1
|
Wang L, Liu K, Song Z, Do H, Yang L, Wu J, Jiang L, Yu H. Influence of coordination number and ionic radius on metal ion preference and activity of lanthanide-dependent alcohol dehydrogenase: Insights from mutational studies and density functional theory. Colloids Surf B Biointerfaces 2025; 251:114596. [PMID: 40031112 DOI: 10.1016/j.colsurfb.2025.114596] [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: 11/04/2024] [Revised: 02/18/2025] [Accepted: 02/21/2025] [Indexed: 03/05/2025]
Abstract
Lanthanide (Ln) elements form a cofactor complex with pyrroloquinoline quinone (PQQ) in bacterial alcohol dehydrogenases (Ln3 +-ADH). The lanthanide elements did not support Ln3+-ADH activity equally, with only early lanthanides (La3+-Gd3+) promoting high enzyme activity. However, the early lanthanides did not promote the activity equally and the detailed mechanism of Ln3+-ADH exhibiting different activity in the presence of different light Lns remains obscure. To uncover the role of lanthanides in promoting Ln3+-ADH activity, we systemically characterized the activity of an Ln3+-ADH from Pseudomonas putida KT2440 (PedH) in the presence of various Ln3+ ions. In the results, enzyme activity displayed a bell-shaped trend along with the lanthanide series, with Nd3+ providing the highest activity. Active site mutation analysis revealed that modifying the number of coordinating ligands shifted the metal preference of the enzyme. DFT calculation revealed that the HOMO-LUMO gap, substrate interaction energy and metal ions binding distances were critical for the lanthanides in promoting enzyme activity. This work shed light on the critical role of metal ions in Ln3+-ADH catalysis, providing insights for future exploration and engineering of Ln-dependent proteins.
Collapse
Affiliation(s)
- Lun Wang
- Institute of Bioengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang 310027, China; ZJU-Hangzhou Global Scientific and Technological Innovation Centre, Hangzhou, Zhejiang 311200, China
| | - Ke Liu
- Institute of Bioengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang 310027, China; ZJU-Hangzhou Global Scientific and Technological Innovation Centre, Hangzhou, Zhejiang 311200, China
| | - Zhongdi Song
- Key Laboratory of Pollution Exposure and Health Intervention of Zhejiang Province, Interdisciplinary Research Academy, Zhejiang Shuren University, Hangzhou 310015, China
| | - Hainam Do
- Department of Chemical and Environmental Engineering, University of Nottingham Ningbo China, Ningbo 315100, China; New Materials Institute, University of Nottingham Ningbo China, Ningbo 315042, China; Key Laboratory of Carbonaceous Waste Processing and Process Intensification Research of Zhejiang Province, University of Nottingham Ningbo China, Ningbo 315100, China
| | - Lirong Yang
- Institute of Bioengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang 310027, China; ZJU-Hangzhou Global Scientific and Technological Innovation Centre, Hangzhou, Zhejiang 311200, China
| | - Jianping Wu
- Institute of Bioengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang 310027, China; ZJU-Hangzhou Global Scientific and Technological Innovation Centre, Hangzhou, Zhejiang 311200, China
| | - Ling Jiang
- Institute of Bioengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang 310027, China; ZJU-Hangzhou Global Scientific and Technological Innovation Centre, Hangzhou, Zhejiang 311200, China.
| | - Haoran Yu
- Institute of Bioengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang 310027, China; ZJU-Hangzhou Global Scientific and Technological Innovation Centre, Hangzhou, Zhejiang 311200, China.
| |
Collapse
|
2
|
Gu Q, Wang Y, Zhang H, Yang W, Meng X, Zhao M. SS-31: A promising therapeutic agent against bleomycin-induced pulmonary fibrosis in Mice. PLoS One 2025; 20:e0315473. [PMID: 40299935 PMCID: PMC12040141 DOI: 10.1371/journal.pone.0315473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 02/26/2025] [Indexed: 05/01/2025] Open
Abstract
OBJECTIVE The aim of this research was to investigate if the mitochondria- targeting peptide SS-31 could serve as a protective measure against bleomycin-induced pulmonary fibrosis in mice. METHOD Mice were split into four groups named CON group, SS-31 group, BLM group, and the BLM + SS-31 group. SS-31 (intraperitoneal injection, 5mg/Kg) was administered daily from the day prior to the experiment for the control and model groups. Mice were euthanized after 28 days of the experiment, following which blood, bronchoalveolar lavage fluid, and lung tissue were collected for analysis. RESULTS BLM caused a large decrease in body weight in mice. However, the intraperitoneal injection of SS-31 slowed down the body weight loss in the mice. It was observed through HE and Masson staining, immunohistochemistry, hydroxyproline detection, and fibrosis index measurement via Western blot that SS-31 could alleviate pulmonary fibrosis caused by BLM. Electron microscopy and ATP detection further suggested that SS-31 might help protect mitochondrial structure and function. It was also found that SS-31 could reduce reactive oxygen species and myeloperoxidase, thereby alleviating the reduction of antioxidant factor MPO and SOD, as well as diminishing the inflammatory factors TNF-α, IL-1 β, and IL-6. CONCLUSION The mitochondria-targeting drug SS-31 exhibited potential in mitigating bleomycin-induced pulmonary fibrosis, improving mitochondrial structural and functional damage, stabilizing the balance between oxidative and antioxidant systems, reducing inflammatory factor expression, and improving apoptosis in lung tissue.
Collapse
Affiliation(s)
- Quankuan Gu
- Department of Critical Care Medicine, the First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
- Heilongjiang Provincial Key Laboratory of Critical Care Medicine, Harbin, Heilongjiang Province, China
| | - Yunlong Wang
- Department of Critical Care Medicine, the First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
- Heilongjiang Provincial Key Laboratory of Critical Care Medicine, Harbin, Heilongjiang Province, China
| | - Haichao Zhang
- Department of Critical Care Medicine, the First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
- Heilongjiang Provincial Key Laboratory of Critical Care Medicine, Harbin, Heilongjiang Province, China
| | - Wei Yang
- Department of Critical Care Medicine, the First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
- Heilongjiang Provincial Key Laboratory of Critical Care Medicine, Harbin, Heilongjiang Province, China
| | - Xianglin Meng
- Department of Critical Care Medicine, the First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
- Heilongjiang Provincial Key Laboratory of Critical Care Medicine, Harbin, Heilongjiang Province, China
| | - Mingyan Zhao
- Department of Critical Care Medicine, the First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
- Heilongjiang Provincial Key Laboratory of Critical Care Medicine, Harbin, Heilongjiang Province, China
| |
Collapse
|
3
|
Ferrini MG, Abraham A, Millán R, Graciano L, Eleswarapu SV, Rajfer J. Nutraceutical COMP-4 confers protection against endothelial dysfunction through the eNOS/iNOS-NO-cGMP pathway. PLoS One 2025; 20:e0316798. [PMID: 39913351 PMCID: PMC11801596 DOI: 10.1371/journal.pone.0316798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 12/17/2024] [Indexed: 02/09/2025] Open
Abstract
The nutraceutical COMP-4 -consisting of L-citrulline, ginger extract, and herbal components Paullinia cupana and muira puama-has been shown previously to stimulate the production of nitric oxide (NO) in a variety of tissue types. We hypothesized that COMP-4 may have a protective, stimulatory effect on the vascular endothelial cell. Human umbilical arterial endothelial cells were incubated for 24 hours with or without COMP-4 and, to replicate impairment of endothelial function, co-incubated with or without H2O2. NO intracellular content, nitrite formation and cGMP content in culture media, nitric oxide synthase (NOS) isoforms and mRNA content, pro-inflammatory cytokines, and PAI-1 expression and activity were measured. COMP-4 increased endothelial cell production of NO and cGMP and the expression of both endothelial NOS (eNOS) and inducible NOS (iNOS), in tandem with a reduction in cytokine expression and activity of PAI-1. Co-incubation of COMP-4 with H2O2 reversed detrimental effects of H2O2 on endothelial function, evidenced by improvement in NO availability and abrogation of the pro-inflammatory milieu. These results suggest that COMP-4 exerts a stimulatory effect on endothelial cell eNOS and iNOS to increase NO bioavailability, leading to a reduction in pro-inflammatory cytokines, particularly the prothrombotic PAI-1.
Collapse
Affiliation(s)
- Monica G. Ferrini
- Department of Health and Life Sciences, Charles R. Drew University of Medicine and Science, Los Angeles, California, United States of America
| | - Andrea Abraham
- Department of Health and Life Sciences, Charles R. Drew University of Medicine and Science, Los Angeles, California, United States of America
| | - Revecca Millán
- Department of Health and Life Sciences, Charles R. Drew University of Medicine and Science, Los Angeles, California, United States of America
| | - Leslie Graciano
- Department of Health and Life Sciences, Charles R. Drew University of Medicine and Science, Los Angeles, California, United States of America
| | - Sriram V. Eleswarapu
- Department of Urology, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Jacob Rajfer
- Department of Urology, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| |
Collapse
|
4
|
Moideen FM, Rahamathulla MP, Charavu R, Alghofaili F, Sha M, Bhandary YP. PAI-1 influences and curcumin destabilizes MMP-2, MMP-9 and basement membrane proteins during lung injury and fibrosis. Int Immunopharmacol 2024; 143:113587. [PMID: 39549545 DOI: 10.1016/j.intimp.2024.113587] [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: 06/07/2024] [Revised: 10/29/2024] [Accepted: 11/05/2024] [Indexed: 11/18/2024]
Abstract
One of the characteristic feature of idiopathic pulmonary fibrosis is an imbalanced fibrinolytic system. Plasminogen activator inhibitor-1 (PAI-1), an essential serine protease in the fibrinolytic system, has an anti-fibrotic tendency in some organs and a pro-fibrotic nature in others. Curcumin is reported to regulate the fibrinolytic system. In this study, we sought to determine how curcumin affected alterations in tissue remodelling mediated by PAI-1 in lung fibrosis. For in vitro studies, NIH3T3 fibroblasts were either exposed to TGF-β or overexpressed with PAI-1, and/or treated with curcumin. For in vivo studies, C57BL/6 mice were either instilled with bleomycin, overexpressed with PAI-1, and/or intervened with curcumin. Protein and gene expression studies were performed by western blotting and RT-PCR techniques, respectively. Curcumin intervention, in vitro and in vivo, could inhibit the the expression of collagen, fibronectin, MMP-2, and MMP-9, which was otherwise elevated by TGF-β or bleomycin. In conclusion, curcumin reduces pulmonary fibrosis by suppressing excessive basement membrane protein deposition and, likely, preventing the thickening of the alveolar septum.
Collapse
Affiliation(s)
- Fathimath Muneesa Moideen
- Yenepoya Research Centre, Yenepoya (Deemed to be University), Deralakatte, Mangalore 575 018, Karnataka, India
| | - Mohamudha Parveen Rahamathulla
- Department of Basic Medical Sciences, College of Medicine, Prince Sattam bin Abdulaziz University, Al Kharj 11942, Saudi Arabia.
| | - Rakshitha Charavu
- Yenepoya Research Centre, Yenepoya (Deemed to be University), Deralakatte, Mangalore 575 018, Karnataka, India
| | - Fayez Alghofaili
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Majmaah 11952, Saudi Arabia
| | - Mohemmed Sha
- Department of Software Engineering, College of Computer Engineering and Sciences, Prince Sattam bin Abdulaziz University, Al Kharj 11942, Saudi Arabia
| | - Yashodhar P Bhandary
- Yenepoya Research Centre, Yenepoya (Deemed to be University), Deralakatte, Mangalore 575 018, Karnataka, India.
| |
Collapse
|
5
|
Cenerini G, Chimera D, Pagnini M, Bazzan E, Conti M, Turato G, Celi A, Neri T. The Intricate Relationship Between Pulmonary Fibrosis and Thrombotic Pathology: A Narrative Review. Cells 2024; 13:2099. [PMID: 39768190 PMCID: PMC11674501 DOI: 10.3390/cells13242099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is associated with a significantly increased risk of thrombotic events and mortality. This review explores the complex bidirectional relationship between pulmonary fibrosis and thrombosis, discussing epidemiological evidence, pathogenetic mechanisms, and therapeutic implications, with a particular focus on the emerging role of extracellular vesicles (EVs) as crucial mediators linking fibrosis and coagulation. Coagulation factors directly promote fibrosis, while fibrosis itself activates thrombotic pathways. Retrospective studies suggest the benefits of anticoagulants in IPF, but prospective trials have faced challenges. Novel anticoagulants, profibrinolytic therapies, and agents targeting protease-activated receptors (PARs) show promise in preclinical studies and early clinical trials. EVs have emerged as key players in the pathogenesis of interstitial lung diseases (ILDs), serving as vehicles for intercellular communication and contributing to both fibrosis and coagulation. EV-based approaches, such as EV modulation, engineered EVs as drug delivery vehicles, and mesenchymal stem cell-derived EVs, represent promising therapeutic strategies. Ongoing research should focus on optimizing risk-benefit profiles, identifying predictive biomarkers, evaluating combination strategies targeting thrombotic, fibrotic, and inflammatory pathways, and advancing the understanding of EVs in ILDs to develop targeted interventions.
Collapse
Affiliation(s)
- Giovanni Cenerini
- UO Pneumologia, Azienda Ospedaliero-Universitaria Pisana, 56124 Pisa, Italy; (G.C.); (D.C.)
| | - Davide Chimera
- UO Pneumologia, Azienda Ospedaliero-Universitaria Pisana, 56124 Pisa, Italy; (G.C.); (D.C.)
| | - Marta Pagnini
- Centro Dipartimentale di Biologia Cellulare Cardiorespiratoria, Dipartimento di Patologia Chirurgica, Medica, Molecolare e dell’Area Critica, Università degli Studi di Pisa, 56124 Pisa, Italy; (M.P.); (T.N.)
| | - Erica Bazzan
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova and Padova City Hospital, 35128 Padova, Italy; (E.B.); (M.C.); (G.T.)
| | - Maria Conti
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova and Padova City Hospital, 35128 Padova, Italy; (E.B.); (M.C.); (G.T.)
- Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy
| | - Graziella Turato
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova and Padova City Hospital, 35128 Padova, Italy; (E.B.); (M.C.); (G.T.)
| | - Alessandro Celi
- Centro Dipartimentale di Biologia Cellulare Cardiorespiratoria, Dipartimento di Patologia Chirurgica, Medica, Molecolare e dell’Area Critica, Università degli Studi di Pisa, 56124 Pisa, Italy; (M.P.); (T.N.)
| | - Tommaso Neri
- Centro Dipartimentale di Biologia Cellulare Cardiorespiratoria, Dipartimento di Patologia Chirurgica, Medica, Molecolare e dell’Area Critica, Università degli Studi di Pisa, 56124 Pisa, Italy; (M.P.); (T.N.)
| |
Collapse
|
6
|
Gawronska-Kozak B, Machcinska-Zielinska S, Walendzik K, Kopcewicz M, Pääkkönen M, Wisniewska J. Hypoxia and Foxn1 alter the proteomic signature of dermal fibroblasts to redirect scarless wound healing to scar-forming skin wound healing in Foxn1 -/- mice. BMC Biol 2024; 22:193. [PMID: 39256768 PMCID: PMC11389453 DOI: 10.1186/s12915-024-01990-2] [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: 07/06/2023] [Accepted: 08/22/2024] [Indexed: 09/12/2024] Open
Abstract
BACKGROUND Foxn1-/- deficient mice are a rare model of regenerative skin wound healing among mammals. In wounded skin, the transcription factor Foxn1 interacting with hypoxia-regulated factors affects re-epithelialization, epithelial-mesenchymal transition (EMT) and dermal white adipose tissue (dWAT) reestablishment and is thus a factor regulating scar-forming/reparative healing. Here, we hypothesized that transcriptional crosstalk between Foxn1 and Hif-1α controls the switch from scarless (regenerative) to scar-present (reparative) skin wound healing. To verify this hypothesis, we examined (i) the effect of hypoxia/normoxia and Foxn1 signalling on the proteomic signature of Foxn1-/- (regenerative) dermal fibroblasts (DFs) and then (ii) explored the effect of Hif-1α or Foxn1/Hif-1α introduced by a lentiviral (LV) delivery vector to injured skin of regenerative Foxn1-/- mice with particular attention to the remodelling phase of healing. RESULTS We showed that hypoxic conditions and Foxn1 stimulation modified the proteome of Foxn1-/- DFs. Hypoxic conditions upregulated DF protein profiles, particularly those related to extracellular matrix (ECM) composition: plasminogen activator inhibitor-1 (Pai-1), Sdc4, Plod2, Plod1, Lox, Loxl2, Itga2, Vldlr, Ftl1, Vegfa, Hmox1, Fth1, and F3. We found that Pai-1 was stimulated by hypoxic conditions in regenerative Foxn1-/- DFs but was released by DFs to the culture media exclusively upon hypoxia and Foxn1 stimulation. We also found higher levels of Pai-1 protein in DFs isolated from Foxn1+/+ mice (reparative/scar-forming) than in DFs isolated from Foxn1-/- (regenerative/scarless) mice and triggered by injury increase in Foxn1 and Pai-1 protein in the skin of mice with active Foxn1 (Foxn1+/+ mice). Then, we demonstrated that the introduction of Foxn1 and Hif-1α via lentiviral injection into the wounded skin of regenerative Foxn1-/- mice activates reparative/scar-forming healing by increasing the wounded skin area and decreasing hyaluronic acid deposition and the collagen type III to I ratio. We also identified a stimulatory effect of LV-Foxn1 + LV-Hif-1α injection in the wounded skin of Foxn1-/- mice on Pai-1 protein levels. CONCLUSIONS The present data highlight the effect of hypoxia and Foxn1 on the protein profile and functionality of regenerative Foxn1-/- DFs and demonstrate that the introduction of Foxn1 and Hif-1α into the wounded skin of regenerative Foxn1-/- mice activates reparative/scar-forming healing.
