1
|
Yan S, Zhao Y, Xu W, Zhang J, Zhang Y, Liu B, Li X, Ma Z, Yang Q. ADAM17/PTGS2 Facilitates Pulmonary Fibrosis by Regulating Ferroptosis. J Cell Mol Med 2025; 29:e70466. [PMID: 40077919 PMCID: PMC11903495 DOI: 10.1111/jcmm.70466] [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: 09/19/2024] [Revised: 02/08/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
Pulmonary fibrosis (PF) is a chronic and progressive interstitial lung disease characterised by excessive deposition of extracellular matrix (ECM), resulting in high mortality rates. In this study, we provide evidence that ADAM17/PTGS2 plays a crucial role in inducing ferroptosis in fibroblasts, promoting PF. Initially, an assessment was made of ADAM17 protein levels in patients diagnosed with connective tissue diseases-interstitial lung diseases (CTD-ILD), using ELISA assays. Confirmation of the relationship between ADAM17 and fibrosis was achieved by stimulating cells with PMA or TAPI-1 (the ADAM17 inhibitor), in conjunction with the fibrosis-inducing factor, TGFβ1. To further explore the major downstream proteins of ADAM17 contributing to altered PF, we employed mRNA transcriptomics. To further investigate the role of ADAM17/PTGS2 in promoting ferroptosis and fibrosis, we employed western blot assays, immunofluorescence and transmission electron microscopy (TEM). Furthermore, the effects of the ADAM17/PTGS2/ferroptosis pathway in PF were verified using Adeno-associated virus (AAV)-mediated ADAM17 gene knockdown in mice. In CTD-ILD patients, ADAM17 expression was significantly elevated. Upon PMA stimulation, lung fibroblasts exhibited increased fibrosis-related proteins, and the combined stimulation of PMA and TGFβ1 synergistically promoted cellular fibrosis. Conversely, TAPI-1 alleviated fibrotic stimulation induced by TGFβ1. Transcriptomic analysis of lung fibroblast specimens overexpressing ADAM17 revealed significantly elevated PTGS2 expression levels. Knockdown and ferroptosis inhibition assays demonstrated that ADAM17 regulates ferroptosis in lung fibroblasts via PTGS2, ultimately inducing fibrosis. Furthermore, the deficiency of ADAM17 alleviated bleomycin-induced PF and inflammation in mice. These findings first verified that ADAM17/PTGS2/ferroptosis is a novel mechanism for regulating PF; it provides a new theoretical basis for further exploring the treatment of PF.
Collapse
Affiliation(s)
- Suyan Yan
- Department of Rheumatology and ImmunologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | - Yaqi Zhao
- Department of Rheumatology and ImmunologyShandong Provincial Hospital, Cheeloo College of Medicine, Shandong UniversityJinanShandongChina
| | - Wei Xu
- Department of Rheumatology and ImmunologyShandong Provincial Hospital, Cheeloo College of Medicine, Shandong UniversityJinanShandongChina
| | - Jin Zhang
- Department of Rheumatology and ImmunologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | - Ying Zhang
- Department of Rheumatology and ImmunologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | - Baocheng Liu
- Department of Rheumatology and ImmunologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | - Xinya Li
- Department of Rheumatology and ImmunologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | - Zhenzhen Ma
- Department of Rheumatology and ImmunologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
- Shandong University of Traditional Chinese MedicineJinanShandongChina
| | - Qingrui Yang
- Department of Rheumatology and ImmunologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
- Department of Rheumatology and ImmunologyShandong Provincial Hospital, Cheeloo College of Medicine, Shandong UniversityJinanShandongChina
| |
Collapse
|
2
|
Ricciotti E, Haines PG, Chai W, FitzGerald GA. Prostanoids in Cardiac and Vascular Remodeling. Arterioscler Thromb Vasc Biol 2024; 44:558-583. [PMID: 38269585 PMCID: PMC10922399 DOI: 10.1161/atvbaha.123.320045] [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: 08/22/2023] [Accepted: 01/09/2024] [Indexed: 01/26/2024]
Abstract
Prostanoids are biologically active lipids generated from arachidonic acid by the action of the COX (cyclooxygenase) isozymes. NSAIDs, which reduce the biosynthesis of prostanoids by inhibiting COX activity, are effective anti-inflammatory, antipyretic, and analgesic drugs. However, their use is limited by cardiovascular adverse effects, including myocardial infarction, stroke, hypertension, and heart failure. While it is well established that NSAIDs increase the risk of atherothrombotic events and hypertension by suppressing vasoprotective prostanoids, less is known about the link between NSAIDs and heart failure risk. Current evidence indicates that NSAIDs may increase the risk for heart failure by promoting adverse myocardial and vascular remodeling. Indeed, prostanoids play an important role in modulating structural and functional changes occurring in the myocardium and in the vasculature in response to physiological and pathological stimuli. This review will summarize current knowledge of the role of the different prostanoids in myocardial and vascular remodeling and explore how maladaptive remodeling can be counteracted by targeting specific prostanoids.
