discuss the outcome of celltype specific RAGE signaling in neurodegenerative pathologies. In addition, we will review the different approaches that have been developed to target RAGE signaling and their therapeutic potential. A clear understanding of the outcome of RAGE signaling in a cell type- and disease-specific manner would contribute to advancing the development of new therapies targeting RAGE. The ability to counteract RAGE neurotoxic signaling while preserving its neuroprotective effects would be critical for the success of novel therapies targeting RAGE signaling.
Collapse
Affiliation(s)
- Noah Alexander Kinscherf
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Mariana Pehar
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.,Geriatric Research Education Clinical Center, Veterans Affairs Medical Center, Madison, WI, USA
| |
Collapse
|
9
|
Zhang Q, Wang Y, Tian C, Yu J, Li Y, Yang J. Clinical characteristics and genetic analysis of a Chinese pedigree of type 2 diabetes complicated with interstitial lung disease. Front Endocrinol (Lausanne) 2022; 13:1050200. [PMID: 36733806 PMCID: PMC9887333 DOI: 10.3389/fendo.2022.1050200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 12/28/2022] [Indexed: 01/18/2023] Open
Abstract
PURPOSE Diabetes mellitus is a systemic metabolic disorder which may target the lungs and lead to interstitial lung disease. The clinical characteristics and mechanisms of type 2 diabetes mellitus (T2DM) complicated with interstitial lung disease (ILD) have been studied. However, little work has been done to assess genetic contributions to the development of T2DM complicated with ILD. METHOD A pedigree of T2DM complicated with ILD was investigated, and the whole genome re-sequencing was performed to identify the genetic variations in the pedigree. According to the literature, the most valuable genetic contributors to the pathogenesis of T2DM complicated with ILD were screened out, and the related cellular functional experiments were also performed. RESULTS A large number of SNPs, InDels, SVs and CNVs were identified in eight subjects including two diabetic patients with ILD, two diabetic patients without ILD, and four healthy subjects from the pedigree. After data analysis according to the literature, MUC5B SNP rs2943512 (A > C) was considered to be an important potentially pathogenic gene mutation associated with the pathogenesis of ILD in T2DM patients. In vitro experiments showed that the expression of MUC5B in BEAS-2B cells was significantly up-regulated by high glucose stimulation, accompanied by the activation of ERK1/2 and the increase of IL-1β and IL-6. When silencing MUC5B by RNA interference, the levels of p-ERK1/2 as well as IL-1β and IL-6 in BEAS-2B cells were all significantly decreased. CONCLUSION The identification of these genetic variants in the pedigree enriches our understanding of the potential genetic contributions to T2DM complicated with ILD. MUC5B SNP rs2943512 (A > C) or the up-regulated MUC5B in bronchial epithelial cells may be an important factor in promoting ILD inT2DM patients, laying a foundation for future exploration about the pathogenesis of T2DM complicated with ILD.
Collapse
Affiliation(s)
- Qinghua Zhang
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Yan Wang
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Chang Tian
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Jinyan Yu
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Yanlei Li
- Department of Laboratory Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Junling Yang
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, China
- *Correspondence: Junling Yang,
| |
Collapse
|
10
|
Sex-dependent deterioration of cardiac function and molecular alterations in age- and disease-associated RAGE overexpression. Mech Ageing Dev 2022; 203:111635. [DOI: 10.1016/j.mad.2022.111635] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 12/14/2021] [Accepted: 01/28/2022] [Indexed: 11/27/2022]
|
11
|
Diekmann F, Chouvarine P, Sallmon H, Meyer-Kobbe L, Kieslich M, Plouffe BD, Murthy SK, Lichtinghagen R, Legchenko E, Hansmann G. Soluble Receptor for Advanced Glycation End Products (sRAGE) Is a Sensitive Biomarker in Human Pulmonary Arterial Hypertension. Int J Mol Sci 2021; 22:ijms22168591. [PMID: 34445297 PMCID: PMC8395319 DOI: 10.3390/ijms22168591] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 07/28/2021] [Accepted: 07/29/2021] [Indexed: 01/31/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive condition with an unmet need for early diagnosis, better monitoring, and risk stratification. The receptor for advanced glycation end products (RAGE) is activated in response to hypoxia and vascular injury, and is associated with inflammation, cell proliferation and migration in PAH. For the adult cohort, we recruited 120 patients with PAH, 83 with idiopathic PAH (IPAH) and 37 with connective tissue disease-associated PAH (CTD-PAH), and 48 controls, and determined potential plasma biomarkers by enzyme-linked immunoassay. The established heart failure marker NTproBNP and IL-6 plasma levels were several-fold higher in both adult IPAH and CTD-PAH patients versus controls. Plasma soluble RAGE (sRAGE) was elevated in IPAH patients (3044 ± 215.2 pg/mL) and was even higher in CTD-PAH patients (3332 ± 321.6 pg/mL) versus controls (1766 ± 121.9 pg/mL; p < 0.01). All three markers were increased in WHO functional class II+III PAH versus controls (p < 0.001). Receiver-operating characteristic analysis revealed that sRAGE has diagnostic accuracy comparable to prognostic NTproBNP, and even outperforms NTproBNP in the distinction of PAH FC I from controls. Lung tissue RAGE expression was increased in IPAH versus controls (mRNA) and was located predominantly in the PA intima, media, and inflammatory cells in the perivascular space (immunohistochemistry). In the pediatric cohort, plasma sRAGE concentrations were higher than in adults, but were similar in PH (n = 10) and non-PH controls (n = 10). Taken together, in the largest adult sRAGE PAH study to date, we identify plasma sRAGE as a sensitive and accurate PAH biomarker with better performance than NTproBNP in the distinction of mild PAH from controls.
Collapse
Affiliation(s)
- Franziska Diekmann
- Department of Pediatric Cardiology and Critical Care, Hannover Medical School, 30625 Hannover, Germany; (F.D.); (P.C.); (L.M.-K.); (E.L.)
| | - Philippe Chouvarine
- Department of Pediatric Cardiology and Critical Care, Hannover Medical School, 30625 Hannover, Germany; (F.D.); (P.C.); (L.M.-K.); (E.L.)
| | - Hannes Sallmon
- Department of Pediatric Cardiology, Charité University Medical Center, 13353 Berlin, Germany; (H.S.); (M.K.)
| | - Louisa Meyer-Kobbe
- Department of Pediatric Cardiology and Critical Care, Hannover Medical School, 30625 Hannover, Germany; (F.D.); (P.C.); (L.M.-K.); (E.L.)
| | - Moritz Kieslich
- Department of Pediatric Cardiology, Charité University Medical Center, 13353 Berlin, Germany; (H.S.); (M.K.)
| | - Brian D. Plouffe
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA; (B.D.P.); (S.K.M.)
- Department of STEM, Regis College, Weston, MA 02493, USA
| | - Shashi K. Murthy
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA; (B.D.P.); (S.K.M.)
- Flaskworks, LLC, Boston, MA 02118, USA
| | - Ralf Lichtinghagen
- Institute of Clinical Chemistry, Hannover Medical School, 30625 Hannover, Germany;
| | - Ekaterina Legchenko
- Department of Pediatric Cardiology and Critical Care, Hannover Medical School, 30625 Hannover, Germany; (F.D.); (P.C.); (L.M.-K.); (E.L.)
| | - Georg Hansmann
- Department of Pediatric Cardiology and Critical Care, Hannover Medical School, 30625 Hannover, Germany; (F.D.); (P.C.); (L.M.-K.); (E.L.)