Collapse
Affiliation(s)
- Barbara Gawronska-Kozak
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Ul. Tuwima 10, 10-748, Olsztyn, Poland.
| | - Sylwia Machcinska-Zielinska
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Ul. Tuwima 10, 10-748, Olsztyn, Poland
| | - Katarzyna Walendzik
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Ul. Tuwima 10, 10-748, Olsztyn, Poland
| | - Marta Kopcewicz
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Ul. Tuwima 10, 10-748, Olsztyn, Poland
| | - Mirva Pääkkönen
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Tykistökatu 6, Biocity 5 Floor, 20520, Turku, Finland
| | - Joanna Wisniewska
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Ul. Tuwima 10, 10-748, Olsztyn, Poland
| |
Collapse
|
7
|
Chauhan S, Jhawat V, Singh RP, Yadav A. Topical delivery of insulin using novel organogel formulations: An approach for the management of diabetic wounds. Burns 2024; 50:1068-1082. [PMID: 38350788 DOI: 10.1016/j.burns.2024.01.008] [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: 01/09/2023] [Revised: 04/06/2023] [Accepted: 01/10/2024] [Indexed: 02/15/2024]
Abstract
Diabetes mellitus is a growing chronic form of diabetes, with lengthy health implications. It is predicted as poor diabetic wound recovery affects roughly 25% of all diabetes mellitus patients, frequently resulting in lower traumatic injury and severe external factors and emotional expenses. The insulin-resistant condition increases biofilm development, making diabetic wounds harder to treat. Nowadays, medical treatment and management of diabetic wounds, which have a significant amputation rate, a high-frequency rate, and a high death rate, have become a global concern. Topical formulations have played a significant part in diabetic wound management and have been developed to achieve a number of features. Because of its significant biocompatibility, moisture retention, and therapeutic qualities, topical insulin has emerged as an appealing and feasible wound healing process effector. With a greater comprehension of the etiology of diabetic wounds, numerous functionalized topical insulins have been described and shown good outcomes in recent years, which has improved some diabetic injuries. The healing of wounds is a physiological phenomenon that restores skin integrity and heals damaged tissues. Insulin, a powerful wound-healing factor, is also used in several experimental and clinical studies accelerate healing of diverse injuries.
Collapse
Affiliation(s)
- Sunita Chauhan
- Department of Pharmaceutical Science, School of Medical and Allied Science, GD Goenka University, Gurugram, Haryana, India
| | - Vikas Jhawat
- Department of Pharmaceutical Science, School of Medical and Allied Science, GD Goenka University, Gurugram, Haryana, India.
| | - Rahul Pratap Singh
- Department of Pharmaceutical Science, School of Medical and Allied Science, GD Goenka University, Gurugram, Haryana, India
| | - Abhishek Yadav
- Department of Pharmaceutical Science, School of Medical and Allied Science, GD Goenka University, Gurugram, Haryana, India
| |
Collapse
|
8
|
Zhao JH, Li S, Du SL, Zhang ZQ. The role of mitochondrial dysfunction in macrophages on SiO 2 -induced pulmonary fibrosis: A review. J Appl Toxicol 2024; 44:86-95. [PMID: 37468209 DOI: 10.1002/jat.4517] [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: 05/03/2023] [Revised: 06/13/2023] [Accepted: 06/29/2023] [Indexed: 07/21/2023]
Abstract
Several epidemiologic and toxicological studies have widely regarded that mitochondrial dysfunction is a popular molecular event in the process of silicosis from different perspectives, but the details have not been systematically summarized yet. Thus, it is necessary to investigate how silica dust leads to pulmonary fibrosis by damaging the mitochondria of macrophages. In this review, we first introduce the molecular mechanisms that silica dust induce mitochondrial morphological and functional abnormalities and then introduce the main molecular mechanisms that silica-damaged mitochondria induce pulmonary fibrosis. Finally, we conclude that the mitochondrial abnormalities of alveolar macrophages caused by silica dust are involved deeply in the pathogenesis of silicosis through these two sequential mechanisms. Therefore, reducing the silica-damaged mitochondria will prevent the potential occurrence and fatality of the disease in the future.
Collapse
Affiliation(s)
- Jia-Hui Zhao
- Weifang Medical University, Weifang, Shandong, China
- Department of Public Health, Jining Medical University, Jining, Shandong, China
| | - Shuang Li
- Department of Public Health, Jining Medical University, Jining, Shandong, China
- Binzhou Medical University, Yantai, Shandong, China
| | - Shu-Ling Du
- Weifang Medical University, Weifang, Shandong, China
- Department of Public Health, Jining Medical University, Jining, Shandong, China
| | - Zhao-Qiang Zhang
- Department of Public Health, Jining Medical University, Jining, Shandong, China
| |
Collapse
|
9
|
van der Schoot GGF, Ormel HL, Westerink NDL, Wempe JB, Lefrandt JD, May AM, Vrieling AH, Meijer C, Gietema JA, Walenkamp AME. Physical exercise in patients with testicular cancer treated with bleomycin, etoposide and cisplatin chemotherapy: pulmonary and vascular endothelial function-an exploratory analysis. J Cancer Res Clin Oncol 2023; 149:17467-17478. [PMID: 37889308 PMCID: PMC10657310 DOI: 10.1007/s00432-023-05469-5] [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: 07/21/2023] [Accepted: 10/09/2023] [Indexed: 10/28/2023]
Abstract
PURPOSE Bleomycin, etoposide, and cisplatin combination chemotherapy (BEP) improves the survival of patients with testicular cancer, but is associated with potentially life-threatening toxicities like pneumonitis and thromboembolic events. This study explored the effects of physical exercise in patients with testicular cancer during or after BEP-chemotherapy on pulmonary and vascular endothelial toxicity. METHODS In this post hoc analysis of a multicenter randomized clinical trial (NCT01642680), patients with metastatic testicular cancer scheduled to receive BEP-chemotherapy were randomized to a 24-week exercise intervention, initiated during (group A) or after BEP-chemotherapy (group B). Endpoints were pulmonary function (forced vital capacity (FVC), forced expiratory volume in one second (FEV1), lung transfer-coefficient and transfer factor for carbon monoxide (KCO, DLCO) and markers of vascular endothelial dysfunction (von Willebrand factor (vWF) and factor VIII). RESULTS Thirty patients were included. Post-chemotherapy, patients declined less in FVC, FEV1 and DLCO in group A compared to group B. Post-chemotherapy, vWF and factor VIII were significantly lower in group A compared to group B. After completion of exercise, started either during BEP-chemotherapy or thereafter, no between-group differences were found. At 1-year post-intervention, significant between-group differences were found in favour of group A in DLCO and KCO. CONCLUSIONS Patients who exercised during BEP-chemotherapy better preserved FVC, FEV1 and DLCO, measured directly post-chemotherapy and 1-year post-intervention (DLCO, KCO). This coincided with less increase in vWF and factor VIII measured directly post-chemotherapy. These data support a beneficial role of a physical exercise intervention during BEP-chemotherapy on pulmonary and vascular damage in patients with testicular cancer. TRIAL REGISTRY Optimal Timing of Physical Activity in Cancer Treatment (ACT) Registry URL: https://clinicaltrials.gov/ct2/show/NCT01642680 . TRIAL REGISTRATION NUMBER NCT01642680.
Collapse
Affiliation(s)
- Gabriela G F van der Schoot
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Harm L Ormel
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Nico-Derk L Westerink
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Johan B Wempe
- Department of Pulmonary Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Joop D Lefrandt
- Department of Vascular Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Anne M May
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Aline H Vrieling
- Department of Rehabilitation Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Coby Meijer
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Jourik A Gietema
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Annemiek M E Walenkamp
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands.
| |
Collapse
|
10
|
Caporarello N, Ligresti G. Vascular Contribution to Lung Repair and Fibrosis. Am J Respir Cell Mol Biol 2023; 69:135-146. [PMID: 37126595 PMCID: PMC10399144 DOI: 10.1165/rcmb.2022-0431tr] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 05/01/2023] [Indexed: 05/03/2023] Open
Abstract
Lungs are constantly exposed to environmental perturbations and therefore have remarkable capacity to regenerate in response to injury. Sustained lung injuries, aging, and increased genomic instability, however, make lungs particularly susceptible to disrepair and fibrosis. Pulmonary fibrosis constitutes a major cause of morbidity and is often relentlessly progressive, leading to death from respiratory failure. The pulmonary vasculature, which is critical for gas exchanges and plays a key role during lung development, repair, and regeneration, becomes aberrantly remodeled in patients with progressive pulmonary fibrosis. Although capillary rarefaction and increased vascular permeability are recognized as distinctive features of fibrotic lungs, the role of vasculature dysfunction in the pathogenesis of pulmonary fibrosis has only recently emerged as an important contributor to the progression of this disease. This review summarizes current findings related to lung vascular repair and regeneration and provides recent insights into the vascular abnormalities associated with the development of persistent lung fibrosis.
Collapse
Affiliation(s)
- Nunzia Caporarello
- Department of Medicine, Stritch School of Medicine, Loyola University Chicago, Chicago, Illinois; and
| | - Giovanni Ligresti
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
| |
Collapse
|
11
|
Ali G, Zhang M, Chang J, Zhao R, Jin Y, Zhang J, Ji HL. PAI-1 regulates AT2-mediated re-alveolarization and ion permeability. Stem Cell Res Ther 2023; 14:185. [PMID: 37501095 PMCID: PMC10375781 DOI: 10.1186/s13287-023-03414-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 07/14/2023] [Indexed: 07/29/2023] Open
Abstract
BACKGROUND Acute lung injury is characterized by overwhelmingly elevated PAI-1 in both lung edema fluid and the circulating system. The role of increased PAI-1, encoded by Serpine1 gene, in the regeneration of injured lung epithelium has not been understood completely. This study aimed to investigate the role of Serpine1 in the regulation of alveolar type 2 epithelial cell (AT2) fate in a humanized mouse line carrying diseased mutants (Serpine1Tg). METHODS Wild-type (wt) and Serpine1Tg AT2 cells were either cultured as monolayers or 3D alveolospheres. Colony-forming assay and total surface area of organoids were analyzed. AT1 and AT2 cells in organoids were counted by immunohistochemistry and fluorescence-activated cell sorting (FACS). To test the potential effects of elevated PAI-1 on the permeability in the epithelial monolayers, we digitized the biophysical properties of polarized AT2 monolayers grown at the air-liquid interface. RESULTS A significant reduction in total AT2 cells harvested in Serpine1Tg mice was observed compared with wt controls. AT2 cells harvested from Serpine1Tg mice reduced significantly over the wt controls. Spheroids formed by Serpine1Tg AT2 cells were lesser than wt control. Similarly, the corresponding surface area, a readout of re-alveolarization of injured epithelium, was markedly reduced in Serpine1Tg organoids. FACS analysis revealed a significant suppression in the number of AT2 cells, in particular, the CD44+ subpopulation, in Serpine1Tg organoids. A lesser ratio of AT1:AT2 cells in Serpine1Tg organoids was observed compared with wt cultures. There was a significant increase in transepithelial resistance but not amiloride inhibition. CONCLUSIONS Our study suggests elevated PAI-1 in injured lungs downregulates alveolar epithelial regeneration by reducing the AT2 self-renewal, particularly in the CD44+ cells.
Collapse
Affiliation(s)
- Gibran Ali
- Department of Cellular and Molecular Biology, Texas Lung Injury Institute, University of Texas at Tyler Health Science Center, Tyler, TX, USA
| | - Mo Zhang
- Department of Cellular and Molecular Biology, Texas Lung Injury Institute, University of Texas at Tyler Health Science Center, Tyler, TX, USA
- Xinxiang Medical University, Xinxiang, Henan, China
| | - Jianjun Chang
- Department of Cellular and Molecular Biology, Texas Lung Injury Institute, University of Texas at Tyler Health Science Center, Tyler, TX, USA
| | - Runzhen Zhao
- Department of Cellular and Molecular Biology, Texas Lung Injury Institute, University of Texas at Tyler Health Science Center, Tyler, TX, USA
- Department of Surgery, Burn and Shock Trauma Research Institute, Loyola University Chicago, 2160 S 1St Avenue, Maywood, IL, 60153, USA
| | - Yang Jin
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University, Boston, MA, USA
| | - Jiwang Zhang
- Department of Cancer Biology, Oncology Institute, Cardinal Bernardin Cancer Center, Loyola University Medical Center, Maywood, IL, USA
| | - Hong-Long Ji
- Department of Cellular and Molecular Biology, Texas Lung Injury Institute, University of Texas at Tyler Health Science Center, Tyler, TX, USA.
- Department of Surgery, Burn and Shock Trauma Research Institute, Loyola University Chicago, 2160 S 1St Avenue, Maywood, IL, 60153, USA.
| |
Collapse
|
12
|
He W, Xi Q, Cui H, Zhang P, Huang R, Wang T, Wang D. Liang-Ge Decoction Ameliorates Coagulation Dysfunction in Cecal Ligation and Puncture-Induced Sepsis Model Rats through Inhibiting PAD4-Dependent Neutrophil Extracellular Trap Formation. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2023; 2023:5042953. [PMID: 37159591 PMCID: PMC10163969 DOI: 10.1155/2023/5042953] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 01/30/2023] [Accepted: 02/14/2023] [Indexed: 05/11/2023]
Abstract
Liang-Ge (LG) decoction could ameliorate coagulation dysfunction in septic model rats. However, the mechanism of LG in treating sepsis still needs to be clarified. Our current study established a septic rat model to evaluate the effect of LG on coagulation dysfunction in septic rats first. Second, we investigated the effect of LG on NET formation in septic rats. Finally, NETs and PAD4 inhibitors were further used to clarify if LG could improve the mechanism of sepsis coagulation dysfunction by inhibiting NET formation. Our findings indicated that treatment with LG improved the survival rate, reduced inflammatory factor levels, enhanced hepatic and renal function, and reduced pathological changes in rats with sepsis. LG could also alleviate coagulation dysfunction in septic model rats. Besides, LG treatment reduced NETs formation and decreased PAD4 expression in neutrophiles. In addition, LG treatment showed a similar result in comparison to the treatment with either NET inhibitors or PAD4 inhibitors alone. In conclusion, this study confirmed that LG has therapeutic effects on septic rats. Furthermore, the improvement of coagulation dysfunction in septic rats by LG was achieved through inhibiting PAD4-mediated NET formation.