Collapse
Affiliation(s)
- Emanuela Ricciotti
- Department of Systems Pharmacology and Translational Therapeutics (E.R., G.A.F.), University of Pennsylvania Perelman School of Medicine, Philadelphia
- Institute for Translational Medicine and Therapeutics (E.R., G.A.F.), University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Philip G Haines
- Rhode Island Hospital, Department of Medicine, Warren Alpert Medical School of Brown University, Providence (P.G.H.)
| | - William Chai
- Health and Human Biology, Division of Biology and Medicine, Brown University, Providence, RI (W.C.)
| | - Garret A FitzGerald
- Department of Systems Pharmacology and Translational Therapeutics (E.R., G.A.F.), University of Pennsylvania Perelman School of Medicine, Philadelphia
- Institute for Translational Medicine and Therapeutics (E.R., G.A.F.), University of Pennsylvania Perelman School of Medicine, Philadelphia
- Department of Medicine (G.A.F.), University of Pennsylvania Perelman School of Medicine, Philadelphia
| |
Collapse
|
3
|
Sayour NV, Tóth VÉ, Nagy RN, Vörös I, Gergely TG, Onódi Z, Nagy N, Bödör C, Váradi B, Ruppert M, Radovits T, Bleckwedel F, Zelarayán LC, Pacher P, Ágg B, Görbe A, Ferdinandy P, Varga ZV. Droplet Digital PCR Is a Novel Screening Method Identifying Potential Cardiac G-Protein-Coupled Receptors as Candidate Pharmacological Targets in a Rat Model of Pressure-Overload-Induced Cardiac Dysfunction. Int J Mol Sci 2023; 24:13826. [PMID: 37762130 PMCID: PMC10531061 DOI: 10.3390/ijms241813826] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/31/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
The identification of novel drug targets is needed to improve the outcomes of heart failure (HF). G-protein-coupled receptors (GPCRs) represent the largest family of targets for already approved drugs, thus providing an opportunity for drug repurposing. Here, we aimed (i) to investigate the differential expressions of 288 cardiac GPCRs via droplet digital PCR (ddPCR) and bulk RNA sequencing (RNAseq) in a rat model of left ventricular pressure-overload; (ii) to compare RNAseq findings with those of ddPCR; and (iii) to screen and test for novel, translatable GPCR drug targets in HF. Male Wistar rats subjected to transverse aortic constriction (TAC, n = 5) showed significant systolic dysfunction vs. sham operated animals (SHAM, n = 5) via echocardiography. In TAC vs. SHAM hearts, RNAseq identified 69, and ddPCR identified 27 significantly differentially expressed GPCR mRNAs, 8 of which were identified using both methods, thus showing a correlation between the two methods. Of these, Prostaglandin-F2α-receptor (Ptgfr) was further investigated and localized on cardiomyocytes and fibroblasts in murine hearts via RNA-Scope. Antagonizing Ptgfr via AL-8810 reverted angiotensin-II-induced cardiomyocyte hypertrophy in vitro. In conclusion, using ddPCR as a novel screening method, we were able to identify GPCR targets in HF. We also show that the antagonism of Ptgfr could be a novel target in HF by alleviating cardiomyocyte hypertrophy.
Collapse
Affiliation(s)
- Nabil V. Sayour
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary; (N.V.S.)
- HCEMM-SU Cardiometabolic Immunology Research Group, 1085 Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Semmelweis University, 1085 Budapest, Hungary
| | - Viktória É. Tóth
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary; (N.V.S.)
- HCEMM-SU Cardiometabolic Immunology Research Group, 1085 Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Semmelweis University, 1085 Budapest, Hungary
| | - Regina N. Nagy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary; (N.V.S.)
| | - Imre Vörös
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary; (N.V.S.)
- HCEMM-SU Cardiometabolic Immunology Research Group, 1085 Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Semmelweis University, 1085 Budapest, Hungary
| | - Tamás G. Gergely
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary; (N.V.S.)