- Correspondence: ; Tel.: +49-511-532-9594
| |
Collapse
|
12
|
Perkins TN, Oury TD. The perplexing role of RAGE in pulmonary fibrosis: causality or casualty? Ther Adv Respir Dis 2021; 15:17534666211016071. [PMID: 34275342 PMCID: PMC8293846 DOI: 10.1177/17534666211016071] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive and fatal lung disease in which most patients die within 3 years of diagnosis. With an unknown etiology, IPF results in progressive fibrosis of the lung parenchyma, diminishing normal lung function, which results in respiratory failure, and eventually, death. While few therapies are available to reduce disease progression, patients continue to advance toward respiratory failure, leaving lung transplantation the only viable option for survival. As incidence and mortality rates steadily increase, the need for novel therapeutics is imperative. The receptor for advanced glycation endproducts (RAGE) is most highly expressed in the lungs and plays a significant role in a number of chronic lung diseases. RAGE has long been linked to IPF; however, confounding data from both human and experimental studies have left an incomplete and perplexing story. This review examines the present understanding of the role of RAGE in human and experimental models of IPF, drawing parallels to recent advances in RAGE biology. Moreover, this review discusses the role of RAGE in lung injury response, type 2 immunity, and cellular senescence, and how such mechanisms may relate to RAGE as both a biomarker of disease progression and potential therapeutic target in IPF.The reviews of this paper are available via the supplemental material section.
Collapse
Affiliation(s)
- Timothy N Perkins
- Department of Pathology, University of Pittsburgh School of Medicine, 3550 Terrace Street, S-784 Scaife Hall, Pittsburgh, PA 15261, USA
| | - Tim D Oury
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
13
|
Transforming growth factor-β2-mediated mesenchymal transition in lens epithelial cells is repressed in the absence of RAGE. Biochem J 2021; 478:2285-2296. [PMID: 34143864 DOI: 10.1042/bcj20210069] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 05/19/2021] [Accepted: 05/27/2021] [Indexed: 12/27/2022]
Abstract
Transforming growth factor-β2 (TGFβ2)-mediated epithelial to mesenchymal transition (EMT) in lens epithelial cells (LECs) has been implicated in fibrosis associated with secondary cataracts. In this study, we investigated whether the receptor for advanced glycation end products (RAGE) plays a role in TGFβ2-mediated EMT in LECs. Unlike in the LECs from wild-type mice, TGFβ2 failed to elicit an EMT response in LECs from RAGE knockout mice. The lack of RAGE also diminished TGFβ2-mediated Smad signaling. In addition, treatment with TGFβ2 increased IL-6 levels in LECs from wild-type mice but not in those from RAGE knockout mice. Treatment of human LECs with the RAGE inhibitor FPS-ZM1 reduced TGFβ2-mediated Smad signaling and the EMT response. Unlike that in wild-type lenses, the removal of fiber cell tissue in RAGE knockout lenses did not result in elevated levels of α-smooth muscle actin (α-SMA), fibronectin (FN), and integrin β1 in capsule-adherent LECs. Taken together, these results suggest that TGFβ2 signaling is intricately linked to RAGE. Targeting RAGE could be explored as a therapeutic strategy against secondary cataracts.
Collapse
|
14
|
Rap1a Regulates Cardiac Fibroblast Contraction of 3D Diabetic Collagen Matrices by Increased Activation of the AGE/RAGE Cascade. Cells 2021; 10:cells10061286. [PMID: 34067282 PMCID: PMC8224555 DOI: 10.3390/cells10061286] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 05/12/2021] [Accepted: 05/20/2021] [Indexed: 01/11/2023] Open
Abstract
Cardiovascular disease is a common diabetic complication that can arise when cardiac fibroblasts transition into myofibroblasts. Myofibroblast transition can be induced by advanced glycated end products (AGEs) present in the extracellular matrix (ECM) activating RAGE (receptor for advanced glycated end products) to elicit intracellular signaling. The levels of AGEs are higher under diabetic conditions due to the hyperglycemic conditions present in diabetics. AGE/RAGE signaling has been shown to alter protein expression and ROS production in cardiac fibroblasts, resulting in changes in cellular function, such as migration and contraction. Recently, a small GTPase, Rap1a, has been identified to overlap the AGE/RAGE signaling cascade and mediate changes in protein expression. While Rap1a has been shown to impact AGE/RAGE-induced protein expression, there are currently no data examining the impact Rap1a has on AGE/RAGE-induced cardiac fibroblast function. Therefore, we aimed to determine the impact of Rap1a on AGE/RAGE-mediated cardiac fibroblast contraction, as well as the influence isolated diabetic ECM has on facilitating these effects. In order to address this idea, genetically different cardiac fibroblasts were embedded in 3D collagen matrices consisting of collagen isolated from either non-diabetic of diabetic mice. Fibroblasts were treated with EPAC and/or exogenous AGEs, which was followed by assessment of matrix contraction, protein expression (α-SMA, SOD-1, and SOD-2), and hydrogen peroxide production. The results showed Rap1a overlaps the AGE/RAGE cascade to increase the myofibroblast population and generation of ROS production. The increase in myofibroblasts and oxidative stress appeared to contribute to increased matrix contraction, which was further exacerbated by diabetic conditions. Based off these results, we determined that Rap1a was essential in mediating the response of cardiac fibroblasts to AGEs within diabetic collagen.
Collapse
|
15
|
Sharma A, Kaur S, Sarkar M, Sarin BC, Changotra H. The AGE-RAGE Axis and RAGE Genetics in Chronic Obstructive Pulmonary Disease. Clin Rev Allergy Immunol 2020; 60:244-258. [PMID: 33170477 DOI: 10.1007/s12016-020-08815-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/26/2020] [Indexed: 12/25/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a heterogeneous group of lung diseases limiting the airflow due to narrowing of airways, chronic bronchitis and emphysema that leads to difficulties in breathing. Chronic inflammation is another important characteristic of COPD which leads to immune cell infiltration and helps in the alveolar destruction. Pathology of COPD is driven by various environmental and genetic factors. COPD is mainly associated with the inhalation of toxic agents mainly the cigarette smoke. Receptor for advanced glycation end products (RAGE) has emerged as a pattern recognition receptor and is a multiligand receptor expressed moderately in various cells, tissues and highly in the lungs throughout life. RAGE recognizes various ligands produced by cigarette smoke and its role has been implicated in the pathogenesis of COPD. RAGE ligands have been reported to accumulate in the lungs of patients with COPD. RAGE is a membrane receptor but its truncated form i.e. soluble RAGE (sRAGE) mainly functions as a contender of RAGE and inhibits various RAGE dependent cell signalling. Among the various ligands of RAGE, advanced glycation end products (AGEs) are majorly linked with COPD. Accumulated AGE triggers downstream RAGE-AGE axis in COPD. Moreover, RAGE genetics has long been known to play a vital role in the pathology of various airway diseases including COPD and this gene contains an associated locus. A reliable biomarker is needed for the management of this disease. sRAGE has an inverse correlation with the RAGE showed its importance as a valuable marker in COPD. This review is focused on the role of RAGE, sRAGE, RAGE axis and RAGE genetics in COPD.
Collapse
Affiliation(s)
- Ambika Sharma
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology, Waknaghat, Solan, Himachal Pradesh, 173 234, India
| | - Sargeet Kaur
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology, Waknaghat, Solan, Himachal Pradesh, 173 234, India
| | - Malay Sarkar
- Department of Pulmonary Medicine, Indira Gandhi Medical College, Shimla, Himachal Pradesh, 171 001, India
| | - B C Sarin
- Department of Chest and TB, Sri Guru Ram Das Institute of Medical Sciences and Research, Vallah, Amritsar, 143 501, India
| | - Harish Changotra
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology, Waknaghat, Solan, Himachal Pradesh, 173 234, India.