Collapse
Affiliation(s)
- Wenju He
- Department of Integration of Traditional Chinese and Western Medicine, First Central Hospital Affiliated to Nankai University, Tianjin First Central Hospital, Tianjin, China
| | - Qiang Xi
- Department of Practice and Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Huantian Cui
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Pingping Zhang
- Department of Integration of Traditional Chinese and Western Medicine, First Central Hospital Affiliated to Nankai University, Tianjin First Central Hospital, Tianjin, China
| | - Rui Huang
- Department of Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Taihuan Wang
- Department of Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Dongqiang Wang
- Department of Integration of Traditional Chinese and Western Medicine, First Central Hospital Affiliated to Nankai University, Tianjin First Central Hospital, Tianjin, China
| |
Collapse
|
13
|
Ali G, Zhang M, Chang J, Zhao R, Jin Y, Ji HL. PAI-1 regulates AT2-mediated re-alveolarization and ion permeability. RESEARCH SQUARE 2023:rs.3.rs-2289657. [PMID: 36909505 PMCID: PMC10002791 DOI: 10.21203/rs.3.rs-2289657/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2023]
Abstract
Background Acute lung injury is characterized by overwhelmingly elevated PAI-1 in both lung edema fluid and the circulating system. The role of increased PAI-1, encoded by Serpine1 gene, in the regeneration of injured lung epithelium has not been understood completely. This study aimed to investigate the role of Serpine1 in the regulation of alveolar type 2 epithelial cell (AT2) fate in a humanized mouse line carrying diseased mutants (Serpine1Tg). Methods Wild type (wt) and Serpine1Tg AT2 cells were either cultured as monolayers or 3D alveolospheres. Colony forming assay and total surface area of organoids were analyzed. AT1 and AT2 cells in organoids were counted by immunohistochemistry and fluorescence-activated cell sorting (FACS). To test the potential effects of elevated PAI-1 on the permeability in the epithelial monolayers, we digitized the biophysical properties of polarized AT2 monolayers grown at the air-liquid interface. Results A significant reduction in total AT2 cells harvested in Serpine1Tg mice was observed compared with wt controls. AT2 cells harvested from Serpine1Tg mice reduced significantly over the wt controls. Spheroids formed by Serpine1Tg AT2 cells were lesser than wt control. Similarly, the corresponding surface area, a readout of realveolarization of injured epithelium, was markedly reduced in Serpine1Tg organoids. FACS analysis revealed a significant suppression in the number of AT2 cells, in particular, the CD44+ subpopulation, in Serpine1Tg organoids. A lesser ratio of AT1:AT2 cells in Serpine1Tg organoids was observed compared with wt cultures. There was a significant increase in transepithelial resistance but not amiloride inhibition. Conclusions Our study suggests elevated PAI-1 in injured lungs downregulates alveolar epithelial regeneration by reducing the AT2 self-renewal, particularly in the CD44+ cells.
Collapse
Affiliation(s)
- Gibran Ali
- Department of Cellular and Molecular Biology, University of Texas at Tyler Health Science Center, Tyler, TX, USA
| | - Mo Zhang
- Department of Cellular and Molecular Biology, University of Texas at Tyler Health Science Center, Tyler, TX, USA
- Xinxiang Medical University, Xinxiang, Henan, China
| | - Jianjun Chang
- Department of Cellular and Molecular Biology, University of Texas at Tyler Health Science Center, Tyler, TX, USA
| | - Runzhen Zhao
- Department of Cellular and Molecular Biology, University of Texas at Tyler Health Science Center, Tyler, TX, USA
| | - Yang Jin
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University, Boston, MA, USA
| | - Hong-Long Ji
- Department of Cellular and Molecular Biology, University of Texas at Tyler Health Science Center, Tyler, TX, USA
- Texas Lung Injury Institute, University of Texas at Tyler Health Science Center, Tyler, TX, USA
- Corresponding Author: Dr. Hong-Long (James) Ji, M.D., Ph.D., , University of Texas at Tyler Health Science Center, 11937 US Highway 271, Tyler, TX 75708-3154, USA
| |
Collapse
|
14
|
Plasminogen activator inhibitor 1 is not a major causative factor for exacerbation in a mouse model of SARS-CoV-2 infection. Sci Rep 2023; 13:3103. [PMID: 36813909 PMCID: PMC9944779 DOI: 10.1038/s41598-023-30305-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 02/21/2023] [Indexed: 02/24/2023] Open
Abstract
Coronavirus disease (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) remains a global pandemic. Although several vaccines targeting SARS-CoV-2 spike proteins protect against COVID-19 infection, mutations affecting virus transmissibility and immune evasion potential have reduced their efficacy, leading to the need for a more efficient strategy. Available clinical evidence regarding COVID-19 suggests that endothelial dysfunction with thrombosis is a central pathogenesis of progression to systemic disease, in which overexpression of plasminogen activator inhibitor-1 (PAI-1) may be important. Here we developed a novel peptide vaccine against PAI-1 and evaluated its effect on lipopolysaccharide (LPS)-induced sepsis and SARS-CoV-2 infection in mice. Administration of LPS and mouse-adapted SARS-CoV-2 increased serum PAI-1 levels, although the latter showed smaller levels. In an LPS-induced sepsis model, mice immunized with PAI-1 vaccine showed reduced organ damage and microvascular thrombosis and improved survival compared with vehicle-treated mice. In plasma clot lysis assays, vaccination-induced serum IgG antibodies were fibrinolytic. However, in a SARS-CoV-2 infection model, survival and symptom severity (i.e., body weight reduction) did not differ between vaccine- and vehicle-treated groups. These results indicate that although PAI-1 may promote the severity of sepsis by increasing thrombus formation, it might not be a major contributor to COVID-19 exacerbation.
Collapse
|
15
|
Yang Y, Wang Q, Li G, Guo W, Yang Z, Liu H, Deng X. Cysteine-Derived Chiral Carbon Quantum Dots: A Fibrinolytic Activity Regulator for Plasmin to Target the Human Islet Amyloid Polypeptide for Type 2 Diabetes Mellitus. ACS APPLIED MATERIALS & INTERFACES 2023; 15:2617-2629. [PMID: 36596222 DOI: 10.1021/acsami.2c17975] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
The fibrillization and deposition of the human islet amyloid polypeptide (hIAPP) are the pathological hallmark of type 2 diabetes mellitus (T2DM), and these insoluble fibrotic depositions of hIAPP are considered to strongly affect insulin secretion by inducing toxicity toward pancreatic islet β-cells. The current strategy of preventing amyloid aggregation by nanoparticle-assisted inhibitors can only disassemble fibrotic amyloids into more toxic oligomers and/or protofibrils. Herein, for the first time, we propose a type of cysteine-derived chiral carbon quantum dot (CQD) that targets plasmin, a core natural fibrinolytic protease in humans. These CQDs can serve as fibrinolytic activity regulators for plasmin to cleave hIAPP into nontoxic polypeptides or into even smaller amino acid fragments, thus alleviating hIAPP's fibrotic amyloid-induced cytotoxicity. Our experiments indicate that chiral CQDs have opposing effects on plasmin activity. The l-CQDs promote the cleavage of hIAPP by enhancing plasmin activity at a promotion ratio of 23.2%, thus protecting β-cells from amyloid-induced toxicity. In contrast, the resultant d-CQDs significantly inhibit proteolysis, decreasing plasmin activity by 31.5% under the same reaction conditions. Second harmonic generation (SHG) microscopic imaging is initially used to dynamically characterize hIAPP before and after proteolysis. The l-CQD promotion of plasmin activity thus provides a promising avenue for the hIAPP-targeted treatment of T2DM to treat low fibrinolytic activity, while the d-CQDs, as inhibitors of plasmin activity, may improve patient survival for hyperfibrinolytic conditions, such as those existing during surgeries and traumas.
Collapse
Affiliation(s)
- Yongzhen Yang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou510631, China
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou510631, China
- Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou510631, China
| | - Qin Wang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou510631, China
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou510631, China
- Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou510631, China
| | - Gongjian Li
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou510631, China
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou510631, China
- Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou510631, China
| | - Wenjing Guo
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou510530, China
| | - Zuojun Yang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou510631, China
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou510631, China
- Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou510631, China
| | - Hao Liu
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou510631, China
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou510631, China
- Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou510631, China
| | - Xiaoyuan Deng
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou510631, China
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou510631, China
- Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou510631, China
| |
Collapse
|
16
|
Distinguishing Plasmin-Generating Microvesicles: Tiny Messengers Involved in Fibrinolysis and Proteolysis. Int J Mol Sci 2023; 24:ijms24021571. [PMID: 36675082 PMCID: PMC9860915 DOI: 10.3390/ijms24021571] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/09/2023] [Accepted: 01/10/2023] [Indexed: 01/15/2023] Open
Abstract
A number of stressors and inflammatory mediators (cytokines, proteases, oxidative stress mediators) released during inflammation or ischemia stimulate and activate cells in blood, the vessel wall or tissues. The most well-known functional and phenotypic responses of activated cells are (1) the immediate expression and/or release of stored or newly synthesized bioactive molecules, and (2) membrane blebbing followed by release of microvesicles. An ultimate response, namely the formation of extracellular traps by neutrophils (NETs), is outside the scope of this work. The main objective of this article is to provide an overview on the mechanism of plasminogen reception and activation at the surface of cell-derived microvesicles, new actors in fibrinolysis and proteolysis. The role of microvesicle-bound plasmin in pathological settings involving inflammation, atherosclerosis, angiogenesis, and tumour growth, remains to be investigated. Further studies are necessary to determine if profibrinolytic microvesicles are involved in a finely regulated equilibrium with pro-coagulant microvesicles, which ensures a balanced haemostasis, leading to the maintenance of vascular patency.
Collapse
|
17
|
Role of Oxidative Stress in Peyronie's Disease: Biochemical Evidence and Experiences of Treatment with Antioxidants. Int J Mol Sci 2022; 23:ijms232415969. [PMID: 36555611 PMCID: PMC9781573 DOI: 10.3390/ijms232415969] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 11/29/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Peyronie's disease (PD) is a chronic inflammatory condition affecting adult males, involving the tunica albuginea of the corpora cavernosa of the penis. PD is frequently associated with penile pain, erectile dysfunction, and a secondary anxious-depressive state. The etiology of PD has not yet been completely elucidated, but local injury is generally recognized to be a triggering factor. It has also been widely proven that oxidative stress is an essential, decisive component in all inflammatory processes, whether acute or chronic. Current conservative medical treatment comprises oral substances, penile injections, and physical therapy. AIM This article intends to show how antioxidant therapy is able to interfere with the pathogenetic mechanisms of the disease. METHOD This article consists of a synthetic narrative review of the current scientific literature on antioxidant therapy for this disease. RESULTS The good results of the antioxidant treatment described above also prove that the doses used were adequate and the concentrations of the substances employed did not exceed the threshold at which they might have interacted negatively with the mechanisms of the redox regulation of tissue. CONCLUSIONS We believe new, randomized, controlled studies are needed to confirm the efficacy of treatment with antioxidants. However, we consider the experiences of antioxidant treatment which can already be found in the literature useful for the clinical practice of urologists in the treatment of this chronic inflammatory disease.
Collapse
|
18
|
Liu J, Fan G, Tao N, Feng F, Meng C, Sun T. Ginsenoside Rb1 Alleviates Bleomycin-Induced Pulmonary Inflammation and Fibrosis by Suppressing Central Nucleotide-Binding Oligomerization-, Leucine-Rich Repeat-, and Pyrin Domains-Containing Protein Three Inflammasome Activation and the NF-κB Pathway. Drug Des Devel Ther 2022; 16:1793-1809. [PMID: 35719213 PMCID: PMC9205635 DOI: 10.2147/dddt.s361748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 06/08/2022] [Indexed: 11/23/2022] Open
Abstract
Purpose Idiopathic pulmonary fibrosis is a chronic and irreversible fibrotic interstitial pneumonia of unknown etiology and therapeutic strategies are limited. Emerging evidence suggests that the continuous activation of the central nucleotide-binding oligomerization-, leucine-rich repeat-, and pyrin domain-containing protein 3 (NLRP3) inflammasome is involved in the pathogenesis of pulmonary fibrosis. Ginsenoside Rb1 (G-Rb1) is the most abundant component in the traditional Chinese herb ginseng and has anti-inflammatory and anti-fibrotic activities. The purpose of this study was to explore whether G-Rb1 exerts anti-inflammatory and anti-fibrotic activities in vivo and in vitro by suppressing the activation of the NLRP3 inflammasome and NF-κB pathway. Methods Forty-eight male C57BL/6 mice were randomly divided into four groups (n=12/group) as follows: control, bleomycin (BLM), BLM/G-Rb1, and G-Rb1. A pulmonary fibrosis model was developed via an intratracheal injection of BLM. Six mice from each group were euthanized on days 3 and 21. The degree of pulmonary fibrosis was examined by histological evaluation and assessing α-smooth muscle actin levels. THP-1 cells were differentiated into macrophages, and stimulated by lipopolysaccharide and adenosine triphosphate. Activation of the NLRP3 inflammasome and NF-κB pathway was determined by Western blotting. Interleukin-1 beta and interleukin-18 levels were measured by ELISA. MRC-5 cells were cultured in the conditioned medium of the treated macrophages, after which markers of myofibroblasts were determined by Western blotting. Results G-Rb1 ameliorated BLM-induced pulmonary inflammation and fibrosis in mice, and suppressed NLRP3 inflammasome activation and the NF-κB pathway in lung tissues. Moreover, interleukin-1 beta secreted after NLRP3 inflammasome activation in macrophages promoted fibroblast differentiation. G-Rb1 inhibited lipopolysaccharide- and adenosine triphosphate-induced NLRP3 inflammasome activation in macrophages and disturbed the crosstalk between macrophages and fibroblasts. Conclusion G-Rb1 ameliorates BLM-induced pulmonary inflammation and fibrosis by suppressing NLRP3 inflammasome activation and the NF-κB pathway. Hence, G-Rb1 is a potential novel therapeutic drug for idiopathic pulmonary fibrosis.
Collapse
Affiliation(s)
- Jingjing Liu
- Department of Respiratory Medicine and Critical Care, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
- Graduate School of Peking Union Medical College, Beijing, People’s Republic of China
- The MOH Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing, People’s Republic of China
| | - Guoqing Fan
- Department of Respiratory Medicine and Critical Care, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
- Graduate School of Peking Union Medical College, Beijing, People’s Republic of China
- The MOH Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing, People’s Republic of China
| | - Ningning Tao
- Department of Respiratory & Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People’s Republic of China
| | - Feifei Feng
- Department of Respiratory & Critical Care Medicine, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People’s Republic of China
| | - Chao Meng
- Department of Respiratory Medicine and Critical Care, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
- Graduate School of Peking Union Medical College, Beijing, People’s Republic of China
- The MOH Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing, People’s Republic of China
| | - Tieying Sun
- Department of Respiratory Medicine and Critical Care, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
- Graduate School of Peking Union Medical College, Beijing, People’s Republic of China
| |
Collapse
|
19
|
Fan M, Xiao H, Song D, Zhu L, Zhang J, Zhang X, Wang J, Dai H, Wang C. A Novel N-Arylpyridone Compound Alleviates the Inflammatory and Fibrotic Reaction of Silicosis by Inhibiting the ASK1-p38 Pathway and Regulating Macrophage Polarization. Front Pharmacol 2022; 13:848435. [PMID: 35401236 PMCID: PMC8983992 DOI: 10.3389/fphar.2022.848435] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 03/03/2022] [Indexed: 11/21/2022] Open
Abstract
Silicosis is one of the potentially fatal occupational diseases characterized by respiratory dysfunction, chronic interstitial inflammation, and fibrosis, for which treatment options are limited. Previous studies showed that a novel N-arylpyridone compound named AKEX0011 exhibited anti-inflammatory and anti-fibrotic effects in bleomycin-induced pulmonary fibrosis; however, it is unknown whether it could also be effective against silicosis. Therefore, we sought to investigate the preventive and therapeutic roles of AKEX0011 in a silicosis rodent model and in a silica-stimulated macrophage cell line. In vivo, our results showed that AKEX0011 ameliorated silica-induced imaging lung damages, respiratory dysfunction, reduced the secretion of inflammatory and fibrotic factors (TNF-α, IL-1β, IL-6, TGF-β, IL-4, and IL-10), and the deposition of fibrosis-related proteins (collagen I, fibronectin, and α-SMA), regardless of early or advanced therapy. Specifically, we found that AKEX0011 attenuated silicosis by inhibiting apoptosis, blocking the ASK1-p38 MAPK signaling pathway, and regulating polarization of macrophages. In vitro, AKEX0011 inhibited macrophages from secreting inflammatory cytokines and inhibited apoptosis of macrophages in pre-treated and post-treated models, concurrent with blocking the ASK1-p38 pathway and inhibiting M1 polarization. Collectively, AKEX0011, as a novel N-arylpyridone compound, exerted protective effects for silica-induced pulmonary inflammation and fibrosis both in vivo and in vitro, and hence, it could be a strong drug candidate for the treatment of silicosis.