- HCEMM-SU Cardiometabolic Immunology Research Group, 1085 Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Semmelweis University, 1085 Budapest, Hungary
| | - Zsófia Onódi
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary; (N.V.S.)
- HCEMM-SU Cardiometabolic Immunology Research Group, 1085 Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Semmelweis University, 1085 Budapest, Hungary
| | - Noémi Nagy
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, 1085 Budapest, Hungary
| | - Csaba Bödör
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, 1085 Budapest, Hungary
| | - Barnabás Váradi
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary; (N.V.S.)
- HCEMM-SU Cardiometabolic Immunology Research Group, 1085 Budapest, Hungary
| | - Mihály Ruppert
- Heart and Vascular Center, Semmelweis University, 1085 Budapest, Hungary
| | - Tamás Radovits
- Heart and Vascular Center, Semmelweis University, 1085 Budapest, Hungary
| | - Federico Bleckwedel
- Institute of Pharmacology and Toxicology, University Medical Center Goettingen (UMG), 37075 Göttingen, Germany
- German Center for Cardiovascular Research (DZHK) Partner Site, 37075 Goettingen, Germany
| | - Laura C. Zelarayán
- Institute of Pharmacology and Toxicology, University Medical Center Goettingen (UMG), 37075 Göttingen, Germany
- German Center for Cardiovascular Research (DZHK) Partner Site, 37075 Goettingen, Germany
| | - Pal Pacher
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institute on Alcohol Abuse and Alcoholism, National Institute of Health, Rockville, MD 20852, USA
| | - Bence Ágg
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary; (N.V.S.)
- MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary
- Pharmahungary Group, 6720 Szeged, Hungary
| | - Anikó Görbe
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary; (N.V.S.)
- Pharmahungary Group, 6720 Szeged, Hungary
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary; (N.V.S.)
- MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary
- Pharmahungary Group, 6720 Szeged, Hungary
| | - Zoltán V. Varga
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary; (N.V.S.)
- HCEMM-SU Cardiometabolic Immunology Research Group, 1085 Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Semmelweis University, 1085 Budapest, Hungary
| |
Collapse
|
4
|
Beccacece L, Abondio P, Bini C, Pelotti S, Luiselli D. The Link between Prostanoids and Cardiovascular Diseases. Int J Mol Sci 2023; 24:ijms24044193. [PMID: 36835616 PMCID: PMC9962914 DOI: 10.3390/ijms24044193] [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: 01/31/2023] [Revised: 02/17/2023] [Accepted: 02/18/2023] [Indexed: 02/22/2023] Open
Abstract
Cardiovascular diseases are the leading cause of global deaths, and many risk factors contribute to their pathogenesis. In this context, prostanoids, which derive from arachidonic acid, have attracted attention for their involvement in cardiovascular homeostasis and inflammatory processes. Prostanoids are the target of several drugs, but it has been shown that some of them increase the risk of thrombosis. Overall, many studies have shown that prostanoids are tightly associated with cardiovascular diseases and that several polymorphisms in genes involved in their synthesis and function increase the risk of developing these pathologies. In this review, we focus on molecular mechanisms linking prostanoids to cardiovascular diseases and we provide an overview of genetic polymorphisms that increase the risk for cardiovascular disease.
Collapse
Affiliation(s)
- Livia Beccacece
- Computational Genomics Lab, Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy
- Correspondence: (L.B.); (P.A.)
| | - Paolo Abondio
- aDNA Lab, Department of Cultural Heritage, University of Bologna, Ravenna Campus, 48121 Ravenna, Italy
- Correspondence: (L.B.); (P.A.)