| |
Collapse
|
16
|
Araki K, Kinoshita R, Tomonobu N, Gohara Y, Tomida S, Takahashi Y, Senoo S, Taniguchi A, Itano J, Yamamoto KI, Murata H, Suzawa K, Shien K, Yamamoto H, Okazaki M, Sugimoto S, Ichimura K, Nishibori M, Miyahara N, Toyooka S, Sakaguchi M. The heterodimer S100A8/A9 is a potent therapeutic target for idiopathic pulmonary fibrosis. J Mol Med (Berl) 2020; 99:131-145. [PMID: 33169236 DOI: 10.1007/s00109-020-02001-x] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 10/12/2020] [Accepted: 10/27/2020] [Indexed: 02/06/2023]
Abstract
In patients with interstitial pneumonia, pulmonary fibrosis is an irreversible condition that can cause respiratory failure. Novel treatments for pulmonary fibrosis are necessary. Inflammation is thought to activate lung fibroblasts, resulting in pulmonary fibrosis. Of the known inflammatory molecules, we have focused on S100A8/A9 from the onset of inflammation to the subsequent progression of inflammation. Our findings confirmed the high expression of S100A8/A9 in specimens from patients with pulmonary fibrosis. An active role of S100A8/A9 was demonstrated not only in the proliferation of fibroblasts but also in the fibroblasts' differentiation to myofibroblasts (the active form of fibroblasts). S100A8/A9 also forced fibroblasts to upregulate the production of collagen. These effects were induced via the receptor of S100A8/A9, i.e., the receptor for advanced glycation end products (RAGE), on fibroblasts. The anti-S100A8/A9 neutralizing antibody inhibited the effects of S100A8/A9 on fibroblasts and suppressed the progression of fibrosis in bleomycin (BLM)-induced pulmonary fibrosis mouse model. Our findings strongly suggest a crucial role of S100A8/A9 in pulmonary fibrosis and the usefulness of S100A8/A9-targeting therapy for fibrosis interstitial pneumonia. HIGHLIGHTS: S100A8/A9 level is highly upregulated in the IPF patients' lungs as well as the blood. S100A8/A9 promotes not only the growth of fibroblasts but also differentiation to myofibroblasts. The cell surface RAGE acts as a crucial receptor to the extracellular S100A8/A9 in fibroblasts. The anti-S100A8/A9 antibody effectively suppresses the progression of IPF in a mouse model. In idiopathic pulmonary fibrosis (IPF), S100A8/A9, a heterodimer composed of S100A8 and S100A9 proteins, plays a crucial role in the onset of inflammation and the subsequent formation of a feed-forward inflammatory loop that promotes fibrosis. (1) The local, pronounced increase in S100A8/A9 in the injured inflammatory lung region-which is provided mainly by the activated neutrophils and macrophages-exerts strong inflammatory signals accompanied by dozens of inflammatory soluble factors including cytokines, chemokines, and growth factors that further act to produce and secrete S100A8/A9, eventually making a sustainable inflammatory circuit that supplies an indefinite presence of S100A8/A9 in the extracellular space with a mal-increased level. (2) The elevated S100A8/A9 compels fibroblasts to activate through receptor for advanced glycation end products (RAGE), one of the major S100A8/A9 receptors, resulting in the activation of NFκB, leading to fibroblast mal-events (e.g., elevated cell proliferation and transdifferentiation to myofibroblasts) that actively produce not only inflammatory cytokines but also collagen matrices. (3) Finally, the S100A8/A9-derived activation of lung fibroblasts under a chronic inflammation state leads to fibrosis events and constantly worsens fibrosis in the lung. Taken together, these findings suggest that the extracellular S100A8/A9 heterodimer protein is a novel mainstay soluble factor for IPF that exerts many functions as described above (1-3). Against this background, we herein applied the developed S100A8/A9 neutralizing antibody to prevent IPF. The IPF imitating lung fibrosis in an IPF mouse model was effectively blocked by treatment with the antibody, leading to enhanced survival. The developed S100A8/A9 antibody, as an innovative novel biologic, may help shed light on the difficulties encountered with IPF therapy in clinical settings.
Collapse
Affiliation(s)
- Kota Araki
- Department of Cell Biology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan.,Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Rie Kinoshita
- Department of Cell Biology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Nahoko Tomonobu
- Department of Cell Biology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Yuma Gohara
- Department of Cell Biology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Shuta Tomida
- Center for Comprehensive Genomic Medicine, Okayama University Hospital, Okayama, Japan
| | - Yuta Takahashi
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Satoru Senoo
- Department of Hematology, Oncology and Respiratory Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Akihiko Taniguchi
- Department of Allergy and Respiratory Medicine, Okayama University Hospital, Okayama, Japan
| | - Junko Itano
- Department of Allergy and Respiratory Medicine, Okayama University Hospital, Okayama, Japan
| | - Ken-Ichi Yamamoto
- Department of Cell Biology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Hitoshi Murata
- Department of Cell Biology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Ken Suzawa
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Kazuhiko Shien
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Hiromasa Yamamoto
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Mikio Okazaki
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Seiichiro Sugimoto
- Department of Organ Transplant Center, Okayama University Hospital, Okayama, Japan
| | - Kouichi Ichimura
- Department of Pathology, Hiroshima City Hiroshima Citizens Hospital, Hiroshima, Japan
| | - Masahiro Nishibori
- Department of Pharmacology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Nobuaki Miyahara
- Department of Allergy and Respiratory Medicine, Okayama University Hospital, Okayama, Japan.,Department of Medical Technology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Shinichi Toyooka
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan.
| | - Masakiyo Sakaguchi
- Department of Cell Biology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan.
| |
Collapse
|
17
|
Liu L, Killoy KM, Vargas MR, Yamamoto Y, Pehar M. Effects of RAGE inhibition on the progression of the disease in hSOD1 G93A ALS mice. Pharmacol Res Perspect 2020; 8:e00636. [PMID: 32776498 PMCID: PMC7415959 DOI: 10.1002/prp2.636] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 07/13/2020] [Indexed: 02/06/2023] Open
Abstract
Astrocytes play a key role in the progression of amyotrophic lateral sclerosis (ALS) by actively inducing the degeneration of motor neurons. Motor neurons isolated from receptor for advanced glycation end products (RAGE)-knockout mice are resistant to the neurotoxic signal derived from ALS-astrocytes. Here, we confirmed that in a co-culture model, the neuronal death induced by astrocytes over-expressing the ALS-linked mutant hSOD1G93A is prevented by the addition of the RAGE inhibitors FPS-ZM1 or RAP. These inhibitors also prevented the motor neuron death induced by spinal cord extracts from symptomatic hSOD1G93A mice. To evaluate the relevance of this neurotoxic mechanism in ALS pathology, we assessed the therapeutic potential of FPS-ZM1 in hSOD1G93A mice. FPS-ZM1 treatment significantly improved hind-limb grip strength in hSOD1G93A mice during the progression of the disease, reduced the expression of atrophy markers in the gastrocnemius muscle, improved the survival of large motor neurons, and reduced gliosis in the ventral horn of the spinal cord. However, we did not observe a statistically significant effect of the drug in symptoms onset nor in the survival of hSOD1G93A mice. Maintenance of hind-limb grip strength was also observed in hSOD1G93A mice with RAGE haploinsufficiency [hSOD1G93A ;RAGE(+/-)], further supporting the beneficial effect of RAGE inhibition on muscle function. However, no benefits were observed after complete RAGE ablation. Moreover, genetic RAGE ablation significantly shortened the median survival of hSOD1G93A mice. These results indicate that the advance of new therapies targeting RAGE in ALS demands a better understanding of its physiological role in a cell type/tissue-specific context.
Collapse
Affiliation(s)
- Liping Liu
- Biomedical Sciences Training ProgramDepartment of Pharmacology and Experimental TherapeuticsMedical University of South CarolinaCharlestonSCUSA
| | - Kelby M. Killoy
- Biomedical Sciences Training ProgramDepartment of Pharmacology and Experimental TherapeuticsMedical University of South CarolinaCharlestonSCUSA
| | | | - Yasuhiko Yamamoto
- Department of Biochemistry and Molecular Vascular BiologyKanazawa University Graduate School of Medical SciencesKanazawaJapan
| | - Mariana Pehar
- Division of Geriatrics and GerontologyDepartment of MedicineUniversity of Wisconsin‐MadisonMadisonWIUSA
- Geriatric Research Education Clinical CenterVeterans Affairs Medical CenterMadisonWIUSA
| |
Collapse
|
18
|
Wang D, Ma Y, Tong X, Zhang Y, Fan H. Diabetes Mellitus Contributes to Idiopathic Pulmonary Fibrosis: A Review From Clinical Appearance to Possible Pathogenesis. Front Public Health 2020; 8:196. [PMID: 32582606 PMCID: PMC7285959 DOI: 10.3389/fpubh.2020.00196] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 04/29/2020] [Indexed: 02/05/2023] Open
Abstract
Diabetes mellitus is a systematic metabolic disease characterized by persistent hyperglycemia, which complications often involve multiple organs and systems including vessels, kidneys, retinas, and nervous system. Idiopathic pulmonary fibrosis is a chronic, progressive, fibrotic disease with usual interstitial pneumonia patterns. With in-depth research, diabetic related lung injury has been confirmed, and the lung is also considered as one of the targeted organs of diabetes, which mainly manifests as the pulmonary fibrosis. Based on that, this review discusses the association between diabetes mellitus and idiopathic pulmonary fibrosis from clinical findings to possible mechanisms.