Collapse
Affiliation(s)
- Mingming Fan
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Jilin, China,Department of Pulmonary and Critical Care Medicine Center of Respiratory Medicine, China-Japan Friendship Hospital, Capital Medical University, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Huijuan Xiao
- Department of Pulmonary and Critical Care Medicine Center of Respiratory Medicine, China-Japan Friendship Hospital, Capital Medical University, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China,Department of Pulmonary and Critical Care Medicine, China-Japan Friendship School of Clinical Medicine, Peking University, Beijing, China
| | - Dingyun Song
- Department of Pulmonary and Critical Care Medicine Center of Respiratory Medicine, China-Japan Friendship Hospital, Capital Medical University, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Lili Zhu
- Department of Pulmonary and Critical Care Medicine Center of Respiratory Medicine, China-Japan Friendship Hospital, Capital Medical University, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Jie Zhang
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Jilin, China
| | - Xinran Zhang
- Department of Pulmonary and Critical Care Medicine Center of Respiratory Medicine, China-Japan Friendship Hospital, Capital Medical University, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China,Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Jing Wang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China,*Correspondence: Huaping Dai, ; Jing Wang, ; Chen Wang,
| | - Huaping Dai
- Department of Pulmonary and Critical Care Medicine Center of Respiratory Medicine, China-Japan Friendship Hospital, Capital Medical University, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China,*Correspondence: Huaping Dai, ; Jing Wang, ; Chen Wang,
| | - Chen Wang
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Jilin, China,Department of Pulmonary and Critical Care Medicine Center of Respiratory Medicine, China-Japan Friendship Hospital, Capital Medical University, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China,State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China,*Correspondence: Huaping Dai, ; Jing Wang, ; Chen Wang,
| |
Collapse
|
20
|
Embedded Human Periodontal Ligament Stem Cells Spheroids Enhance Cementogenic Differentiation via Plasminogen Activator Inhibitor 1. Int J Mol Sci 2022; 23:ijms23042340. [PMID: 35216454 PMCID: PMC8878532 DOI: 10.3390/ijms23042340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 02/18/2022] [Accepted: 02/19/2022] [Indexed: 11/17/2022] Open
Abstract
Spheroids reproduce the tissue structure that is found in vivo more accurately than classic two-dimensional (2D) monolayer cultures. We cultured human periodontal ligament stem cells (HPLSCs) as spheroids that were embedded in collagen gel to examine whether their cementogenic differentiation could be enhanced by treatment with recombinant human plasminogen activator inhibitor-1 (rhPAI-1). The upregulated expression of cementum protein 1 (CEMP1) and cementum attachment protein (CAP), established cementoblast markers, was observed in the 2D monolayer HPLSCs that were treated with rhPAI-1 for 3 weeks compared with that in the control and osteogenic-induction medium groups. In the embedded HPLSC spheroids, rhPAI-1 treatment induced interplay between the spheroids and collagenous extracellular matrix (ECM), indicating that disaggregated HPLSCs migrated and spread into the surrounding ECM 72 h after three-dimensional (3D) culture. Western blot and immunocytochemistry analyses showed that the CEMP1 expression levels were significantly upregulated in the rhPAI-1-treated embedded HPLSC spheroids compared with all the 2D monolayer HPLSCs groups and the 3D spheroid groups. Therefore, 3D collagen-embedded spheroid culture in combination with rhPAI-1 treatment may be useful for facilitating cementogenic differentiation of HPLSCs.
Collapse
|
21
|
Ma Y, Meng C, Weng L. Association between trauma exposure and respiratory disease-A Mendelian randomization study. Front Endocrinol (Lausanne) 2022; 13:1001223. [PMID: 36133309 PMCID: PMC9483852 DOI: 10.3389/fendo.2022.1001223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 08/16/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Trauma is a well-known risk factor for many disease, but the effect of trauma on respiratory disease is unclarified. In the present study, we aimed to evaluate the association between trauma and respiratory disease. METHOD Using both United Kingdom biobank and Finnish biobank genome-wide association study data (GWAS), we performed a two-sample Mendelian randomization (MR) analysis to evaluate the relationship between trauma and respiratory disease. We used four methods including inverse-variance weighted (IVW), weighted median, Maximum likelihood, and MR-Egger in this MR analysis. The IVW MR was selected as the main method. We also performed multivariable Mendelian randomization (MVMR) to simultaneously assess the independent impact of trauma exposure on respiratory disease. RESULTS In the main two-sample MR analysis, trauma exposure was significantly associated with increased risk of respiratory disease (OR 1.15, 95%CI: 1.05-1.25). Besides, there was no heterogeneity and horizontal pleiotropy observed in the sensitivity analysis. After adjusting for pack years of smoking and body mass index (BMI), trauma exposure retained its association with respiratory disease (OR, 1.13, 95%CI, 1.04-1.23 adjusted by pack years of smoking; and OR, 1.11, 95%CI, 1.04-1.18 adjusted by BMI). CONCLUSION Our study discovered the association between trauma exposure and the increased risk of respiratory disease, suggesting the prevention and treatment with trauma to reduce the risk of respiratory disease.
Collapse
Affiliation(s)
- Yuchao Ma
- Department of Cardiothoracic Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| | - Changjiang Meng
- Clinical Research Center, Third Xiangya Hospital, Central South University, Changsha, China
| | - Liang Weng
- Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Molecular Radiation Oncology Hunan Province, Xiangya Hospital, Central South University, Changsha, China
- Hunan International Science and Technology Collaboration Base of Precision Medicine for Cancer, Xiangya Hospital, Central South University, Changsha, China
- Institute of Gerontological Cancer Research, National Clinical Research Center for Gerontology, Changsha, China
- Xiangya Lung Cancer Center, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Liang Weng,
| |
Collapse
|
22
|
Jäger B, Seeliger B, Terwolbeck O, Warnecke G, Welte T, Müller M, Bode C, Prasse A. The NLRP3-Inflammasome-Caspase-1 Pathway Is Upregulated in Idiopathic Pulmonary Fibrosis and Acute Exacerbations and Is Inducible by Apoptotic A549 Cells. Front Immunol 2021; 12:642855. [PMID: 33968032 PMCID: PMC8104027 DOI: 10.3389/fimmu.2021.642855] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 04/06/2021] [Indexed: 12/15/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a relentlessly progressive disease harboring significant morbidity and mortality despite recent advances in therapy. Regardless of disease severity acute exacerbations (IPF-AEs) may occur leading to considerable loss of function and are the leading cause of death in IPF. Histologic features of IPF-AE are very similar to acute respiratory distress syndrome (ARDS), but the underlying mechanisms are incompletely understood. We investigated the role of the NLRP3 inflammasome in IPF and IPF-AE. Bronchoalveolar lavage (BAL) cells were sampled from patients with IPF (n = 32), IPF-AE (n = 10), ARDS (n = 7) and healthy volunteers (HV, n = 37) and the NLRP3-inflammasome was stimulated in-vitro. We found the NLRP3 inflammasome to be hyper-inducible in IPF compared to HV with increased IL-1ß and pro-IL-1ß levels on ELISA upon stimulation as well as increased caspase-1 activity measured by caspase-1p20 immunoblotting. In IPF-AE, IL-1ß was massively elevated to an extent similar to ARDS. To evaluate potential mechanisms, we co-cultured BAL cells with radiated A549 cells (a model to simulate apoptotic alveolar epithelial cells), which led to increased NLRP3 mRNA expression and increased caspase-1 dependent IL-1ß production. In the presence of a reactive oxygen species (ROS) inhibitor (diphenyleneiodonium) and a cathepsin B inhibitor (E64D), NLRP3 expression was suppressed indicating that induction of NLRP3 activation following efferocytosis of apoptotic A549 cells is mediated via ROS and cathepsin-B. In summary, we present evidence of involvement of the NLRP3 inflammasome-caspase pathway in the pathogenesis of IPF-AE, similarly to ARDS, which may be mediated by efferocytosis of apoptotic alveolar epithelial cells in IPF.
Collapse
Affiliation(s)
- Benedikt Jäger
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany.,Department of Respiratory Medicine, University Medical Center, Freiburg, Germany.,Faculty of Biology, Albert Ludwig University, Freiburg, Germany
| | - Benjamin Seeliger
- Department of Respiratory Medicine, Hannover Medical School and Biomedical Research in End-stage and Obstructive Lung Disease (BREATH), German Center for Lung Research (DZL), Hannover, Germany
| | - Oliver Terwolbeck
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
| | - Gregor Warnecke
- Department of Heart, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Tobias Welte
- Department of Respiratory Medicine, Hannover Medical School and Biomedical Research in End-stage and Obstructive Lung Disease (BREATH), German Center for Lung Research (DZL), Hannover, Germany
| | - Meike Müller
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
| | - Christian Bode
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Antje Prasse
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany.,Department of Respiratory Medicine, University Medical Center, Freiburg, Germany.,Department of Respiratory Medicine, Hannover Medical School and Biomedical Research in End-stage and Obstructive Lung Disease (BREATH), German Center for Lung Research (DZL), Hannover, Germany
| |
Collapse
|
23
|
Adamcakova J, Mokra D. New Insights into Pathomechanisms and Treatment Possibilities for Lung Silicosis. Int J Mol Sci 2021; 22:ijms22084162. [PMID: 33920534 PMCID: PMC8072896 DOI: 10.3390/ijms22084162] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/14/2021] [Accepted: 04/14/2021] [Indexed: 02/06/2023] Open
Abstract
Inhalation of silica particles is an environmental and occupational cause of silicosis, a type of pneumoconiosis. Development of the lung silicosis is a unique process in which the vicious cycle of ingestion of inhaled silica particles by alveolar macrophages and their release triggers inflammation, generation of nodular lesions, and irreversible fibrosis. The pathophysiology of silicosis is complex, and interactions between the pathomechanisms have not been completely understood. However, elucidation of silica-induced inflammation cascades and inflammation-fibrosis relations has uncovered several novel possibilities of therapeutic targeting. This article reviews new information on the pathophysiology of silicosis and points out several promising treatment approaches targeting silicosis-related pathways.
Collapse
|
24
|
Sillen M, Declerck PJ. A Narrative Review on Plasminogen Activator Inhibitor-1 and Its (Patho)Physiological Role: To Target or Not to Target? Int J Mol Sci 2021; 22:ijms22052721. [PMID: 33800359 PMCID: PMC7962805 DOI: 10.3390/ijms22052721] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 02/28/2021] [Accepted: 03/03/2021] [Indexed: 02/06/2023] Open
Abstract
Plasminogen activator inhibitor-1 (PAI-1) is the main physiological inhibitor of plasminogen activators (PAs) and is therefore an important inhibitor of the plasminogen/plasmin system. Being the fast-acting inhibitor of tissue-type PA (tPA), PAI-1 primarily attenuates fibrinolysis. Through inhibition of urokinase-type PA (uPA) and interaction with biological ligands such as vitronectin and cell-surface receptors, the function of PAI-1 extends to pericellular proteolysis, tissue remodeling and other processes including cell migration. This review aims at providing a general overview of the properties of PAI-1 and the role it plays in many biological processes and touches upon the possible use of PAI-1 inhibitors as therapeutics.
Collapse
|
25
|
Bayram B, Owen AR, Dudakovic A, Bettencourt JW, Limberg AK, Morrey ME, Sanchez-Sotelo J, Berry DJ, Kocher JP, van Wijnen AJ, Abdel MP. Elevated Expression of Plasminogen Activator Inhibitor (PAI-1/SERPINE1) is Independent from rs1799889 Genotypes in Arthrofibrosis. Meta Gene 2021; 28. [PMID: 33816122 DOI: 10.1016/j.mgene.2021.100877] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Arthrofibrosis is characterized by excessive extracellular matrix deposition in patients with total knee arthroplasties (TKAs) and causes undesirable joint stiffness. The pathogenesis of arthrofibrosis remains elusive and currently there are no diagnostic biomarkers for the pathological formation of this connective tissue. Fibrotic soft tissues are known to have elevated levels of plasminogen activator inhibitor-1 (PAI-1) (encoded by SERPINE1), a secreted serine protease inhibitor that moderates extracellular matrix remodeling and tissue homeostasis. The 4G/5G insertion/deletion (rs1799889) is a well-known SERPINE1 polymorphism that directly modulates PAI-1 levels. Homozygous 4G/4G allele carriers typically have higher PAI-1 levels and may predispose patients to soft tissue fibrosis (e.g., liver, lung, and kidney). Here, we examined the genetic contribution of the SERPINE1 rs1799889 polymorphism to musculoskeletal fibrosis in arthrofibrotic (n = 100) and non-arthrofibrotic (n = 100) patients using Sanger Sequencing. Statistical analyses revealed that the allele frequencies of the SERPINE1 rs1799889 polymorphism are similar in arthrofibrotic and non-arthrofibrotic patient cohorts. Because the fibrosis related SERPINE1 rs1799889 polymorphism is independent of arthrofibrosis susceptibility in TKA patients, the possibility arises that fibrosis of joint connective tissues may involve unique genetic determinants distinct from those linked to classical soft tissue fibrosis.
Collapse
Affiliation(s)
- Banu Bayram
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, United States
| | - Aaron R Owen
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, United States
| | - Amel Dudakovic
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, United States.,Department of Biochemistry & Molecular Biology, Mayo Clinic, Rochester, MN, United States
| | | | - Afton K Limberg
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, United States
| | - Mark E Morrey
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, United States
| | | | - Daniel J Berry
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, United States
| | - Jean-Pierre Kocher
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, United States
| | - Andre J van Wijnen
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, United States.,Department of Biochemistry & Molecular Biology, Mayo Clinic, Rochester, MN, United States
| | - Matthew P Abdel
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
26
|
Kwak SY, Park S, Kim H, Lee SJ, Jang WS, Kim MJ, Lee S, Jang WI, Kim AR, Kim EH, Shim S, Jang H. Atorvastatin Inhibits Endothelial PAI-1-Mediated Monocyte Migration and Alleviates Radiation-Induced Enteropathy. Int J Mol Sci 2021; 22:ijms22041828. [PMID: 33673196 PMCID: PMC7917640 DOI: 10.3390/ijms22041828] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 02/08/2021] [Accepted: 02/09/2021] [Indexed: 01/15/2023] Open
Abstract
Intestinal injury is observed in cancer patients after radiotherapy and in individuals exposed to radiation after a nuclear accident. Radiation disrupts normal vascular homeostasis in the gastrointestinal system by inducing endothelial damage and senescence. Despite advances in medical technology, the toxicity of radiation to healthy tissue remains an issue. To address this issue, we investigated the effect of atorvastatin, a commonly prescribed hydroxy-3-methylglutaryl-coenzyme A reductase inhibitor of cholesterol synthesis, on radiation-induced enteropathy and inflammatory responses. We selected atorvastatin based on its pleiotropic anti-fibrotic and anti-inflammatory effects. We found that atorvastatin mitigated radiation-induced endothelial damage by regulating plasminogen activator inhibitor-1 (PAI-1) using human umbilical vein endothelial cells (HUVECs) and mouse model. PAI-1 secreted by HUVECs contributed to endothelial dysfunction and trans-endothelial monocyte migration after radiation exposure. We observed that PAI-1 production and secretion was inhibited by atorvastatin in irradiated HUVECs and radiation-induced enteropathy mouse model. More specifically, atorvastatin inhibited PAI-1 production following radiation through the JNK/c-Jun signaling pathway. Together, our findings suggest that atorvastatin alleviates radiation-induced enteropathy and supports the investigation of atorvastatin as a radio-mitigator in patients receiving radiotherapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Sehwan Shim
- Correspondence: (S.S.); (H.J.); Tel.: +82-2-3399-5873 (S.S.); +82-2-970-1302 (H.J.)
| | - Hyosun Jang
- Correspondence: (S.S.); (H.J.); Tel.: +82-2-3399-5873 (S.S.); +82-2-970-1302 (H.J.)
| |
Collapse
|
27
|
Role of various imbalances centered on alveolar epithelial cell/fibroblast apoptosis imbalance in the pathogenesis of idiopathic pulmonary fibrosis. Chin Med J (Engl) 2021; 134:261-274. [PMID: 33522725 PMCID: PMC7846426 DOI: 10.1097/cm9.0000000000001288] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
There have been recent extensive studies and rapid advancement on the pathogenesis underlying idiopathic pulmonary fibrosis (IPF), and intricate pathogenesis of IPF has been suggested. The purpose of this study was to clarify the logical relationship between these mechanisms. An extensive search was undertaken of the PubMed using the following keywords: “etiology,” “pathogenesis,” “alveolar epithelial cell (AEC),” “fibroblast,” “lymphocyte,” “macrophage,” “epigenomics,” “histone,” acetylation,” “methylation,” “endoplasmic reticulum stress,” “mitochondrial dysfunction,” “telomerase,” “proteases,” “plasminogen,” “epithelial-mesenchymal transition,” “oxidative stress,” “inflammation,” “apoptosis,” and “idiopathic pulmonary fibrosis.” This search covered relevant research articles published up to April 30, 2020. Original articles, reviews, and other articles were searched and reviewed for content; 240 highly relevant studies were obtained after screening. IPF is likely the result of complex interactions between environmental, genetic, and epigenetic factors: environmental exposures affect epigenetic marks; epigenetic processes translate environmental exposures into the regulation of chromatin; epigenetic processes shape gene expression profiles; in turn, an individual's genetic background determines epigenetic marks; finally, these genetic and epigenetic factors act in concert to dysregulate gene expression in IPF lung tissue. The pathogenesis of IPF involves various imbalances including endoplasmic reticulum, telomere length homeostasis, mitochondrial dysfunction, oxidant/antioxidant imbalance, Th1/Th2 imbalance, M1–M2 polarization of macrophages, protease/antiprotease imbalance, and plasminogen activation/inhibition imbalance. These affect each other, promote each other, and ultimately promote AEC/fibroblast apoptosis imbalance directly or indirectly. Excessive AEC apoptosis and impaired apoptosis of fibroblasts contribute to fibrosis. IPF is likely the result of complex interactions between environmental, genetic, and epigenetic factors. The pathogenesis of IPF involves various imbalances centered on AEC/fibroblast apoptosis imbalance.