| | - Carla Bini
- Unit of Legal Medicine, Department of Medical and Surgical Sciences, University of Bologna, 40126 Bologna, Italy
| | - Susi Pelotti
- Unit of Legal Medicine, Department of Medical and Surgical Sciences, University of Bologna, 40126 Bologna, Italy
| | - Donata Luiselli
- aDNA Lab, Department of Cultural Heritage, University of Bologna, Ravenna Campus, 48121 Ravenna, Italy
| |
Collapse
|
5
|
Alotaibi M, Shao J, Pauciulo MW, Nichols WC, Hemnes AR, Malhotra A, Kim NH, Yuan JXJ, Fernandes T, Kerr KM, Alshawabkeh L, Desai AA, Bujor AM, Lafyatis R, Watrous JD, Long T, Cheng S, Chan SY, Jain M. Metabolomic Profiles Differentiate Scleroderma-PAH From Idiopathic PAH and Correspond With Worsened Functional Capacity. Chest 2023; 163:204-215. [PMID: 36087794 PMCID: PMC9899641 DOI: 10.1016/j.chest.2022.08.2230] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 07/12/2022] [Accepted: 08/19/2022] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND The prognosis and therapeutic responses are worse for pulmonary arterial hypertension associated with systemic sclerosis (SSc-PAH) compared with idiopathic pulmonary arterial hypertension (IPAH). This discrepancy could be driven by divergence in underlying metabolic determinants of disease. RESEARCH QUESTION Are circulating bioactive metabolites differentially altered in SSc-PAH vs IPAH, and can this alteration explain clinical disparity between these PAH subgroups? STUDY DESIGN AND METHODS Plasma biosamples from 400 patients with SSc-PAH and 1,082 patients with IPAH were included in the study. Another cohort of 100 patients with scleroderma with no PH and 44 patients with scleroderma with PH was included for external validation. More than 700 bioactive lipid metabolites, representing a range of vasoactive and immune-inflammatory pathways, were assayed in plasma samples from independent discovery and validation cohorts using liquid chromatography/high-resolution mass spectrometry-based approaches. Regression analyses were used to identify metabolites that exhibited differential levels between SSc-PAH and IPAH and associated with disease severity. RESULTS From hundreds of circulating bioactive lipid molecules, five metabolites were found to distinguish between SSc-PAH and IPAH, as well as associate with markers of disease severity. Relative to IPAH, patients with SSc-PAH carried increased levels of fatty acid metabolites, including lignoceric acid and nervonic acid, as well as eicosanoids/oxylipins and sex hormone metabolites. INTERPRETATION Patients with SSc-PAH are characterized by an unfavorable bioactive metabolic profile that may explain the poor and limited response to therapy. These data provide important metabolic insights into the molecular heterogeneity underlying differences between subgroups of PAH.
Collapse
Affiliation(s)
- Mona Alotaibi
- Division of Pulmonary, Critical Care and Sleep Medicine, University of California San Diego, La Jolla, CA; Department of Medicine, University of California San Diego, La Jolla, CA
| | - Junzhe Shao
- School of Life Sciences, Peking University, Beijing, China
| | - Michael W Pauciulo
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA; Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - William C Nichols
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA; Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Anna R Hemnes
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Atul Malhotra
- Division of Pulmonary, Critical Care and Sleep Medicine, University of California San Diego, La Jolla, CA; Department of Medicine, University of California San Diego, La Jolla, CA
| | - Nick H Kim
- Division of Pulmonary, Critical Care and Sleep Medicine, University of California San Diego, La Jolla, CA; Department of Medicine, University of California San Diego, La Jolla, CA
| | - Jason X-J Yuan
- Division of Pulmonary, Critical Care and Sleep Medicine, University of California San Diego, La Jolla, CA; Department of Medicine, University of California San Diego, La Jolla, CA
| | - Timothy Fernandes
- Division of Pulmonary, Critical Care and Sleep Medicine, University of California San Diego, La Jolla, CA; Department of Medicine, University of California San Diego, La Jolla, CA
| | - Kim M Kerr
- Division of Pulmonary, Critical Care and Sleep Medicine, University of California San Diego, La Jolla, CA; Department of Medicine, University of California San Diego, La Jolla, CA
| | - Laith Alshawabkeh
- Division of Cardiovascular Medicine, Sulpizio Cardiovascular Institute, University of California San Diego, La Jolla, CA
| | - Ankit A Desai
- Department of Medicine, Indiana University, Indianapolis, IN
| | - Andreea M Bujor
- Division of Rheumatology, Boston University Medical Center, Boston, MA
| | - Robert Lafyatis
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Jeramie D Watrous
- Department of Medicine, University of California San Diego, La Jolla, CA
| | - Tao Long
- Department of Medicine, University of California San Diego, La Jolla, CA
| | - Susan Cheng
- Barbra Streisand Women's Heart Center, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Stephen Y Chan
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA.