Collapse
Affiliation(s)
- Dongguang Wang
- Department of Respiratory and Critical Care Medicine, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, China
| | - Yao Ma
- The Center of Gerontology and Geriatrics, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, China
| | - Xiang Tong
- Department of Respiratory and Critical Care Medicine, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, China
| | - Yonggang Zhang
- Department of Periodical Press, West China Hospital, Sichuan University, Chengdu, China
| | - Hong Fan
- Department of Respiratory and Critical Care Medicine, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, China
| |
Collapse
|
19
|
Kim DH, Gu A, Lee JS, Yang EJ, Kashif A, Hong MH, Kim G, Park BS, Lee SJ, Kim IS. Suppressive effects of S100A8 and S100A9 on neutrophil apoptosis by cytokine release of human bronchial epithelial cells in asthma. Int J Med Sci 2020; 17:498-509. [PMID: 32174780 PMCID: PMC7053304 DOI: 10.7150/ijms.37833] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Accepted: 09/05/2019] [Indexed: 12/28/2022] Open
Abstract
S100A8 and S100A9 are important proteins in the pathogenesis of allergy. Asthma is an allergic lung disease, characterized by bronchial inflammation due to leukocytes, bronchoconstriction, and allergen-specific IgE. In this study, we examined the role of S100A8 and S100A9 in the interaction of cytokine release from bronchial epithelial cells, with constitutive apoptosis of neutrophils. S100A8 and S100A9 induce increased secretion of neutrophil survival cytokines such as MCP-1, IL-6 and IL-8. This secretion is suppressed by TLR4 inhibitor), LY294002, AKT inhibitor, PD98059, SB202190, SP600125, and BAY-11-7085. S100A8 and S100A9 also induce the phosphorylation of AKT, ERK, p38 MAPK and JNK, and activation of NF-κB, which were blocked after exposure to TLR4i, LY294002, AKTi, PD98059, SB202190 or SP600125. Furthermore, supernatants collected from bronchial epithelial cells after S100A8 and S100A9 stimulation suppressed the apoptosis of normal and asthmatic neutrophils. These inhibitory mechanisms are involved in suppression of caspase 9 and caspase 3 activation, and BAX expression. The degradation of MCL-1 and BCL-2 was also blocked by S100A8 and S100A9 stimulation. Essentially, neutrophil apoptosis was blocked by co-culture of normal and asthmatic neutrophils with BEAS-2B cells in the presence of S100A8 and S100A9. These findings will enable elucidation of asthma pathogenesis.
Collapse
Affiliation(s)
- Da Hye Kim
- Department of Senior Healthcare, BK21 Plus Program, Graduate School, Eulji University, Daejeon 34824
| | - Ayoung Gu
- Department of Senior Healthcare, BK21 Plus Program, Graduate School, Eulji University, Daejeon 34824
| | - Ji-Sook Lee
- Department of Clinical Laboratory Science, Wonkwang Health Science University, Iksan, 54538
| | - Eun Ju Yang
- Department of Clinical Laboratory Science, Daegu Haany University, Gyeongsan, 38610
| | - Ayesha Kashif
- Department of Senior Healthcare, BK21 Plus Program, Graduate School, Eulji University, Daejeon 34824
| | - Min Hwa Hong
- Department of Senior Healthcare, BK21 Plus Program, Graduate School, Eulji University, Daejeon 34824
| | - Geunyeong Kim
- Department of Senior Healthcare, BK21 Plus Program, Graduate School, Eulji University, Daejeon 34824
| | - Beom Seok Park
- Department of Senior Healthcare, BK21 Plus Program, Graduate School, Eulji University, Daejeon 34824.,Department of Biomedical Laboratory Science, College of Health Science, Eulji University, Seongnam 13135
| | - Soo Jin Lee
- Department of Pediatrics, School of Medicine, Eulji University, Daejeon, 301-746
| | - In Sik Kim
- Department of Senior Healthcare, BK21 Plus Program, Graduate School, Eulji University, Daejeon 34824.,Department of Biomedical Laboratory Science, School of Medicine, Eulji University, Daejeon 34824, Republic of Korea
| |
Collapse
|
20
|
Zhao S, Luo G, Wu H, Zhang L. Placental growth factor gene silencing mitigates the epithelial‑to‑mesenchymal transition via the p38 MAPK pathway in rats with hyperoxia‑induced lung injury. Mol Med Rep 2019; 20:4867-4874. [PMID: 31702808 PMCID: PMC6854522 DOI: 10.3892/mmr.2019.10785] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 07/26/2019] [Indexed: 02/06/2023] Open
Abstract
Hyperoxia may cause pulmonary fibrosis in neonates and is characterized by the epithelial-to-mesenchymal 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 PLGF-silencing lentiviral plasmid. Western blot analysis and immunohistochemistry revealed that expression levels of the EMT-related protein epithelial-cadherin were increased, whereas its inhibitor protein zinc-finger E-box binding homeobox 2 was decreased in these rats. These data demonstrated that PLGF silencing may significantly mitigate hyperoxia-induced EMT in rat lung tissue. Additionally, an increase in phosphorylated-p38 MAPK protein expression indicated that PLGF may be able to regulate hyperoxia-induced lung injury in rats via the p38 MAPK pathway.
Collapse
Affiliation(s)
- Shuang Zhao
- Department of Pediatrics, The Fourth People's Hospital of Shenyang, Shenyang, Liaoning 110003, P.R. China
| | - Gang Luo
- Department of Pediatrics, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Hongmin Wu
- Department of Neonatology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Liang Zhang
- Department of Neonatology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
21
|
Khaket TP, Kang SC, Mukherjee TK. The Potential of Receptor for Advanced Glycation End Products (RAGE) as a Therapeutic Target for Lung Associated Diseases. Curr Drug Targets 2019; 20:679-689. [DOI: 10.2174/1389450120666181120102159] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 10/17/2018] [Accepted: 11/02/2018] [Indexed: 12/27/2022]
Abstract
The receptor for advanced glycation end products (RAGE) is a multi-ligand pattern recognition
receptor that is highly expressed in lung epithelial cells. It helps alveolar epithelial cells to
maintain their morphology and specific architecture. However, in various pathophysiological conditions,
pulmonary tissues express a supraphysiological level of RAGE and its ligands including advanced
glycation end products, high mobility group box 1 proteins, and S100 proteins. On interaction
with RAGE, these ligands stimulate downstream signaling that generates inflammation and oxidative
stress leading to asthma, chronic obstructive pulmonary disease, lung cancers, idiopathic pulmonary
fibrosis, acute lung injury, pneumonia, bronchopulmonary dysplasia, cystic fibrosis, and sepsis. Thus,
pharmacological agents that can either suppress the production of RAGE or block its biological activity
would offer promising therapeutic value against pathogenesis of the aforementioned lungassociated
diseases. This review presents a comprehensive overview of the recent progress made in
defining the functions of RAGE in lung-associated diseases.