Collapse
|
28
|
Wani K, AlHarthi H, Alghamdi A, Sabico S, Al-Daghri NM. Role of NLRP3 Inflammasome Activation in Obesity-Mediated Metabolic Disorders. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:E511. [PMID: 33435142 PMCID: PMC7826517 DOI: 10.3390/ijerph18020511] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 12/22/2020] [Accepted: 01/06/2021] [Indexed: 12/12/2022]
Abstract
NLRP3 inflammasome is one of the multimeric protein complexes of the nucleotide-binding domain, leucine-rich repeat (NLR)-containing pyrin and HIN domain family (PYHIN). When activated, NLRP3 inflammasome triggers the release of pro-inflammatory interleukins (IL)-1β and IL-18, an essential step in innate immune response; however, defective checkpoints in inflammasome activation may lead to autoimmune, autoinflammatory, and metabolic disorders. Among the consequences of NLRP3 inflammasome activation is systemic chronic low-grade inflammation, a cardinal feature of obesity and insulin resistance. Understanding the mechanisms involved in the regulation of NLRP3 inflammasome in adipose tissue may help in the development of specific inhibitors for the treatment and prevention of obesity-mediated metabolic diseases. In this narrative review, the current understanding of NLRP3 inflammasome activation and regulation is highlighted, including its putative roles in adipose tissue dysfunction and insulin resistance. Specific inhibitors of NLRP3 inflammasome activation which can potentially be used to treat metabolic disorders are also discussed.
Collapse
Affiliation(s)
| | | | | | | | - Nasser M. Al-Daghri
- Biochemistry Department, College of Science, King Saud University, Riyadh 11451, Saudi Arabia; (K.W.); (H.A.); (A.A.); (S.S.)
| |
Collapse
|
29
|
Cheng Y, Liu B, Qian H, Yang H, Wang Y, Wu Y, Shen F. BAY11-7082 inhibits the expression of tissue factor and plasminogen activator inhibitor-1 in type-II alveolar epithelial cells following TNF-α stimulation via the NF-κB pathway. Exp Ther Med 2020; 21:177. [PMID: 33552241 DOI: 10.3892/etm.2020.9608] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 11/17/2020] [Indexed: 12/26/2022] Open
Abstract
Pulmonary inflammation strongly promotes alveolar hypercoagulation and fibrinolytic inhibition. NF-κB signaling regulates the expression of molecules associated with coagulation and fibrinolytic inhibition in type-II alveolar epithelial cells (AECII) stimulated by lipopolysaccharide. However, whether TNF-α-induced alveolar hypercoagulation and fibrinolysis inhibition is also associated with the NF-κB pathway remains to be determined. The aim of the present study was to determine whether BAY11-7082, an inhibitor of the NF-κB pathway, inhibits the expressions of tissue factor (TF) and plasminogen activator inhibitor-1 (PAI-1) in AECⅡ in response to TNF-α. Rat AECII were treated with BAY11-7082 for 24 h and stimulated with TNF-α for 1 h. The expression of TF and PAI-1 were determined using western blotting and reverse transcription-quantitative PCR. The concentrations of TF and PAI-1 in culture supernatant were also measured by ELISA. Moreover, levels of NF-κB p65 (p65), phosphorylated (p)-p65 (p-p65), inhibitor of NF-κB α (IκBα) and p-IκBα were also evaluated. Immunofluorescence was used to detect p65 levels in cell nuclei. TNF-α significantly promoted TF and PAI-1 expression either at the mRNA or protein level in AECII cells. Concentrations of TF and PAI-1 in supernatant also significantly increased upon TNF-α stimulation. Furthermore, TNF-α upregulated the levels of p-IκBα, p65, and p-p65 in the cytoplasm. Immunofluorescence analysis indicated that TNF-α increased p65 translocation from the cytoplasm to the nucleus. However, AECII pre-treated with BAY11-7082 expressed lower levels of TF and PAI-1 following TNF-α treatment. Levels of p-IκBα, p65 and p-p65 in the cytoplasm also decreased, and translocation of p65 from cytoplasm into the nucleus was inhibited by BAY11-7082 pretreatment. These findings suggest that BAY11-7082 improves the hypercoagulation and fibrinolytic inhibition induced by TNF-α in alveolar epithelial cells via the NF-κB signaling pathway. BAY11-7082 might represent a therapeutic option for alveolar hypercoagulation and fibrinolytic inhibition in acute respiratory distress syndrome.
Collapse
Affiliation(s)
- Yumei Cheng
- Department of Critical Care Medicine, Guizhou Medical University Affiliated Hospital, Guiyang, Guizhou 550001, P.R. China
| | - Bo Liu
- Department of Critical Care Medicine, Guizhou Medical University Affiliated Hospital, Guiyang, Guizhou 550001, P.R. China
| | - Hong Qian
- Department of Critical Care Medicine, Guizhou Medical University Affiliated Hospital, Guiyang, Guizhou 550001, P.R. China
| | - Huilin Yang
- Department of Critical Care Medicine, Guizhou Medical University Affiliated Hospital, Guiyang, Guizhou 550001, P.R. China
| | - Yahui Wang
- Department of Critical Care Medicine, Guizhou Medical University Affiliated Hospital, Guiyang, Guizhou 550001, P.R. China
| | - Yanqi Wu
- Department of Critical Care Medicine, Guizhou Medical University Affiliated Hospital, Guiyang, Guizhou 550001, P.R. China
| | - Feng Shen
- Department of Critical Care Medicine, Guizhou Medical University Affiliated Hospital, Guiyang, Guizhou 550001, P.R. China
| |
Collapse
|
30
|
Deng P, Wang C, Wahlang B, Sexton T, Morris AJ, Hennig B. Co-exposure to PCB126 and PFOS increases biomarkers associated with cardiovascular disease risk and liver injury in mice. Toxicol Appl Pharmacol 2020; 409:115301. [PMID: 33096110 DOI: 10.1016/j.taap.2020.115301] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 09/08/2020] [Accepted: 10/18/2020] [Indexed: 02/07/2023]
Abstract
Polychlorinated biphenyl (PCB)126 and perfluorooctane sulfonic acid (PFOS) are halogenated organic pollutants of high concern. Exposure to these chemicals is ubiquitous, and can lead to potential synergistic adverse effects in individuals exposed to both classes of chemicals. The present study was designed to identify interactions between PCB126 and PFOS that might promote acute changes in inflammatory pathways associated with cardiovascular disease and liver injury. Male C57BL/6 mice were exposed to vehicle, PCB126, PFOS, or a mixture of both pollutants. Plasma and liver samples were collected at 48 h after exposure. Changes in the expression of hepatic genes involved in oxidative stress, inflammation, and atherosclerosis were investigated. Plasma and liver samples was analyzed using untargeted lipidomic method. Hepatic mRNA levels for Nqo1, Icam1, and PAI1 were significantly increased in the mixture-exposed mice. Plasma levels of PAI1, a marker of fibrosis and thrombosis, were also significantly elevated in the mixture-exposed group. Liver injury was observed only in the mixture-exposed mice. Lipidomic analysis revealed that co-exposure to the mixture enhanced hepatic lipid accumulation and elevated oxidized phospholipids levels. In summary, this study shows that acute co-exposure to PCB126 and PFOS in mice results in liver injury and increased cardiovascular disease risk.
Collapse
Affiliation(s)
- Pan Deng
- Superfund Research Center, University of Kentucky, Lexington, KY 40536, USA; Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536, USA
| | - Chunyan Wang
- Superfund Research Center, University of Kentucky, Lexington, KY 40536, USA
| | - Banrida Wahlang
- Superfund Research Center, University of Louisville, Louisville, KY 40202, USA
| | - Travis Sexton
- Division of Cardiovascular Medicine, The Gill Heart and Vascular Institute, College of Medicine, University of Kentucky, and Lexington Veterans Affairs Medical Center, Lexington, KY, 40536, USA
| | - Andrew J Morris
- Superfund Research Center, University of Kentucky, Lexington, KY 40536, USA; Division of Cardiovascular Medicine, The Gill Heart and Vascular Institute, College of Medicine, University of Kentucky, and Lexington Veterans Affairs Medical Center, Lexington, KY, 40536, USA
| | - Bernhard Hennig
- Superfund Research Center, University of Kentucky, Lexington, KY 40536, USA; Department of Animal and Food Sciences, College of Agriculture, Food and Environment, University of Kentucky, Lexington, KY 40536, USA.
| |
Collapse
|
31
|
Malek Mahdavi A. A brief review of interplay between vitamin D and angiotensin-converting enzyme 2: Implications for a potential treatment for COVID-19. Rev Med Virol 2020; 30:e2119. [PMID: 32584474 PMCID: PMC7362103 DOI: 10.1002/rmv.2119] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/03/2020] [Accepted: 05/07/2020] [Indexed: 01/08/2023]
Abstract
The novel coronavirus disease 2019 (COVID-19) is rapidly expanding and causing many deaths all over the world with the World Health Organization (WHO) declaring a pandemic in March 2020. Current therapeutic options are limited and there is no registered and/or definite treatment or vaccine for this disease or the causative infection, severe acute respiratory coronavirus 2 syndrome (SARS-CoV-2). Angiotensin-converting enzyme 2 (ACE2), a part of the renin-angiotensin system (RAS), serves as the major entry point into cells for SARS-CoV-2 which attaches to human ACE2, thereby reducing the expression of ACE2 and causing lung injury and pneumonia. Vitamin D, a fat-soluble-vitamin, is a negative endocrine RAS modulator and inhibits renin expression and generation. It can induce ACE2/Ang-(1-7)/MasR axis activity and inhibits renin and the ACE/Ang II/AT1R axis, thereby increasing expression and concentration of ACE2, MasR and Ang-(1-7) and having a potential protective role against acute lung injury (ALI)/acute respiratory distress syndrome (ARDS). Therefore, targeting the unbalanced RAS and ACE2 down-regulation with vitamin D in SARS-CoV-2 infection is a potential therapeutic approach to combat COVID-19 and induced ARDS.
Collapse
MESH Headings
- Acute Lung Injury/pathology
- Acute Lung Injury/prevention & control
- Acute Lung Injury/virology
- Angiotensin I/genetics
- Angiotensin I/metabolism
- Angiotensin-Converting Enzyme 2
- Angiotensin-Converting Enzyme Inhibitors/therapeutic use
- Betacoronavirus/genetics
- Betacoronavirus/metabolism
- Betacoronavirus/pathogenicity
- COVID-19
- Coronavirus Infections/drug therapy
- Coronavirus Infections/pathology
- Coronavirus Infections/virology
- Gene Expression Regulation/drug effects
- Humans
- Pandemics
- Peptide Fragments/genetics
- Peptide Fragments/metabolism
- Peptidyl-Dipeptidase A/genetics
- Peptidyl-Dipeptidase A/metabolism
- Pneumonia, Viral/drug therapy
- Pneumonia, Viral/pathology
- Pneumonia, Viral/virology
- Protein Binding
- Proto-Oncogene Mas
- Receptor, Angiotensin, Type 1/genetics
- Receptor, Angiotensin, Type 1/metabolism
- Receptors, Virus/antagonists & inhibitors
- Receptors, Virus/genetics
- Receptors, Virus/metabolism
- Renin-Angiotensin System/drug effects
- SARS-CoV-2
- Severity of Illness Index
- Spike Glycoprotein, Coronavirus/genetics
- Spike Glycoprotein, Coronavirus/metabolism
- Vitamin D/therapeutic use
Collapse
Affiliation(s)
- Aida Malek Mahdavi
- Connective Tissue Diseases Research CenterTabriz University of Medical SciencesTabrizIran
| |
Collapse
|
32
|
Murphy EJ, Masterson C, Rezoagli E, O'Toole D, Major I, Stack GD, Lynch M, Laffey JG, Rowan NJ. β-Glucan extracts from the same edible shiitake mushroom Lentinus edodes produce differential in-vitro immunomodulatory and pulmonary cytoprotective effects - Implications for coronavirus disease (COVID-19) immunotherapies. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 732:139330. [PMID: 32413619 PMCID: PMC7211630 DOI: 10.1016/j.scitotenv.2020.139330] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/07/2020] [Accepted: 05/08/2020] [Indexed: 05/17/2023]
Abstract
Coronavirus pneumonia is accompanied by rapid virus replication, where a large number of inflammatory cell infiltration and cytokine storm may lead to acute lung injury, acute respiratory distress syndrome (ARDS) and death. The uncontrolled release of pro-inflammatory cytokines, including interleukin (IL)-1β and IL-6, is associated with ARDS. This constituted the first study to report on the variability in physicochemical properties of β-glucans extracts from the same edible mushroom Lentinus edodes on the reduction of these pro-inflammatory cytokines and oxidative stress. Specifically, the impact on the immunomodulatory and cytoprotective properties of our novel in 'house' (IH-Lentinan, IHL) and a commercial (Carbosynth-Lentinan, CL) Lentinan extract were investigated using in vitro models of lung injury and macrophage phagocytosis. CL comprised higher amounts of α-glucans and correspondingly less β-glucans. The two lentinan extracts demonstrated varying immunomodulatory activities. Both Lentinan extracts reduced cytokine-induced NF-κB activation in human alveolar epithelial A549 cells, with the IHL extract proving more effective at lower doses. In contrast, in activated THP-1 derived macrophages, the CL extract more effectively attenuated pro-inflammatory cytokine production (TNF-α, IL-8, IL-2, IL-6, IL-22) as well as TGF-β and IL-10. The CL extract attenuated oxidative stress-induced early apoptosis, while the IHL extract attenuated late apoptosis. Our findings demonstrate significant physicochemical differences between Lentinan extracts, which produce differential in vitro immunomodulatory and pulmonary cytoprotective effects that may also have positive relevance to candidate COVID-19 therapeutics targeting cytokine storm.
Collapse
Affiliation(s)
- Emma J Murphy
- Bioscience Research Institute, Athlone Institute of Technology, Athlone, Ireland.
| | - Claire Masterson
- Lung Biology Group, Regenerative Medicine Institute at CURAM Centre for Medical Devices, National University of Ireland Galway, Galway, Ireland
| | - Emanuele Rezoagli
- Lung Biology Group, Regenerative Medicine Institute at CURAM Centre for Medical Devices, National University of Ireland Galway, Galway, Ireland; Anaesthesia and Intensive Care Medicine, University Hospital Galway, Galway, Ireland; Department of Medicine and Surgery, University of Milano-Bicocca, Monza (MB), Italy
| | - Daniel O'Toole
- Lung Biology Group, Regenerative Medicine Institute at CURAM Centre for Medical Devices, National University of Ireland Galway, Galway, Ireland
| | - Ian Major
- Materials Research Institute, Athlone Institute of Technology, Athlone, Ireland
| | - Gary D Stack
- Bioscience Research Institute, Athlone Institute of Technology, Athlone, Ireland
| | - Mark Lynch
- Bioscience Research Institute, Athlone Institute of Technology, Athlone, Ireland
| | - John G Laffey
- Lung Biology Group, Regenerative Medicine Institute at CURAM Centre for Medical Devices, National University of Ireland Galway, Galway, Ireland; Anaesthesia and Intensive Care Medicine, University Hospital Galway, Galway, Ireland
| | - Neil J Rowan
- Bioscience Research Institute, Athlone Institute of Technology, Athlone, Ireland; Centre for Disinfection, Sterilization and Biosecurity, Athlone Institute of Technology, Athlone, Ireland
| |
Collapse
|
33
|
Diagnostic and Prognostic Biomarkers for Chronic Fibrosing Interstitial Lung Diseases With a Progressive Phenotype. Chest 2020; 158:646-659. [PMID: 32268131 DOI: 10.1016/j.chest.2020.03.037] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 03/05/2020] [Accepted: 03/19/2020] [Indexed: 12/19/2022] Open
Abstract
Biomarkers have the potential to become central to the clinical evaluation and monitoring of patients with chronic fibrosing interstitial lung diseases (ILDs) with a progressive phenotype. Here we summarize the current understanding of putative serum, BAL fluid, and genetic biomarkers in this setting, according to their hypothesized pathobiologic mechanisms: evidence of epithelial cell dysfunction (eg, Krebs von den Lungen-6 antigen), fibroblast proliferation and extracellular matrix production or turnover (eg, matrix metalloproteinase-1), or immune dysregulation (eg, CC chemokine ligand 18). While most of the available data come from idiopathic pulmonary fibrosis (IPF), the prototypic progressive fibrosing ILD, data are available in the broader patient population of chronic fibrosing ILDs. A number of these biomarkers show promise, however, none have been validated. In this review article, we assess both the status of proposed biomarkers for chronic fibrosing lung diseases with a progressive phenotype in predicting disease risk or predisposition, diagnosis, prognosis, and treatment response and provide a direct comparison between IPF and other chronic fibrotic ILDs. We also reflect on the current clinical usefulness and future direction of research for biomarkers in the setting of chronic fibrosing ILDs with a progressive phenotype.
Collapse
|
34
|
Janssen-Heininger Y, Reynaert NL, van der Vliet A, Anathy V. Endoplasmic reticulum stress and glutathione therapeutics in chronic lung diseases. Redox Biol 2020; 33:101516. [PMID: 32249209 PMCID: PMC7251249 DOI: 10.1016/j.redox.2020.101516] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 03/20/2020] [Accepted: 03/20/2020] [Indexed: 02/07/2023] Open
Affiliation(s)
- Yvonne Janssen-Heininger
- Department of Pathology and Laboratory Medicine, University of Vermont, Larner College of Medicine, Burlington, VT, 05405, USA.
| | - Niki L Reynaert
- Department of Respiratory Medicine and School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center, Maastricht, the Netherlands
| | - Albert van der Vliet
- Department of Pathology and Laboratory Medicine, University of Vermont, Larner College of Medicine, Burlington, VT, 05405, USA
| | - Vikas Anathy
- Department of Pathology and Laboratory Medicine, University of Vermont, Larner College of Medicine, Burlington, VT, 05405, USA
| |
Collapse
|
35
|
Govoni M, Bassi M, Vezzoli S, Lucci G, Emirova A, Nandeuil MA, Petruzzelli S, Jellema GL, Afolabi EK, Colgan B, Leaker B, Kornmann O, Beeh KM, Watz H, Singh D. Sputum and blood transcriptomics characterisation of the inhaled PDE4 inhibitor CHF6001 on top of triple therapy in patients with chronic bronchitis. Respir Res 2020; 21:72. [PMID: 32197620 PMCID: PMC7085203 DOI: 10.1186/s12931-020-1329-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 02/27/2020] [Indexed: 02/06/2023] Open
Abstract
Background Although phosphodiesterase-4 (PDE4) inhibitors have been shown to reduce COPD exacerbation rate, their biological mechanism of action is not completely elucidated at the molecular level. We aimed to characterise the whole genome gene expression profile of the inhaled PDE4-inhibitor CHF6001 on top of triple therapy in sputum cells and whole blood of patients with COPD and chronic bronchitis. Methods Whole genome gene expression analysis was carried out by microarray in 54 patients before and after 32 days treatment with CHF6001 800 and 1600 μg and placebo twice daily (BID) in a randomised crossover study. Results CHF6001 had a strong effect in sputum, with 1471 and 2598 significantly differentially-expressed probe-sets relative to placebo (p-adjusted for False Discovery Rate < 0.05) with 800 and 1600 μg BID, respectively. Functional enrichment analysis showed significant modulation of key inflammatory pathways involved in cytokine activity, pathogen-associated-pattern-recognition activity, oxidative stress and vitamin D with associated inhibition of downstream inflammatory effectors. A large number of pro-inflammatory genes coding for cytokines and matrix-metalloproteinases were significantly differentially expressed for both doses; the majority (> 87%) were downregulated, including macrophage inflammatory protein-1-alpha and 1-beta, interleukin-27-beta, interleukin-12-beta, interleukin-32, tumour necrosis factor-alpha-induced-protein-8, ligand-superfamily-member-15, and matrix-metalloproteinases-7,12 and 14. The effect in blood was not significant. Conclusions Inhaled PDE4 inhibition by CHF6001 on top of triple therapy in patients with COPD and chronic bronchitis significantly modulated key inflammatory targets and pathways in the lung but not in blood. Mechanistically these findings support a targeted effect in the lung while minimising unwanted systemic class-effects. Trial registration ClinicalTrial.gov, EudraCT, 2015–005550-35. Registered 15 July 2016.
Collapse
Affiliation(s)
- Mirco Govoni
- Global Clinical Development, Personalised Medicine and Biomarkers, Chiesi, Parma, Italy.
| | - Michele Bassi
- Global Clinical Development, Personalised Medicine and Biomarkers, Chiesi, Parma, Italy
| | - Stefano Vezzoli
- Global Clinical Development, Personalised Medicine and Biomarkers, Chiesi, Parma, Italy
| | - Germano Lucci
- Global Clinical Development, Personalised Medicine and Biomarkers, Chiesi, Parma, Italy
| | - Aida Emirova
- Global Clinical Development, Personalised Medicine and Biomarkers, Chiesi, Parma, Italy
| | - Marie Anna Nandeuil
- Global Clinical Development, Personalised Medicine and Biomarkers, Chiesi, Parma, Italy
| | - Stefano Petruzzelli
- Global Clinical Development, Personalised Medicine and Biomarkers, Chiesi, Parma, Italy
| | | | | | | | | | - Oliver Kornmann
- IKF Pneumologie Frankfurt, Clinical Research Centre Respiratory Diseases, Frankfurt, Germany
| | | | - Henrik Watz
- Pulmonary Research Institute at Lung Clinic Grosshansdorf, Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Grosshansdorf, Germany
| | - Dave Singh
- Medicines Evaluation Unit, The University of Manchester, Manchester University NHS Foundation Trust, Manchester, UK
| |
Collapse
|
36
|
Head BM, Mao R, Keynan Y, Rueda ZV. Inflammatory mediators and lung abnormalities in HIV: A systematic review. PLoS One 2019; 14:e0226347. [PMID: 31830103 PMCID: PMC6907827 DOI: 10.1371/journal.pone.0226347] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 11/25/2019] [Indexed: 12/16/2022] Open
Abstract
HIV and pneumonia infections have both been shown to negatively impact lung function. However, evidence of the role of inflammation on lung dysfunction in HIV and pneumonia co-infected individuals remains limited. We aimed to systematically review the association of inflammatory markers and lung abnormalities in HIV and pneumonia co-infected individuals. This systematic review was registered with the International Prospective Register of Systematic Reviews on August 15, 2017 (registration number CRD42017069254) and used 4 databases (Cochrane Central Register of Controlled Trials, PubMed Central, Clinical Trials.gov and Google Scholar). All clinical trial, observational, and comparative studies targeting adult (> 18 years old) populations with HIV, pneumonia, or both, that report on immune response (cytokine, chemokine, or biomarker), and lung abnormality as an outcome were eligible. Data selection, risk of bias and extraction were performed independently by 2 blinded reviewers. Due to heterogeneity among the articles, a qualitative synthesis was performed. Our search strategy identified 4454 articles of which, 7 met our inclusion criteria. All of the studies investigated the ability of circulating biomarkers to predict lung damage in HIV. None of the articles included patients with both HIV and pneumonia, nor pneumonia alone. Markers of inflammation (IL-6, TNF-α, CRP), innate defense (cathelicidin), monocyte and macrophage activation (sCD14, sCD163 and, IL-2sRα), endothelial dysfunction (ET-1) and general immune health (CD4/CD8 ratio) were associated with lung abnormalities in HIV. This review highlights the lack of available information regarding the impact of inflammatory mediators on lung function in HIV and pneumonia populations, therefore opportunities to prevent lung damage with available anti-inflammatory treatment or to investigate new ones still remain.
Collapse
Affiliation(s)
- Breanne M. Head
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Ruochen Mao
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Yoav Keynan
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Zulma Vanessa Rueda
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
- Facultad de Medicina, Universidad Pontificia Bolivariana, Medellín, Antioquia, Colombia
| |
Collapse
|
37
|
Cui Y, Jiang L, Yu R, Shao Y, Mei L, Tao Y. β-carboline alkaloids attenuate bleomycin induced pulmonary fibrosis in mice through inhibiting NF-kb/p65 phosphorylation and epithelial-mesenchymal transition. JOURNAL OF ETHNOPHARMACOLOGY 2019; 243:112096. [PMID: 31323300 DOI: 10.1016/j.jep.2019.112096] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Revised: 07/06/2019] [Accepted: 07/15/2019] [Indexed: 06/10/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The plant Arenaria kansuensis is used in traditional medicine to treat lung inflammation for a long time. However, the anti-pulmonary fibrosis effect and its corresponding bioactive constituents of this plant have not been studied extensively. AIM OF THE STUDY The purpose of this study was to investigate the anti-pulmonary fibrosis effect and its corresponding bioactive constituents of A. kansuensis and its possible mechanism. MATERIALS AND METHODS In vivo experiment, the anti-pulmonary fibrosis effects of the fraction (Part1) enriched from ethyl acetate extracts of the whole plant A. kansuensis were evaluated through bleomycin (BLM)-induced pulmonary fibrosis mice (five groups, n = 10) daily at doses of 50, 100 and 150 mg/kg for 15 days. In vitro experiment, the anti-inflammation and reversed epithelial-mesenchymal transition (EMT) effect of 12 β-carboline alkaloids isolated from Part1 were evaluated through lipopolysaccharide (LPS)-induced RAW264.7 inflammatory cell model and TGF-β1 induced A549 cell model. RESULTS In this study, a fraction named Part1 extracted from Arenaria kansuensis presented strong anti-pulmonary fibrosis effect at the dose of 150 mg/kg. Vivo experiments showed that the survival rate and body weight of mice significantly increased after Part1 treatment. Part1 could significantly inhibit the initial of inflammation, deposition of collagen and expression of TGF-β1 and α-SMA, moreover, the expression of E-cadherin was significantly elevated after administration of Part1. All the cure effects of Part1 were in dose dependent manner. A total of 12 β-carboline alkaloids were identified in Part1 and they all showed suppressive effect on inflammatory cytokines including MCP-1, TNF-α, IL-6 and IL-1β through inhibition of NF-kb/p65 phosphorylation, and that epithelial-mesenchymal transition (EMT) process was reversed by different compounds in different levels. The expression of indicators of EMT including α-SMA, vimentin and E-cadherin was significantly improved after given different β-carboline alkaloids. CONCLUSIONS This study showed that antifibrogenic effect of β-carboline alkaloids was due to inhibiting the initial of inflammation through NF-kb/p65 pathway and reversing the process of EMT.
Collapse
Affiliation(s)
- Yulei Cui
- Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, 810008, Qinghai, PR China; Key Laboratory of Tibetan Medicine Research, Chinese Academy of Sciences, Xining, 810008, Qinghai, PR China; Key Laboratory of Tibetan Medicine Research of Qinghai Province, Xining, 810008, Qinghai, PR China; University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Lei Jiang
- Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, 810008, Qinghai, PR China; Key Laboratory of Tibetan Medicine Research, Chinese Academy of Sciences, Xining, 810008, Qinghai, PR China; Key Laboratory of Tibetan Medicine Research of Qinghai Province, Xining, 810008, Qinghai, PR China
| | - Ruitao Yu
- Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, 810008, Qinghai, PR China; Key Laboratory of Tibetan Medicine Research, Chinese Academy of Sciences, Xining, 810008, Qinghai, PR China; Key Laboratory of Tibetan Medicine Research of Qinghai Province, Xining, 810008, Qinghai, PR China
| | - Yun Shao
- Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, 810008, Qinghai, PR China; Key Laboratory of Tibetan Medicine Research, Chinese Academy of Sciences, Xining, 810008, Qinghai, PR China; Key Laboratory of Tibetan Medicine Research of Qinghai Province, Xining, 810008, Qinghai, PR China
| | - Lijuan Mei
- Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, 810008, Qinghai, PR China; Key Laboratory of Tibetan Medicine Research, Chinese Academy of Sciences, Xining, 810008, Qinghai, PR China; Key Laboratory of Tibetan Medicine Research of Qinghai Province, Xining, 810008, Qinghai, PR China.
| | - Yanduo Tao
- Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, 810008, Qinghai, PR China; Key Laboratory of Tibetan Medicine Research, Chinese Academy of Sciences, Xining, 810008, Qinghai, PR China; Key Laboratory of Tibetan Medicine Research of Qinghai Province, Xining, 810008, Qinghai, PR China.
| |
Collapse
|
38
|
Deng P, Hoffman JB, Petriello MC, Wang CY, Li XS, Kraemer MP, Morris AJ, Hennig B. Dietary inulin decreases circulating ceramides by suppressing neutral sphingomyelinase expression and activity in mice. J Lipid Res 2019; 61:45-53. [PMID: 31604806 DOI: 10.1194/jlr.ra119000346] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 10/09/2019] [Indexed: 01/26/2023] Open
Abstract
Elevated circulating levels of ceramides (Cers) are associated with increased risk of cardiometabolic diseases, and Cers may play a causative role in metabolic dysfunction that precedes cardiac events, such as mortality as a result of coronary artery disease. Although the mechanisms involved are likely complex, these associations suggest that lowering circulating Cer levels could be protective against cardiovascular diseases. Conversely, dietary fibers, such as inulin, have been reported to promote cardiovascular and metabolic health. However, the mechanisms involved in these protective processes also are not well understood. We studied the effects of inulin on lipid metabolism with a model of atherosclerosis in LDL receptor-deficient mice using lipidomics and transcriptomics. Plasma and tissues were collected at 10 days and/or 12 weeks after feeding mice an atherogenic diet supplemented with inulin or cellulose (control). Compared with controls, inulin-fed mice displayed a decreased C16:0/C24:0 plasma Cer ratio and lower levels of circulating Cers associated with VLDL and LDL. Liver transcriptomic analysis revealed that Smpd3, a gene that encodes neutral SMase (NSMase), was downregulated by 2-fold in inulin-fed mice. Hepatic NSMase activity was 3-fold lower in inulin-fed mice than in controls. Furthermore, liver redox status and compositions of phosphatidylserine and FFA species, the major factors that determine NSMase activity, were also modified by inulin. Taken together, these results showed that, in mice, inulin can decrease plasma Cer levels through reductions in NSMase expression and activity, suggesting a mechanism by which fiber could reduce cardiometabolic disease risk.