Collapse
Affiliation(s)
| | - Sun Chul Kang
- Department of Biotechnology, Daegu University, Gyeongsan, Gyeongbuk, Korea
| | - Tapan Kumar Mukherjee
- Department of Biotechnology, Maharishi Markandeshwar University, Mullana, Haryana, India
| |
Collapse
|
22
|
Machahua C, Montes-Worboys A, Planas-Cerezales L, Buendia-Flores R, Molina-Molina M, Vicens-Zygmunt V. Serum AGE/RAGEs as potential biomarker in idiopathic pulmonary fibrosis. Respir Res 2018; 19:215. [PMID: 30409203 PMCID: PMC6225674 DOI: 10.1186/s12931-018-0924-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 10/28/2018] [Indexed: 12/16/2022] Open
Abstract
Background The soluble receptor for advanced glycation end-products (sRAGE) has been suggested that it acts as a decoy for capturing advanced glycation end-products (AGEs) and inhibits the activation of the oxidative stress and apoptotic pathways. Lung AGEs/sRAGE is increased in idiopathic pulmonary fibrosis (IPF). The objective of the study was to evaluate the AGEs and sRAGE levels in serum as a potential biomarker in IPF. Methods Serum samples were collected from adult patients: 62 IPF, 22 chronic hypersensitivity pneumonitis (cHP), 20 fibrotic non-specific interstitial pneumonia (fNSIP); and 12 healthy controls. In addition, 23 IPF patients were re-evaluated after 3-year follow-up period. Epidemiological and clinical features were recorded: age, sex, smoking habits, and lung function. AGEs and sRAGE were evaluated by ELISA, and the results were correlated with pulmonary functional test values. Results IPF and cHP groups presented a significant increase of AGE/sRAGE serum concentration compared with fNSIP patients. Moreover, an inverse correlation between AGEs and sRAGE levels were found in IPF, and serum sRAGE at diagnosis correlated with FVC and DLCO values. Additionally, changes in serum AGEs and sRAGE correlated with % change of FVC, DLCO and TLC during the follow-up. sRAGE levels below 428.25 pg/ml evolved poor survival rates. Conclusions These findings demonstrate that the increase of AGE/sRAGE ratio is higher in IPF, although the levels were close to cHP. AGE/sRAGE increase correlates with respiratory functional progression. Furthermore, the concentration of sRAGE in blood stream at diagnosis and follow-up could be considered as a potential prognostic biomarker. Electronic supplementary material The online version of this article (10.1186/s12931-018-0924-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Carlos Machahua
- Pneumology Research Group, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain.,Biomedical Research Network Centers in Respiratory Diseases (CIBERES), Barcelona, Spain
| | - Ana Montes-Worboys
- Pneumology Research Group, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain.,Biomedical Research Network Centers in Respiratory Diseases (CIBERES), Barcelona, Spain.,Unit of Interstitial Lung Diseases, Department of Pneumology, University Hospital of Bellvitge, C. Feixa Llarga sn., 08907 L'Hospitalet de Llobregat, Barcelona, Spain
| | - Lurdes Planas-Cerezales
- Pneumology Research Group, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain.,Unit of Interstitial Lung Diseases, Department of Pneumology, University Hospital of Bellvitge, C. Feixa Llarga sn., 08907 L'Hospitalet de Llobregat, Barcelona, Spain
| | | | - Maria Molina-Molina
- Pneumology Research Group, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain. .,Biomedical Research Network Centers in Respiratory Diseases (CIBERES), Barcelona, Spain. .,Unit of Interstitial Lung Diseases, Department of Pneumology, University Hospital of Bellvitge, C. Feixa Llarga sn., 08907 L'Hospitalet de Llobregat, Barcelona, Spain.
| | - Vanesa Vicens-Zygmunt
- Pneumology Research Group, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain.,Unit of Interstitial Lung Diseases, Department of Pneumology, University Hospital of Bellvitge, C. Feixa Llarga sn., 08907 L'Hospitalet de Llobregat, Barcelona, Spain
| |
Collapse
|
23
|
Antognelli C, Cecchetti R, Riuzzi F, Peirce MJ, Talesa VN. Glyoxalase 1 sustains the metastatic phenotype of prostate cancer cells via EMT control. J Cell Mol Med 2018; 22:2865-2883. [PMID: 29504694 PMCID: PMC5908125 DOI: 10.1111/jcmm.13581] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 01/23/2018] [Indexed: 01/07/2023] Open
Abstract
Metastasis is the primary cause of death in prostate cancer (PCa) patients. Effective therapeutic intervention in metastatic PCa is undermined by our poor understanding of its molecular aetiology. Defining the mechanisms underlying PCa metastasis may lead to insights into how to decrease morbidity and mortality in this disease. Glyoxalase 1 (Glo1) is the detoxification enzyme of methylglyoxal (MG), a potent precursor of advanced glycation end products (AGEs). Hydroimidazolone (MG-H1) and argpyrimidine (AP) are AGEs originating from MG-mediated post-translational modification of proteins at arginine residues. AP is involved in the control of epithelial to mesenchymal transition (EMT), a crucial determinant of cancer metastasis and invasion, whose regulation mechanisms in malignant cells are still emerging. Here, we uncover a novel mechanism linking Glo1 to the maintenance of the metastatic phenotype of PCa cells by controlling EMT by engaging the tumour suppressor miR-101, MG-H1-AP and TGF-β1/Smad signalling. Moreover, circulating levels of Glo1, miR-101, MG-H1-AP and TGF-β1 in patients with metastatic compared with non-metastatic PCa support our in vitro results, demonstrating their clinical relevance. We suggest that Glo1, together with miR-101, might be potential therapeutic targets for metastatic PCa, possibly by metformin administration.
Collapse
Affiliation(s)
- Cinzia Antognelli
- Department of Experimental MedicineUniversity of PerugiaPerugiaItaly
| | - Rodolfo Cecchetti
- Department of Experimental MedicineUniversity of PerugiaPerugiaItaly
| | - Francesca Riuzzi
- Department of Experimental MedicineUniversity of PerugiaPerugiaItaly
| | - Matthew J. Peirce
- Department of Experimental MedicineUniversity of PerugiaPerugiaItaly
| | | |
Collapse
|
24
|
Zhao J, Geng L, Duan G, Xu W, Cheng Y, Huang Z, Zhou Z, Gong S. REC8 inhibits EMT by downregulating EGR1 in gastric cancer cells. Oncol Rep 2018; 39:1583-1590. [PMID: 29393474 PMCID: PMC5868373 DOI: 10.3892/or.2018.6244] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 12/07/2017] [Indexed: 12/11/2022] Open
Abstract
REC8 is a component of the meiotic cohesion complex that plays a critical role in chromosome dynamics during meiosis. However, the functional role of REC8 in gastric cancer has not been elucidated. In the present study, REC8 suppressed the growth and metastasis of gastric cancer cells in vitro. Whole Human Genome Oligo Microarray results revealed that a wide range of genes with broad function were targeted by REC8. Among them early growth response-1 (EGR1), a transcription factor and an epithelial-mesenchymal transition (EMT)-associated protein in the AGR-RAGE pathway was significantly downregulated when REC8 was overexpressed in gastric cancer cells. We hypothesized that REC8 inhibits EMT by downregulating EGR1 in gastric cancer cells. Consistent with our prediction, REC8 overexpression decreased EMT in gastric cancer cells, whereas the REC8 ablation reversed these effects. In addition, the phenotypes of EGR1 overexpressed cells were similar to the phenotypes of REC8 ablated cells. Furthermore, we determined that REC8 interacted with EGR1, and inhibited EMT in gastric cancer cells. We thus propose further studies of the pathways associated with REC8 and EGR1 to potentially find novel targets in the treatment for gastric cancer.
Collapse
Affiliation(s)
- Junhong Zhao
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510120, P.R. China
| | - Lanlan Geng
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510120, P.R. China
| | - Gaoyang Duan
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510120, P.R. China
| | - Wanfu Xu
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510120, P.R. China
| | - Yang Cheng
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510120, P.R. China
| | - Zhiliang Huang
- Cancer Center of Guangzhou Medical University, Guangzhou, Guangdong 510089, P.R. China
| | - Zhenwen Zhou
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510120, P.R. China
| | - Sitang Gong
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510120, P.R. China
| |
Collapse
|
25
|
Byun K, Yoo Y, Son M, Lee J, Jeong GB, Park YM, Salekdeh GH, Lee B. Advanced glycation end-products produced systemically and by macrophages: A common contributor to inflammation and degenerative diseases. Pharmacol Ther 2017; 177:44-55. [PMID: 28223234 DOI: 10.1016/j.pharmthera.2017.02.030] [Citation(s) in RCA: 244] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Advanced glycation end products (AGEs) and their receptor have been implicated in the progressions of many intractable diseases, such as diabetes and atherosclerosis, and are also critical for pathologic changes in chronic degenerative diseases, such as Alzheimer's disease, Parkinson's disease, and alcoholic brain damage. Recently activated macrophages were found to be a source of AGEs, and the most abundant form of AGEs, AGE-albumin excreted by macrophages has been implicated in these diseases and to act through common pathways. AGEs inhibition has been shown to prevent the pathogenesis of AGEs-related diseases in human, and therapeutic advances have resulted in several agents that prevent their adverse effects. Recently, anti-inflammatory molecules that inhibit AGEs have been shown to be good candidates for ameliorating diabetic complications as well as degenerative diseases. This review was undertaken to present, discuss, and clarify current understanding regarding AGEs formation in association with macrophages, different diseases, therapeutic and diagnostic strategy and links with RAGE inhibition.