Collapse
Affiliation(s)
- Pan Deng
- Superfund Research Center, University of Kentucky, Lexington, KY 40536; Department of Animal and Food Sciences, College of Agriculture, Food, and Environment, University of Kentucky, Lexington, KY 40536
| | - Jessie B Hoffman
- Superfund Research Center, University of Kentucky, Lexington, KY 40536; Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536
| | - Michael C Petriello
- Superfund Research Center, University of Kentucky, Lexington, KY 40536; Division of Cardiovascular Medicine, College of Medicine and Lexington Veterans Affairs Medical Center, University of Kentucky, Lexington, KY 40536
| | - Chun-Yan Wang
- Superfund Research Center, University of Kentucky, Lexington, KY 40536; Department of Animal and Food Sciences, College of Agriculture, Food, and Environment, University of Kentucky, Lexington, KY 40536
| | - Xu-Sheng Li
- Superfund Research Center, University of Kentucky, Lexington, KY 40536; Department of Food Science and Engineering, Jinan University, Guangzhou, China 510632
| | - Maria P Kraemer
- Division of Cardiovascular Medicine, College of Medicine and Lexington Veterans Affairs Medical Center, University of Kentucky, Lexington, KY 40536
| | - Andrew J Morris
- Superfund Research Center, University of Kentucky, Lexington, KY 40536; Division of Cardiovascular Medicine, College of Medicine and Lexington Veterans Affairs Medical Center, University of Kentucky, Lexington, KY 40536
| | - Bernhard Hennig
- Superfund Research Center, University of Kentucky, Lexington, KY 40536; Department of Animal and Food Sciences, College of Agriculture, Food, and Environment, University of Kentucky, Lexington, KY 40536.
| |
Collapse
|
39
|
Yi Y, Liu Y, Wu K, Wu W, Zhang W. The core genes involved in the promotion of depression in patients with ovarian cancer. Oncol Lett 2019; 18:5995-6007. [PMID: 31788074 PMCID: PMC6865084 DOI: 10.3892/ol.2019.10934] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 08/08/2019] [Indexed: 12/09/2022] Open
Abstract
The present study aimed to identify the core genes and pathways involved in depression in patients with ovarian cancer (OC) who suffer from high or low-grade depression. The dataset GSE9116 from Gene Expression Omnibus database was analyzed to identify differentially expressed genes (DEGs) in these patients. To elucidate how certain genes could promote depression in patients with OC, pathway crosstalk, protein-protein interaction (PPI) and comprehensive gene-pathway analyses were determined using WebGestalt, ToppGene and Search Tool for the Retrieval of Interacting Genes and gene ontology analysis. Key genes and pathways were extracted from the gene-pathway network, and gene expression and survival analysis were evaluated. A total of 93 DEGs were identified from GSE9116 dataset, including 84 upregulated genes and nine downregulated genes. The PPI, pathway crosstalk and comprehensive gene-pathway analyses highlighted C-C motif chemokine ligand 2 (CCL2), Fos proto-oncogene, AP-1 transcription factor subunit (FOS), serpin family E member 1 (SERPINE1) and serpin family G member 1 (SERPING1) as core genes involved in the promotion of depression in patients with OC. These core genes were involved in the following four pathways 'Ensemble of genes encoding ECM-associated proteins including ECM-affiliated proteins', 'ECM regulators and secreted factors', 'Ensemble of genes encoding extracellular matrix and extracellular matrix-associated proteins' and 'MAPK signaling pathway and IL-17 signaling pathway'. The results from gene expression and survival analysis demonstrated that these four key genes were upregulated in patients with OC and high-grade depression and could worsen patients' survival. These results suggested that CCL2, FOS, SERPINE1 and SERPING1 may serve a crucial role in the promotion of depression in patients with OC. This finding may provide novel markers for predicting and treating depression in patients with OC; however, the underlying mechanisms remain unknown and require further investigation.
Collapse
Affiliation(s)
- Yuexiong Yi
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Yanyan Liu
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Kejia Wu
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Wanrong Wu
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Wei Zhang
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| |
Collapse
|
40
|
Liu B, Wang Y, Wu Y, Cheng Y, Qian H, Yang H, Shen F. IKKβ regulates the expression of coagulation and fibrinolysis factors through the NF-κB canonical pathway in LPS-stimulated alveolar epithelial cells type II. Exp Ther Med 2019; 18:2859-2866. [PMID: 31572531 PMCID: PMC6755483 DOI: 10.3892/etm.2019.7928] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 07/12/2019] [Indexed: 01/11/2023] Open
Abstract
Aim: Hypercoagulation and fibrinolysis inhibition in the alveolar cavity are important characteristics in acute respiratory distress syndrome (ARDS). Alveolar epithelial cells type II (AEC II) have been confirmed to have significant role in regulating alveolar hypercoagulation and fibrinolysis inhibition, but the mechanism is unknown. Nuclear factor-κB (NF-κB) signaling pathway has been demonstrated to participate in the pathogenesis of these two abnormalities in ARDS. The purpose of the present study is to explore whether controlling the upstream crucial factor IκB kinase (IKK)β could regulate coagulation and fibrinolysis factors in LPS-stimulated AEC II. Materials and methods: An IKKβ gene regulation model (IKKβ+/+ and IKKβ−/−) was prepared using lentiviral vector transfection. The models with wild type cells were all stimulated by lipopolysaccharide (LPS) or saline for 24 h. Expression of the related proteins were determined by western-blotting, ELISA and revere transcription-PCR respectively. Tissue factor (TF) procoagulant activity and nuclear p65 protein level were also detected. Results: IKKβ increased in IKKβ+/+ cells but decreased in IKKβ−/− cells. LPS stimulation promoted the expression of p-IκBα, p65, p-p65 and p-IKKβ as well as TF and plasminogen activator inhibitor (PAI)-1, at the mRNA or protein level, and this was significantly enhanced by IKKβ upregulation but weakened by IKKβ downregulation. TF procoagulant activity presented the same changes as the molecules above. ELISAs showed additional increases in the concentrations of as thrombin antithrombin, procollagen III propeptide, thrombomodulin and PAI-1 in IKKβ+/+ cell supernatant under LPS stimulation, however they decreased in IKKβ−/−. The level of as antithrombin III however, appeared to show the opposite change to those other factors. Immunofluorescence demonstrated a greatly enhanced expression of p65 in the nucleus by IKKβ upregulation, which was reduced by IKKβ downregulation. Conclusions: IKKβ could regulate the expression and secretion of coagulation and fibrinolysis factors in LPS-stimulated AEC II via the NF-κB p65 signaling pathway. The IKKβ molecule is expected to be a new target for prevention of coagulation and fibrinolysis abnormalities in ARDS.
Collapse
Affiliation(s)
- Bo Liu
- Department of Critical Care Medicine, Guizhou Medical University Affiliated Hospital, Guiyang, Guizhou 550001, P.R. China
| | - Yahui Wang
- Department of Critical Care Medicine, Guizhou Medical University Affiliated Hospital, Guiyang, Guizhou 550001, P.R. China
| | - Yanqi Wu
- Department of Critical Care Medicine, Guizhou Medical University Affiliated Hospital, Guiyang, Guizhou 550001, P.R. China
| | - Yumei Cheng
- Department of Critical Care Medicine, Guizhou Medical University Affiliated Hospital, Guiyang, Guizhou 550001, P.R. China
| | - Hong Qian
- Department of Critical Care Medicine, Guizhou Medical University Affiliated Hospital, Guiyang, Guizhou 550001, P.R. China
| | - Huilin Yang
- Department of Critical Care Medicine, Guizhou Medical University Affiliated Hospital, Guiyang, Guizhou 550001, P.R. China
| | - Feng Shen
- Department of Critical Care Medicine, Guizhou Medical University Affiliated Hospital, Guiyang, Guizhou 550001, P.R. China
| |
Collapse
|
41
|
Alotaibi FT, Peng B, Klausen C, Lee AF, Abdelkareem AO, Orr NL, Noga H, Bedaiwy MA, Yong PJ. Plasminogen activator inhibitor-1 (PAI-1) expression in endometriosis. PLoS One 2019; 14:e0219064. [PMID: 31315131 PMCID: PMC6637014 DOI: 10.1371/journal.pone.0219064] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 06/14/2019] [Indexed: 12/13/2022] Open
Abstract
Purpose Deep infiltrating endometriosis (DIE) is defined as an endometriotic lesion penetrating to a depth of >5 mm and is associated with pelvic pain, but the underlying mechanisms are unclear. Our objective is to investigate whether plasminogen activator inhibitor-1 expression (PAI-1) in endometriotic tissues is increased in women with DIE. Methods In this blinded in vitro study, immunohistochemistry and Histoscore were used to examine the expression of PAI-1 in glandular epithelium (GECs) and stroma (SCs) in a total of 62 women: deep infiltrating uterosacral/rectovaginal endometriosis (DIE; n = 13), ovarian endometrioma (OMA; n = 14), superficial peritoneal uterosacral/cul-de-sac endometriosis (SUP; n = 23), uterine (eutopic) endometrium from women with endometriosis (UE; n = 6), and non-endometriosis eutopic endometrium (UC; n = 6). The following patient characteristics were also collected: age, American Fertility Society stage, hormonal suppression, phase of menstrual cycle, dysmenorrhea score and deep dyspareunia score. Results PAI-1 expression in GECs and SCs of the DIE group was significantly higher than that of SUP group (p = 0.01, p = 0.01, respectively) and UE group (p = 0.03, p = 0.04, respectively). Interestingly, increased PAI-1 expression in GECs and SCs was also significantly correlated with increased dysmenorrhea (r = 0.38, p = 0.01; r = 0.34, p = 0.02, respectively). Conclusions We found higher expression of PAI-1 in DIE, and an association between PAI-1 and worse dysmenorrhea.
Collapse
Affiliation(s)
- Fahad T. Alotaibi
- Department of Obstetrics & Gynaecology, BC Children’s Hospital Research Institute, The University of British Columbia, Vancouver, Canada
| | - Bo Peng
- Department of Obstetrics & Gynaecology, BC Children’s Hospital Research Institute, The University of British Columbia, Vancouver, Canada
| | - Christian Klausen
- Department of Obstetrics & Gynaecology, BC Children’s Hospital Research Institute, The University of British Columbia, Vancouver, Canada
| | - Anna F. Lee
- Department of Obstetrics & Gynaecology, BC Children’s Hospital Research Institute, The University of British Columbia, Vancouver, Canada
| | - Amr O. Abdelkareem
- Department of Obstetrics & Gynaecology, BC Children’s Hospital Research Institute, The University of British Columbia, Vancouver, Canada
| | - Natasha L. Orr
- Department of Obstetrics & Gynaecology, BC Children’s Hospital Research Institute, The University of British Columbia, Vancouver, Canada
| | - Heather Noga
- Department of Obstetrics & Gynaecology, BC Children’s Hospital Research Institute, The University of British Columbia, Vancouver, Canada
| | - Mohamed A. Bedaiwy
- Department of Obstetrics & Gynaecology, BC Children’s Hospital Research Institute, The University of British Columbia, Vancouver, Canada
| | - Paul J. Yong
- Department of Obstetrics & Gynaecology, BC Children’s Hospital Research Institute, The University of British Columbia, Vancouver, Canada
- * E-mail:
| |
Collapse
|
42
|
Tsantarliotou MP, Lavrentiadou SN, Psalla DA, Margaritis IE, Kritsepi MG, Zervos IA, Latsari MI, Sapanidou VG, Taitzoglou IA, Sinakos ZM. Suppression of plasminogen activator inhibitor-1 (PAI-1) activity by crocin ameliorates lipopolysaccharide-induced thrombosis in rats. Food Chem Toxicol 2019; 125:190-197. [PMID: 30610936 DOI: 10.1016/j.fct.2019.01.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 12/11/2018] [Accepted: 01/01/2019] [Indexed: 02/07/2023]
Abstract
The imbalance between clot formation and fibrinolysis is mainly attributed to increased levels of plasminogen activator inhibitor type 1 (PAI-1), an inhibitor of fibrinolysis closely involved in inflammatory responses such as septic shock. This increase is mediated by many factors, including reactive oxygen species (ROS). The present study was designed to evaluate the prophylactic effect of crocin, a potent natural antioxidant, on PAI-1 in the rat model of endotoxic shock. Lipopolysaccharide-infused rats (500 μg/kg) showed significant changes in thrombosis-related haematological parameters such as decrease of platelet blood counts and increase (7 fold) of PAI-1 concentration in blood plasma. No effect on t-PA activity was observed. Crocin administration in two different doses (10 mg/kg and 100 mg/kg) 30 min prior to the injection of LPS, inhibited the reduction of platelet counts and ameliorated the concentration of PAI-1 in the liver and the brain. Moreover, crocin inhibited the deposition of fibrin in the renal glomeruli. No significant changes were recorded in the healthy groups of crocin (10 mg/kg and 100 mg/kg) compared to the control group. These data demonstrate the potential of crocin to prevent LPS-induced organ injury and suggest it is worthwhile to investigate the use of antioxidants for the treatment of septicemia.
Collapse
Affiliation(s)
- M P Tsantarliotou
- School of Veterinary Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Greece.
| | - S N Lavrentiadou
- School of Veterinary Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Greece
| | - D A Psalla
- School of Veterinary Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Greece
| | - I E Margaritis
- School of Veterinary Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Greece
| | - M G Kritsepi
- School of Veterinary Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Greece
| | - I A Zervos
- School of Veterinary Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Greece
| | - M I Latsari
- School of Veterinary Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Greece
| | - V G Sapanidou
- School of Veterinary Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Greece
| | - I A Taitzoglou
- School of Veterinary Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Greece
| | - Z M Sinakos
- School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Greece
| |
Collapse
|
43
|
Liu Q, Shi X, Tang L, Xu W, Jiang S, Ding W, Feng Q, Chu H, Ma Y, Li Y, Lu J, Pu W, Zhou X, Jin L, Wang J, Wu W. Salvianolic acid B attenuates experimental pulmonary inflammation by protecting endothelial cells against oxidative stress injury. Eur J Pharmacol 2018; 840:9-19. [PMID: 30273543 DOI: 10.1016/j.ejphar.2018.09.030] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Revised: 09/21/2018] [Accepted: 09/26/2018] [Indexed: 12/21/2022]
Abstract
Endothelial cell injury and subsequent inflammation play pivotal roles in the pathogenesis of pulmonary fibrosis, a progressive and fatal disorder. We found previously that salvianolic acid B (SAB) attenuated experimental pulmonary fibrosis. Pulmonary fibrosis is driven by inflammation, but the anti-inflammatory role and mechanism of SAB on the treatment of pulmonary fibrosis is still unknown. Here, our in vivo studies showed that SAB had a strong anti-inflammatory effect on bleomycin-instilled mice by inhibiting inflammatory cell infiltration and inflammatory cytokine production. Moreover, SAB protected endothelial cells against oxidative stress injury and inhibited endothelial cell apoptosis in bleomycin-treated mice. The in vitro studies also showed that SAB decreased the H2O2-induced overproduction of reactive oxygen species to protect EA.hy926 endothelial cells from oxidative damage, and further inhibited H2O2-induced permeability and overexpression of pro-inflammatory molecules. The next studies revealed that SAB inhibited the H2O2-induced cell apoptosis and attenuated the decrease of tight junction-related gene expression, resulting in a decrease of the endothelial permeability in injured endothelial cells. Furthermore, Western blot analysis suggested that SAB decreased endothelial cell permeability and expression of pro-inflammatory cytokines by inhibiting MAPK and NF-κB signaling pathways. Taken together, these data indicate that SAB exerted anti-inflammatory roles in pulmonary fibrosis by protection of the endothelial cells against oxidative stress injury, mediated by inhibition of endothelial permeability and expression of pro-inflammatory cytokine via the MAPK and NF-κB signaling pathways.
Collapse
Affiliation(s)
- Qingmei Liu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China; State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Xiangguang Shi
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China; State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Luyan Tang
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Weihong Xu
- The Clinical Laboratory of Shanghai Tongren Hosipital, Shanghai, China
| | - Shuai Jiang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Weifeng Ding
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Qian Feng
- Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China
| | - Haiyan Chu
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Yanyun Ma
- Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China
| | - Yuan Li
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Jiaying Lu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Weilin Pu
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Xiaodong Zhou
- University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Li Jin
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Jiucun Wang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China; Human Phenome Institute, Fudan University, Shanghai, China.
| | - Wenyu Wu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China; Department of Dermatology, Jing'an District Central Hospital, Shanghai, China.
| |
Collapse
|
44
|
Shioya S, Masuda T, Senoo T, Horimasu Y, Miyamoto S, Nakashima T, Iwamoto H, Fujitaka K, Hamada H, Hattori N. Plasminogen activator inhibitor-1 serves an important role in radiation-induced pulmonary fibrosis. Exp Ther Med 2018; 16:3070-3076. [PMID: 30214528 PMCID: PMC6125865 DOI: 10.3892/etm.2018.6550] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 07/19/2018] [Indexed: 12/11/2022] Open
Abstract
Radiation-induced pulmonary fibrosis is a serious complication. Plasminogen activator inhibitor-1 (PAI-1) has been indicated to be a key factor in the progression of pulmonary fibrosis. In the present study, the effect of PAI-1 deficiency on radiation-induced pulmonary fibrosis was analyzed. Wild-type (WT) and PAI-1-deficient (PAI-1−/−) mice were treated with thoracic irradiation of 15 Gy to induce pulmonary fibrosis. Analyses of bronchoalveolar lavage (BAL) fluids were performed 0, 4, 12, 18, and 24 weeks after irradiation. The degree of pulmonary fibrosis was assessed according to the histology of lung tissues and hydroxyproline contents. The results demonstrated that the irradiation of WT mice increased PAI-1 expression in the lungs after 18 weeks and established lung fibrosis at 24 weeks. The number of total cells and transforming growth factor-β levels in BAL fluid were significantly lower at 24 weeks after irradiation in PAI-1−/− mice compared with WT mice. Furthermore, histological examination revealed that the extent of pulmonary fibrosis was attenuated in PAI-1−/− mice compared with that in WT mice. Hydroxyproline content was also significantly lower in PAI-1−/− mice compared with WT mice at 24 weeks after irradiation. In conclusion, PAI-1 serves an important role in the development of radiation-induced pulmonary fibrosis and may represent a novel therapeutic target for pulmonary fibrosis.
Collapse
Affiliation(s)
- Sachiko Shioya
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| | - Takeshi Masuda
- Department of Respiratory Internal Medicine, Hiroshima University Hospital, Hiroshima 734-8551, Japan
| | - Tadashi Senoo
- Department of Clinical Oncology, Hiroshima University Hospital, Hiroshima 734-8551, Japan
| | - Yasushi Horimasu
- Department of Respiratory Internal Medicine, Hiroshima University Hospital, Hiroshima 734-8551, Japan
| | - Shintaro Miyamoto
- Department of Respiratory Internal Medicine, Hiroshima University Hospital, Hiroshima 734-8551, Japan
| | - Taku Nakashima
- Department of Respiratory Internal Medicine, Hiroshima University Hospital, Hiroshima 734-8551, Japan
| | - Hiroshi Iwamoto
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| | - Kazunori Fujitaka
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| | - Hironobu Hamada
- Department of Physical Analysis and Therapeutic Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| | - Noboru Hattori
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| |
Collapse
|
45
|
Effect of progesterone on Smad signaling and TGF-β/Smad-regulated genes in lung epithelial cells. PLoS One 2018; 13:e0200661. [PMID: 30001393 PMCID: PMC6042760 DOI: 10.1371/journal.pone.0200661] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 06/29/2018] [Indexed: 11/20/2022] Open
Abstract
The effect of endogenous progesterone and/or exogenous pre- or postnatal progesterone application on lung function of preterm infants is poorly defined. While prenatal progesterone substitution may prevent preterm birth, in vitro and in vivo data suggest a benefit of postnatal progesterone replacement on the incidence and severity of bronchopulmonary dysplasia (BPD). However, the molecular mechanisms responsible for progesterone's effects are undefined. Numerous factors are involved in lung development, airway inflammation, and airway remodeling: the transforming growth factor beta (TGF-β)/mothers against decapentaplegic homolog (Smad) signaling pathway and TGF-β-regulated genes, such as connective tissue growth factor (CTGF), transgelin (TAGLN), and plasminogen activator inhibitor-1 (PAI-1). These processes contribute to the development of BPD. The aim of the present study was to clarify whether progesterone could affect TGF-β1-activated Smad signaling and CTGF/transgelin/PAI-1 expression in lung epithelial cells. The pharmacological effect of progesterone on Smad signaling was investigated using a TGF-β1-inducible luciferase reporter and western blotting analysis of phosphorylated Smad2/3 in A549 lung epithelial cells. The regulation of CTGF, transgelin, and PAI-1 expression by progesterone was studied using a promoter-based luciferase reporter, quantitative real-time PCR, and western blotting in the same cell line. While progesterone alone had no direct effect on Smad signaling in lung epithelial cells, it dose-dependently inhibited TGF-β1-induced Smad3 phosphorylation, as shown by luciferase assays and western blotting analysis. Progesterone also antagonized the TGF-β1/Smad-induced upregulation of CTGF, transgelin, and PAI-1 at the promoter, mRNA, and/or protein levels. The present study highlights possible new molecular mechanisms involving progesterone, including inhibition of TGF-β1-activated Smad signaling and TGF-β1-regulated genes involved in BPD pathogenesis, which are likely to attenuate the development of BPD by inhibiting TGF-β1-mediated airway remodeling. Understanding these mechanisms might help to explain the effects of pre- or postnatal application of progesterone on lung diseases of preterm infants.
Collapse
|
46
|
Ding L, Zhu C, Yu F, Wu P, Chen G, Ullah A, Wang K, Sun M, Li J, Oupický D. Pulmonary delivery of polyplexes for combined PAI-1 gene silencing and CXCR4 inhibition to treat lung fibrosis. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2018; 14:1765-1776. [PMID: 29777878 DOI: 10.1016/j.nano.2018.05.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 04/14/2018] [Accepted: 05/04/2018] [Indexed: 02/06/2023]
Abstract
This report describes the development of polyplexes based on CXCR4-inhibiting poly(ethylenimine) derivative (PEI-C) for pulmonary delivery of siRNA to silence plasminogen activator inhibitor-1 (siPAI-1) as a new combination treatment of pulmonary fibrosis (PF). Safety and delivery efficacy of the PEI-C/siPAI-1 polyplexes was investigated in vitro in primary lung fibroblasts isolated from mice with bleomycin-induced PF. Biodistribution analysis following intratracheal administration of fluorescently labeled polyplexes showed prolonged retention in the lungs. Treatment of mice with bleomycin-induced PF using the PEI-C/siPAI-1 polyplexes resulted in a significant down-regulation of the PAI-1 expression and decreased collagen deposition in the lung. The results of this study provide first evidence of the potential benefits of combined inhibition of CXCR4 and PAI-1 in the pulmonary treatment of PF.
Collapse
Affiliation(s)
- Ling Ding
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, China
| | - Chenfei Zhu
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, China
| | - Fei Yu
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Pengkai Wu
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, China
| | - Gang Chen
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, China
| | - Aftab Ullah
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, China
| | - Kaikai Wang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, China
| | - Minjie Sun
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, China
| | - Jing Li
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - David Oupický
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, China; Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
47
|
Milenkovic J, Milojkovic M, Jevtovic Stoimenov T, Djindjic B, Miljkovic E. Mechanisms of plasminogen activator inhibitor 1 action in stromal remodeling and related diseases. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2017; 161:339-347. [PMID: 29097819 DOI: 10.5507/bp.2017.046] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Accepted: 10/20/2017] [Indexed: 01/11/2023] Open
Abstract
Plasminogen activator inhibitor type 1 (PAI-1) is the main physiologic inhibitor of fibrinolysis. However, it is also involved in many physiological processes such as extracellular matrix (ECM) proteolysis and remodeling, cell adhesion, motility, and apoptosis, angiogenesis, etc. The aim of the study was to summarize current knowledge and gain insights into the mechanisms of PAI-1 action in the processes of stromal remodeling and diseases with considerable matrix pathologies (atherosclerosis, tissue fibrosis, cancer metastasis, pregnancy related complications, etc). As a component of an early cellular response to injury, PAI-1 reacts with membrane surface proteins and participates in the initiation of intracellular signaling, specifically cytoskeletal reorganization and motility. Complexity of ECM homeostasis resides in varying relation of the plasminogen system components and other matrix constituents. Inflammatory mediators (transforming growth factor-β and interferon-γ) and hormones (angiotensin II) are in the close interdependent relation with PAI-1. Also, special attention is devoted to the role of increased PAI-1 concentrations due to the common 4G/5G polymorphism. Some of the novel mechanisms of ECM modification consider PAI-1 dependent stabilization of urokinase mediated cell adhesion, control of the vascular endothelial cadherin trafficking and interaction with endothelial cells proteasome, its relation to matrix metalloproteinase 2 and osteopontin, and oxidative inhibition by myeloperoxidase. Targeting and/or alteration of PAI-1 functions might bring benefit to the future therapeutic approaches in diseases where ECM undergoes substantial remodeling.
Collapse
Affiliation(s)
- Jelena Milenkovic
- Institute of Pathophysiology, Faculty of Medicine University of Nis, Serbia
| | - Maja Milojkovic
- Institute of Pathophysiology, Faculty of Medicine University of Nis, Serbia
| | | | - Boris Djindjic
- Institute of Pathophysiology, Faculty of Medicine University of Nis, Serbia
| | - Edita Miljkovic
- Hematology and Clinical Immunology Clinic, Clinical Center in Nis, Serbia
| |
Collapse
|
48
|
Recent Pathophysiological Aspects of Peyronie's Disease: Role of Free Radicals, Rationale, and Therapeutic Implications for Antioxidant Treatment-Literature Review. Adv Urol 2017; 2017:4653512. [PMID: 28744308 PMCID: PMC5514334 DOI: 10.1155/2017/4653512] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 05/30/2017] [Indexed: 12/13/2022] Open
Abstract
Peyronie's disease (PD) is a chronic inflammation of tunica albuginea of the corpora cavernosa that causes an inelastic plaque resulting in penis deformation. Although its etiology is not completely known, there is general consensus that PD is genetically transmitted and secondary to penile trauma. In recent years, numerous studies demonstrated the role played by oxidative stress in PD pathogenesis, and other studies have described successful use of antioxidants in PD treatment. Oxidative stress is an integral part of this disease, influencing its progression. In the early stages of PD, the inflammatory infiltrate cells produce high quantities of free radicals and proinflammatory and profibrotic cytokines, with consequent activation of transcription factor NF-κB. While conservative therapies commonly used in the early stages of PD include oral substances (Potaba, tamoxifen, colchicine, and vitamin E), intralesional treatment (verapamil, interferon, steroids, and more recently collagenase clostridium histolyticum-Xiaflex), and local physical treatment (iontophoresis, extracorporeal shock wave therapy, and penile extender), the significant results obtained by emerging treatments with the antioxidants cited in this article suggest these therapeutic agents interfere at several levels with the disease's pathogenetic mechanisms. Antioxidants therapy outcomes are interesting for good clinical practice and also confirm the fundamental role played by oxidative stress in PD.
Collapse
|
49
|
Stout-Delgado HW, Cho SJ, Chu SG, Mitzel DN, Villalba J, El-Chemaly S, Ryter SW, Choi AMK, Rosas IO. Age-Dependent Susceptibility to Pulmonary Fibrosis Is Associated with NLRP3 Inflammasome Activation. Am J Respir Cell Mol Biol 2017; 55:252-63. [PMID: 26933834 DOI: 10.1165/rcmb.2015-0222oc] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Aging has been implicated in the development of pulmonary fibrosis, which has seen a sharp increase in incidence in those older than 50 years. Recent studies demonstrate a role for the nucleotide-binding domain and leucine rich repeat containing family, pyrin domain containing 3 (NLRP3) inflammasome and its regulated cytokines in experimental lung fibrosis. In this study, we tested the hypothesis that age-related NLRP3 inflammasome activation is an important predisposing factor in the development of pulmonary fibrosis. Briefly, young and aged wild-type and NLRP3(-/-) mice were subjected to bleomycin-induced lung injury. Pulmonary fibrosis was determined by histology and hydroxyproline accumulation. Bone marrow and alveolar macrophages were isolated from these mice. NLRP3 inflammasome activation was assessed by co-immunoprecipitation experiments. IL-1β and IL-18 production was measured by ELISA. The current study demonstrated that aged wild-type mice developed more lung fibrosis and exhibited increased morbidity and mortality after bleomycin-induced lung injury, when compared with young mice. Bleomycin-exposed aged NLRP3(-/-) mice had reduced fibrosis compared with their wild-type age-matched counterparts. Bone marrow-derived and alveolar macrophages from aged mice displayed higher levels of NLRP3 inflammasome activation and caspase-1-dependent IL-1β and IL-18 production, which was associated with altered mitochondrial function and increased production of reactive oxygen species. Our study demonstrated that age-dependent increases in alveolar macrophage mitochondrial reactive oxygen species production and NLRP3 inflammasome activation contribute to the development of experimental fibrosis.
Collapse
Affiliation(s)
- Heather W Stout-Delgado
- 1 Pulmonary and Critical Care, Department of Medicine, Weill Cornell Medical College, New York, New York.,2 Pulmonary Fibrosis Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico; and
| | - Soo Jung Cho
- 1 Pulmonary and Critical Care, Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Sarah G Chu
- 3 Pulmonary and Critical Care, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Dana N Mitzel
- 2 Pulmonary Fibrosis Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico; and
| | - Julian Villalba
- 2 Pulmonary Fibrosis Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico; and.,3 Pulmonary and Critical Care, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Souheil El-Chemaly
- 2 Pulmonary Fibrosis Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico; and.,3 Pulmonary and Critical Care, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Stefan W Ryter
- 1 Pulmonary and Critical Care, Department of Medicine, Weill Cornell Medical College, New York, New York.,3 Pulmonary and Critical Care, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Augustine M K Choi
- 1 Pulmonary and Critical Care, Department of Medicine, Weill Cornell Medical College, New York, New York.,3 Pulmonary and Critical Care, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Ivan O Rosas
- 2 Pulmonary Fibrosis Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico; and.,3 Pulmonary and Critical Care, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
50
|
Chu H, Shi Y, Jiang S, Zhong Q, Zhao Y, Liu Q, Ma Y, Shi X, Ding W, Zhou X, Cui J, Jin L, Guo G, Wang J. Treatment effects of the traditional Chinese medicine Shenks in bleomycin-induced lung fibrosis through regulation of TGF-beta/Smad3 signaling and oxidative stress. Sci Rep 2017; 7:2252. [PMID: 28533545 PMCID: PMC5440393 DOI: 10.1038/s41598-017-02293-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 04/05/2017] [Indexed: 12/20/2022] Open
Abstract
Pulmonary fibrosis is a kind of devastating interstitial lung disease due to the limited therapeutic strategies. Traditional Chinese medicine (TCM) practices have put forth Shenks as a promising treatment approach. Here, we performed in vivo study and in vitro study to delineate the anti-fibrotic mechanisms behind Shenks treatment for pulmonary fibrosis. We found that regardless of the prophylactic or therapeutic treatment, Shenks was able to attenuate BLM-induced-fibrosis in mice, down regulate extracellular matrix genes expression, and reduce collagen production. The aberrantly high Smad3 phosphorylation levels and SBE activity in TGF-β-induced fibroblasts were dramatically decreased as a result of Shenks treatment. At the same time, Shenks was able to increase the expression of antioxidant-related genes, including Gclc and Ec-sod, while reduce the transcription levels of oxidative-related genes, such as Rac1 and Nox4 demonstrated by both in vivo and in vitro studies. Further investigations found that Shenks could decrease the oxidative productions of protein (3-nitrotyrosine) and lipid (malondialdehyde) and increase GSH content both in bleomycin treated mouse lungs and TGF-β stimulated fibroblasts, as well as inhibit the production of ROS stimulated by TGF-β to fight against oxidative stress. Overall, Shenks inhibited fibrosis by blocking TGF-β pathway and modulating the oxidant/antioxidant balance.
Collapse
Affiliation(s)
- Haiyan Chu
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, 2005 Songhu Road, Shanghai, 200438, P. R. China
| | - Ying Shi
- Department of Rheumatology and Immunology, Yiling Affiliated Hospital of Hebei Medical University, Shijiazhuang, 050091, China
- Department of Traditional Chinese Medicine, Geriatric Hospital of Hebei Province, Shijiazhuang, 050011, China
| | - Shuai Jiang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, 2005 Songhu Road, Shanghai, 200438, P. R. China
| | - Qicheng Zhong
- Department of Rheumatology and Immunology, Yiling Affiliated Hospital of Hebei Medical University, Shijiazhuang, 050091, China
| | - Yongqiang Zhao
- Department of Rheumatology and Immunology, Yiling Affiliated Hospital of Hebei Medical University, Shijiazhuang, 050091, China
| | - Qingmei Liu
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, 2005 Songhu Road, Shanghai, 200438, P. R. China
| | - Yanyun Ma
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, 2005 Songhu Road, Shanghai, 200438, P. R. China
| | - Xiangguang Shi
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, 2005 Songhu Road, Shanghai, 200438, P. R. China
| | - Weifeng Ding
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, 2005 Songhu Road, Shanghai, 200438, P. R. China
| | - Xiaodong Zhou
- University of Texas Health Science Center at Houston, 6431 Fannin St., Houston, Texas, 77030, USA
| | - Jimin Cui
- Department of Rheumatology and Immunology, Yiling Affiliated Hospital of Hebei Medical University, Shijiazhuang, 050091, China
| | - Li Jin
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, 2005 Songhu Road, Shanghai, 200438, P. R. China
| | - Gang Guo
- Department of Rheumatology and Immunology, Yiling Affiliated Hospital of Hebei Medical University, Shijiazhuang, 050091, China.
| | - Jiucun Wang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, 2005 Songhu Road, Shanghai, 200438, P. R. China.
- Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, 200040, P. R. China.
| |
Collapse
|