Collapse
Affiliation(s)
- Kyunghee Byun
- Center for Genomics and Proteomics, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 406-840, Republic of Korea; Department of Anatomy and Cell Biology, Gachon University Graduate School of Medicine, Incheon 406-799, Republic of Korea
| | - YongCheol Yoo
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon 305-811, Republic of Korea
| | - Myeongjoo Son
- Department of Anatomy and Cell Biology, Gachon University Graduate School of Medicine, Incheon 406-799, Republic of Korea
| | - Jaesuk Lee
- Center for Genomics and Proteomics, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 406-840, Republic of Korea
| | - Goo-Bo Jeong
- Department of Anatomy and Cell Biology, Gachon University Graduate School of Medicine, Incheon 406-799, Republic of Korea
| | - Young Mok Park
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon 305-811, Republic of Korea.
| | - Ghasem Hosseini Salekdeh
- Department of Molecular Systems Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | - Bonghee Lee
- Center for Genomics and Proteomics, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 406-840, Republic of Korea; Department of Anatomy and Cell Biology, Gachon University Graduate School of Medicine, Incheon 406-799, Republic of Korea.
| |
Collapse
|
26
|
Clair G, Piehowski PD, Nicola T, Kitzmiller JA, Huang EL, Zink EM, Sontag RL, Orton DJ, Moore RJ, Carson JP, Smith RD, Whitsett JA, Corley RA, Ambalavanan N, Ansong C. Spatially-Resolved Proteomics: Rapid Quantitative Analysis of Laser Capture Microdissected Alveolar Tissue Samples. Sci Rep 2016; 6:39223. [PMID: 28004771 PMCID: PMC5177886 DOI: 10.1038/srep39223] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 11/16/2016] [Indexed: 01/12/2023] Open
Abstract
Laser capture microdissection (LCM)-enabled region-specific tissue analyses are critical to better understand complex multicellular processes. However, current proteomics workflows entail several manual sample preparation steps and are challenged by the microscopic mass-limited samples generated by LCM, impacting measurement robustness, quantification and throughput. Here, we coupled LCM with a proteomics workflow that provides fully automated analysis of proteomes from microdissected tissues. Benchmarking against the current state-of-the-art in ultrasensitive global proteomics (FASP workflow), our approach demonstrated significant improvements in quantification (~2-fold lower variance) and throughput (>5 times faster). Using our approach we for the first time characterized, to a depth of >3,400 proteins, the ontogeny of protein changes during normal lung development in microdissected alveolar tissue containing only 4,000 cells. Our analysis revealed seven defined modules of coordinated transcription factor-signaling molecule expression patterns, suggesting a complex network of temporal regulatory control directs normal lung development with epigenetic regulation fine-tuning pre-natal developmental processes.
Collapse
Affiliation(s)
- Geremy Clair
- Biological Science Division, Pacific Northwest National Laboratory, Richland, WA 99352, USA
| | - Paul D Piehowski
- Biological Science Division, Pacific Northwest National Laboratory, Richland, WA 99352, USA
| | - Teodora Nicola
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL 35249, USA
| | - Joseph A Kitzmiller
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Eric L Huang
- Biological Science Division, Pacific Northwest National Laboratory, Richland, WA 99352, USA
| | - Erika M Zink
- Biological Science Division, Pacific Northwest National Laboratory, Richland, WA 99352, USA
| | - Ryan L Sontag
- Biological Science Division, Pacific Northwest National Laboratory, Richland, WA 99352, USA
| | - Daniel J Orton
- Biological Science Division, Pacific Northwest National Laboratory, Richland, WA 99352, USA
| | - Ronald J Moore
- Biological Science Division, Pacific Northwest National Laboratory, Richland, WA 99352, USA
| | - James P Carson
- Texas Advanced Computing Center, University of Texas at Austin, Austin, TX 78712, USA
| | - Richard D Smith
- Biological Science Division, Pacific Northwest National Laboratory, Richland, WA 99352, USA
| | - Jeffrey A Whitsett
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Richard A Corley
- Biological Science Division, Pacific Northwest National Laboratory, Richland, WA 99352, USA
| | | | - Charles Ansong
- Biological Science Division, Pacific Northwest National Laboratory, Richland, WA 99352, USA
| |
Collapse
|
27
|
Machahua C, Montes-Worboys A, Llatjos R, Escobar I, Dorca J, Molina-Molina M, Vicens-Zygmunt V. Increased AGE-RAGE ratio in idiopathic pulmonary fibrosis. Respir Res 2016; 17:144. [PMID: 27816054 PMCID: PMC5097848 DOI: 10.1186/s12931-016-0460-2] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 10/28/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The abnormal epithelial-mesenchymal restorative capacity in idiopathic pulmonary fibrosis (IPF) has been recently associated with an accelerated aging process as a key point for the altered wound healing. The advanced glycation end-products (AGEs) are the consequence of non-enzymatic reactions between lipid and protein with several oxidants in the aging process. The receptor for AGEs (RAGEs) has been implicated in the lung fibrotic process and the alveolar homeostasis. However, this AGE-RAGE aging pathway has been under-explored in IPF. METHODS Lung samples from 16 IPF and 9 control patients were obtained through surgical lung biopsy. Differences in AGEs and RAGE expression between both groups were evaluated by RT-PCR, Western blot and immunohistochemistry. The effect of AGEs on cell viability of primary lung fibrotic fibroblasts and alveolar epithelial cells was assessed. Cell transformation of fibrotic fibroblasts cultured into glycated matrices was evaluated in different experimental conditions. RESULTS Our study demonstrates an increase of AGEs together with a decrease of RAGEs in IPF lungs, compared with control samples. Two specific AGEs involved in aging, pentosidine and Nε-Carboxymethyl lysine, were significantly increased in IPF samples. The immunohistochemistry identified higher staining of AGEs related to extracellular matrix (ECM) proteins and the apical surface of the alveolar epithelial cells (AECs) surrounding fibroblast foci in fibrotic lungs. On the other hand, RAGE location was present at the cell membrane of AECs in control lungs, while it was almost missing in pulmonary fibrotic tissue. In addition, in vitro cultures showed that the effect of AGEs on cell viability was different for AECs and fibrotic fibroblasts. AGEs decreased cell viability in AECs, even at low concentration, while fibroblast viability was less affected. Furthermore, fibroblast to myofibroblast transformation could be enhanced by ECM glycation. CONCLUSIONS All of these findings suggest a possible role of the increased ratio AGEs-RAGEs in IPF, which could be a relevant accelerating aging tissue reaction in the abnormal wound healing of the lung fibrotic process.
Collapse
Affiliation(s)
- Carlos Machahua
- Pneumology Research Group, IDIBELL, University of Barcelona, Barcelona, Spain
- Department of Pneumology, Unit of Interstitial Lung Diseases, University Hospital of Bellvitge, Barcelona, Spain
| | - Ana Montes-Worboys
- Pneumology Research Group, IDIBELL, University of Barcelona, Barcelona, Spain
- Department of Pneumology, Unit of Interstitial Lung Diseases, University Hospital of Bellvitge, Barcelona, Spain
- Research Network in Respiratory Diseases (CIBERES), Madrid, Spain
| | - Roger Llatjos
- Department of Pathology, University Hospital of Bellvitge, Barcelona, Spain
| | - Ignacio Escobar
- Department of Thoracic Surgery, University Hospital of Bellvitge, Barcelona, Spain
| | - Jordi Dorca
- Pneumology Research Group, IDIBELL, University of Barcelona, Barcelona, Spain
- Department of Pneumology, Unit of Interstitial Lung Diseases, University Hospital of Bellvitge, Barcelona, Spain
- Research Network in Respiratory Diseases (CIBERES), Madrid, Spain
| | - Maria Molina-Molina
- Pneumology Research Group, IDIBELL, University of Barcelona, Barcelona, Spain
- Department of Pneumology, Unit of Interstitial Lung Diseases, University Hospital of Bellvitge, Barcelona, Spain
- Research Network in Respiratory Diseases (CIBERES), Madrid, Spain
| | - Vanesa Vicens-Zygmunt
- Pneumology Research Group, IDIBELL, University of Barcelona, Barcelona, Spain
- Department of Pneumology, Unit of Interstitial Lung Diseases, University Hospital of Bellvitge, Barcelona, Spain
| |
Collapse
|
28
|
Raghavan CT, Smuda M, Smith AJO, Howell S, Smith DG, Singh A, Gupta P, Glomb MA, Wormstone IM, Nagaraj RH. AGEs in human lens capsule promote the TGFβ2-mediated EMT of lens epithelial cells: implications for age-associated fibrosis. Aging Cell 2016; 15:465-76. [PMID: 26853893 PMCID: PMC4854921 DOI: 10.1111/acel.12450] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/10/2016] [Indexed: 11/30/2022] Open
Abstract
Proteins in basement membrane (BM) are long‐lived and accumulate chemical modifications during aging; advanced glycation endproduct (AGE) formation is one such modification. The human lens capsule is a BM secreted by lens epithelial cells. In this study, we have investigated the effect of aging and cataracts on the AGE levels in the human lens capsule and determined their role in the epithelial‐to‐mesenchymal transition (EMT) of lens epithelial cells. EMT occurs during posterior capsule opacification (PCO), also known as secondary cataract formation. We found age‐dependent increases in several AGEs and significantly higher levels in cataractous lens capsules than in normal lens capsules measured by LC‐MS/MS. The TGFβ2‐mediated upregulation of the mRNA levels (by qPCR) of EMT‐associated proteins was significantly enhanced in cells cultured on AGE‐modified BM and human lens capsule compared with those on unmodified proteins. Such responses were also observed for TGFβ1. In the human capsular bag model of PCO, the AGE content of the capsule proteins was correlated with the synthesis of TGFβ2‐mediated α‐smooth muscle actin (αSMA). Taken together, our data imply that AGEs in the lens capsule promote the TGFβ2‐mediated fibrosis of lens epithelial cells during PCO and suggest that AGEs in BMs could have a broader role in aging and diabetes‐associated fibrosis.
Collapse
Affiliation(s)
- Cibin T. Raghavan
- Department of Ophthalmology & Visual Sciences Case Western Reserve University School of Medicine Cleveland OH USA
- Department of Ophthalmology University of Colorado School of Medicine Aurora CO USA
| | - Mareen Smuda
- Institute of Chemistry Martin‐Luther‐University Halle‐Wittenberg Halle/Saale Germany
| | | | - Scott Howell
- Visual Sciences Research Center Case Western Reserve University School of Medicine Cleveland OH USA
| | - Dawn G. Smith
- Visual Sciences Research Center Case Western Reserve University School of Medicine Cleveland OH USA
| | | | - Pankaj Gupta
- University Hospitals Eye Institute Cleveland OH USA
| | - Marcus A. Glomb
- Institute of Chemistry Martin‐Luther‐University Halle‐Wittenberg Halle/Saale Germany
| | | | - Ram H. Nagaraj
- Department of Ophthalmology & Visual Sciences Case Western Reserve University School of Medicine Cleveland OH USA
- Department of Ophthalmology University of Colorado School of Medicine Aurora CO USA
| |
Collapse
|
29
|
Antognelli C, Gambelunghe A, Muzi G, Talesa VN. Glyoxalase I drives epithelial-to-mesenchymal transition via argpyrimidine-modified Hsp70, miR-21 and SMAD signalling in human bronchial cells BEAS-2B chronically exposed to crystalline silica Min-U-Sil 5: Transformation into a neoplastic-like phenotype. Free Radic Biol Med 2016; 92:110-125. [PMID: 26784015 DOI: 10.1016/j.freeradbiomed.2016.01.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 01/12/2016] [Accepted: 01/13/2016] [Indexed: 12/15/2022]
Abstract
Glyoxalase I (Glo1) is the main scavenging enzyme of methylglyoxal (MG), a potent precursor of advanced glycation end products (AGEs). AGEs are known to control multiple biological processes, including epithelial to mesenchymal transition (EMT), a multistep phenomenon associated with cell transformation, playing a major role in a variety of diseases, including cancer. Crystalline silica is a well-known occupational health hazard, responsible for a great number of human pulmonary diseases, such as silicosis. There is still much debate concerning the carcinogenic role of crystalline silica, mainly due to the lack of a causal demonstration between silica exposure and carcinogenesis. It has been suggested that EMT might play a role in crystalline silica-induced lung neoplastic transformation. The aim of this study was to investigate whether, and by means of which mechanism, the antiglycation defence Glo1 is involved in Min-U-Sil 5 (MS5) crystalline silica-induced EMT in BEAS-2B human bronchial epithelial cells chronically exposed, and whether this is associated with the beginning of a neoplastic-like transformation process. By using gene silencing/overexpression and scavenging/inhibitory agents, we demonstrated that MS5 induced hydrogen peroxide-mediated c-Jun-dependent Glo1 up-regulation which resulted in a decrease in the Argpyrimidine-modified Hsp70 protein level which triggered EMT in a novel mechanism involving miR-21 and SMAD signalling. The observed EMT was associated with a neoplastic-like phenotype. The results obtained provide a causal in vitro demonstration of the MS5 pro-carcinogenic transforming role and more importantly they provide new insights into the mechanisms involved in this process, thus opening new paths in research concerning the in vivo study of the carcinogenic potential of crystalline silica.
Collapse
Affiliation(s)
- Cinzia Antognelli
- Department of Experimental Medicine, School of Medicine and Surgery, University of Perugia, Piazzale L. Severi 1, 06129 Perugia, Italy.
| | - Angela Gambelunghe
- Department of Medicine, School of Medicine and Surgery, University of Perugia, Piazzale L. Severi 1, 06129 Perugia, Italy.
| | - Giacomo Muzi
- Department of Medicine, School of Medicine and Surgery, University of Perugia, Piazzale L. Severi 1, 06129 Perugia, Italy.
| | - Vincenzo Nicola Talesa
- Department of Experimental Medicine, School of Medicine and Surgery, University of Perugia, Piazzale L. Severi 1, 06129 Perugia, Italy.
| |
Collapse
|
30
|
Antifibrotic properties of receptor for advanced glycation end products in idiopathic pulmonary fibrosis. Pulm Pharmacol Ther 2015; 35:34-41. [PMID: 26545872 DOI: 10.1016/j.pupt.2015.10.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Revised: 10/02/2015] [Accepted: 10/28/2015] [Indexed: 11/23/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive chronic interstitial lung disease with poor survival. Previous reports suggested the contributory effect of receptor for advanced glycation end products (RAGE) to the pathogenesis of IPF. But the findings are controversial. The present in vivo study with RAGE null mice, we further confirmed the evidence that lack of RAGE evolves worse bleomycin-induced pulmonary fibrosis compared with control mice. Moreover, RAGE null mice spontaneously developed similar pathogenesis of lung fibrosis via immunohistochemical staining. In addition, we investigated the negative roles of RAGE on epithelial-mesenchymal transition (EMT) indicated by elevated α-smooth muscle actin (α-SMA) and collagen-I (Col-I) deposition in A549 cell treated with transforming growth factor-β (TGF-β), all of which were blocked by sRAGE, a decoy receptor. Furthermore, interacting with the specific ligand as AGE, RAGE blocked TGF-β-induced activation of Smad2, ERK and JNK signals in A549 cells, which were also challenged by sRAGE administration. This present study confirmed an important role of RAGE in vivo and vitro models of pulmonary fibrosis and suggested the therapeutic possibility for pulmonary fibrosis via RAGE regulation.
Collapse
|
31
|
Possible participation of receptor for advanced glycation end products (RAGE) in the origin of cancer stem cells in diabetic patients with colon cancer. Med Hypotheses 2013; 80:620-3. [PMID: 23466064 DOI: 10.1016/j.mehy.2013.02.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Revised: 01/28/2013] [Accepted: 02/07/2013] [Indexed: 01/06/2023]
Abstract
The association between diabetes and the associated increased risk of several solid malignancies has been the subject of investigation for many years, while potential biologic links between the two diseases are incompletely understood. The receptor for advanced glycation end-products (RAGE) signal transduction may represent a focal point in their respective contributions to malignant transformation associated diabetes. While the physiopathology of RAGE axis in promoting malignancies cannot be explained completely by the available mechanism as perpetuating inflammation at tumor microenvironment. In addition, experimental researches revealed a crucial role for upstreams of RAGE signaling pathway in maintaining the stemness properties and tumorigenicity of cancer stem cells. Hence, we hypothesized that RAGE inducing cancer stem cells may be a key determinant in the origin and progression of colon malignant tumors concomitant diabetes. Such an opinion not only bands together the seemingly disparate various complications in diabetes and colon cancers, but also has future implications for risk assessment and biopharmaceutical treatment.
Collapse
|
32
|
Fineschi S, De Cunto G, Facchinetti F, Civelli M, Imbimbo BP, Carnini C, Villetti G, Lunghi B, Stochino S, Gibbons DL, Hayday A, Lungarella G, Cavarra E. Receptor for advanced glycation end products contributes to postnatal pulmonary development and adult lung maintenance program in mice. Am J Respir Cell Mol Biol 2013; 48:164-71. [PMID: 23144333 DOI: 10.1165/rcmb.2012-0111oc] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The role of the receptor for advanced glycation end products (RAGE) in promoting the inflammatory response through activation of NF-κB pathway is well established. Recent findings indicate that RAGE may also have a regulative function in apoptosis, as well as in cellular proliferation, differentiation, and adhesion. Unlike other organs, lung tissue in adulthood and during organ development shows relatively high levels of RAGE expression. Thus a role for the receptor in lung organogenesis and homeostasis may be proposed. To evaluate the role of RAGE in lung development and adult lung homeostasis, we generated hemizygous and homozygous transgenic mice overexpressing human RAGE, and analyzed their lungs from the fourth postnatal day to adulthood. Moderate RAGE hyperexpression during lung development influenced secondary septation, resulting in an impairment of alveolar morphogenesis and leading to significant changes in morphometric parameters such as airspace number and the size of alveolar ducts. An increase in alveolar cell apoptosis and a decrease in cell proliferation were demonstrated by the terminal deoxy-nucleotidyltransferase-mediated dUTP nick end labeling reaction, active caspase-3, and Ki-67 immunohistochemistry. Alterations in elastin organization and deposition and in TGF-β expression were observed. In homozygous mice, the hyperexpression of RAGE resulted in histological changes resembling those changes characterizing human bronchopulmonary dysplasia (BPD). RAGE hyperexpression in the adult lung is associated with an increase of the alveolar destructive index and persistent inflammatory status leading to "destructive" emphysema. These results suggest an important role for RAGE in both alveolar development and lung homeostasis, and open new doors to working hypotheses on the pathogenesis of BPD and chronic obstructive pulmonary disease.
Collapse
Affiliation(s)
- Silvia Fineschi
- Department of Physiopathology, University of Siena, Via Aldo Moro 6, I-53100 Siena, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Abstract
PURPOSE OF REVIEW Pathogenesis of interstitial lung diseases (ILD) has largely been investigated in the context of the most frequent ILD, idiopathic pulmonary fibrosis (IPF). We review studies of epithelial-to-mesenchymal transition (EMT) and discuss its potential contribution to collagen-producing (myo)fibroblasts in IPF. RECENT FINDINGS Endoplasmic reticulum (ER) stress leading to epithelial apoptosis has been reported as a potential etiologic factor in fibrosis. Recent studies further suggest EMT as a link between ER stress and fibrosis. Combinatorial interactions among Smad3, β-catenin and other transcriptional co-activators at the α-smooth muscle actin (α-SMA) promoter provide direct evidence for crosstalk between transforming growth factor-β (TGFβ) and β-catenin pathways during EMT. Lineage tracing yielded conflicting results, with two recent studies supporting and one opposing a role for EMT in lung fibrosis. SUMMARY Advances have been made in elucidating causes and mechanisms of EMT, potentially leading to new treatment options, although contributions of EMT to lung fibrosis in vivo remain controversial. In addition to EMT providing a direct source of (myo)fibroblasts, expression of mesenchymal markers may reflect epithelial injury, in which case inhibition of EMT might be deleterious. EMT-derived cells may also contribute to aberrant epithelial-mesenchymal crosstalk that promotes fibrogenesis.
Collapse
|
34
|
Li M, Song L, Gao X, Chang W, Qin X. Toll-like receptor 4 on islet β cells senses expression changes in high-mobility group box 1 and contributes to the initiation of type 1 diabetes. Exp Mol Med 2012; 44:260-7. [PMID: 22217446 PMCID: PMC3349908 DOI: 10.3858/emm.2012.44.4.021] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Type 1 diabetes mellitus is caused by the autoimmune destruction of β cells within the islets. In recent years, innate immunity has been proposed to play a key role in this process. High-mobility group box 1 (HMGB1), an inflammatory trigger in a number of autoimmune diseases, activates proinflammatory responses following its release from necrotic cells. Our aim was to determine the significance of HMGB1 in the natural history of diabetes in non-obese diabetic (NOD) mice. We observed that the rate of HMGB1 expression in the cytoplasm of islets was much greater in diabetic mice compared with non-diabetic mice. The majority of cells positively stained for toll-like receptor 4 (TLR4) were β cells; few α cells were stained for TLR4. Thus, we examined the effects of anti-TLR4 antibodies on HMGB1 cell surface binding, which confirmed that HMGB1 interacts with TLR4 in isolated islets. Expression changes in HMGB1 and TLR4 were detected throughout the course of diabetes. Our findings indicate that TLR4 is the main receptor on β cells and that HMGB1 may signal via TLR4 to selectively damage β cells rather than α cells during the development of type 1 diabetes mellitus.
Collapse
Affiliation(s)
- Min Li
- Department of General Surgery, Zhongshan Hospital, Shanghai Medical School, Fudan University, Shanghai 200032, China
| | | | | | | | | |
Collapse
|
35
|
Inhibition of TGF-β signaling and decreased apoptosis in IUGR-associated lung disease in rats. PLoS One 2011; 6:e26371. [PMID: 22028866 PMCID: PMC3197638 DOI: 10.1371/journal.pone.0026371] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2011] [Accepted: 09/25/2011] [Indexed: 01/04/2023] Open
Abstract
Intrauterine growth restriction is associated with impaired lung function in adulthood. It is unknown whether such impairment of lung function is linked to the transforming growth factor (TGF)-β system in the lung. Therefore, we investigated the effects of IUGR on lung function, expression of extracellular matrix (ECM) components and TGF-β signaling in rats. IUGR was induced in rats by isocaloric protein restriction during gestation. Lung function was assessed with direct plethysmography at postnatal day (P) 70. Pulmonary activity of the TGF-β system was determined at P1 and P70. TGF-β signaling was blocked in vitro using adenovirus-delivered Smad7. At P70, respiratory airway compliance was significantly impaired after IUGR. These changes were accompanied by decreased expression of TGF-β1 at P1 and P70 and a consistently dampened phosphorylation of Smad2 and Smad3. Furthermore, the mRNA expression levels of inhibitors of TGF-β signaling (Smad7 and Smurf2) were reduced, and the expression of TGF-β-regulated ECM components (e.g. collagen I) was decreased in the lungs of IUGR animals at P1; whereas elastin and tenascin N expression was significantly upregulated. In vitro inhibition of TGF-β signaling in NIH/3T3, MLE 12 and endothelial cells by adenovirus-delivered Smad7 demonstrated a direct effect on the expression of ECM components. Taken together, these data demonstrate a significant impact of IUGR on lung development and function and suggest that attenuated TGF-β signaling may contribute to the pathological processes of IUGR-associated lung disease.
Collapse
|