| | - Mohit Jain
- Department of Medicine, University of California San Diego, La Jolla, CA
| |
Collapse
|
6
|
Evolution of the Concepts of Endometrosis, Post Breeding Endometritis, and Susceptibility of Mares. Animals (Basel) 2022; 12:ani12060779. [PMID: 35327176 PMCID: PMC8944725 DOI: 10.3390/ani12060779] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/12/2022] [Accepted: 03/16/2022] [Indexed: 12/24/2022] Open
Abstract
In this paper, the evolution of our understanding about post breeding endometritis (PBE), the susceptibility of mares, and events leading to endometrosis are reviewed. When sperm arrive in the uterus, pro-inflammatory cytokines and chemokines are released. They attract neutrophils and induce modulatory cytokines which control inflammation. In susceptible mares, this physiological defense can be prolonged since the pattern of cytokine release differs from that of resistant mares being delayed and weaker for anti-inflammatory cytokines. Delayed uterine clearance due to conformational defects, deficient myometrial contractions, and failure of the cervix to relax is detected by intrauterine fluid accumulation and is an important reason for susceptibility to endometritis. Multiparous aged mares are more likely to be susceptible. Untreated prolonged PBE can lead to bacterial or fungal endometritis called persistent or chronic endometritis. Exuberant or prolonged neutrophilia and cytokine release can have deleterious and permanent effects in inducing endometrosis. Interactions of neutrophils, cytokines, and prostaglandins in the formation of collagen and extracellular matrix in the pathogenesis of fibrosis are discussed. Endometritis and endometrosis are interconnected, influencing each other. It is suggested that they represent epigenetic changes induced by age and hostile uterine environment.
Collapse
|
7
|
Chen ZY, Xiao HW, Dong JL, Li Y, Wang B, Fan SJ, Cui M. Gut Microbiota-Derived PGF2α Fights against Radiation-Induced Lung Toxicity through the MAPK/NF-κB Pathway. Antioxidants (Basel) 2021; 11:antiox11010065. [PMID: 35052569 PMCID: PMC8773112 DOI: 10.3390/antiox11010065] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 12/24/2021] [Accepted: 12/24/2021] [Indexed: 12/28/2022] Open
Abstract
Radiation pneumonia is a common and intractable side effect associated with radiotherapy for chest cancer and involves oxidative stress damage and inflammation, prematurely halting the remedy and reducing the life quality of patients. However, the therapeutic options for the complication have yielded disappointing results in clinical application. Here, we report an effective avenue for fighting against radiation pneumonia. Faecal microbiota transplantation (FMT) reduced radiation pneumonia, scavenged oxidative stress and improved lung function in mouse models. Local chest irradiation shifted the gut bacterial taxonomic proportions, which were preserved by FMT. The level of gut microbiota-derived PGF2α decreased following irradiation but increased after FMT. Experimental mice with PGF2α replenishment, via an oral route, exhibited accumulated PGF2α in faecal pellets, peripheral blood and lung tissues, resulting in the attenuation of inflammatory status of the lung and amelioration of lung respiratory function following local chest irradiation. PGF2α activated the FP/MAPK/NF-κB axis to promote cell proliferation and inhibit apoptosis with radiation challenge; silencing MAPK attenuated the protective effect of PGF2α on radiation-challenged lung cells. Together, our findings pave the way for the clinical treatment of radiotherapy-associated complications and underpin PGF2α as a gut microbiota-produced metabolite.
Collapse
Affiliation(s)
- Zhi-Yuan Chen
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300110, China; (Z.-Y.C.); (J.-L.D.); (Y.L.); (B.W.)
| | - Hui-Wen Xiao
- Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China;
| | - Jia-Li Dong
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300110, China; (Z.-Y.C.); (J.-L.D.); (Y.L.); (B.W.)
| | - Yuan Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300110, China; (Z.-Y.C.); (J.-L.D.); (Y.L.); (B.W.)
| | - Bin Wang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300110, China; (Z.-Y.C.); (J.-L.D.); (Y.L.); (B.W.)
| | - Sai-Jun Fan
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300110, China; (Z.-Y.C.); (J.-L.D.); (Y.L.); (B.W.)
- Correspondence: (S.-J.F.); (M.C.)
| | - Ming Cui
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300110, China; (Z.-Y.C.); (J.-L.D.); (Y.L.); (B.W.)
- Correspondence: (S.-J.F.); (M.C.)
| |
Collapse
|
8
|
Rebordão MR, Amaral A, Fernandes C, Silva E, Lukasik K, Szóstek-Mioduchowska A, Pinto-Bravo P, Galvão A, Skarzynski DJ, Ferreira-Dias G. Enzymes Present in Neutrophil Extracellular Traps May Stimulate the Fibrogenic PGF 2α Pathway in the Mare Endometrium. Animals (Basel) 2021; 11:ani11092615. [PMID: 34573581 PMCID: PMC8469524 DOI: 10.3390/ani11092615] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 09/02/2021] [Accepted: 09/02/2021] [Indexed: 12/23/2022] Open
Abstract
Simple Summary Endometrosis is a fibrotic disease in mare endometrium whose pathological mechanisms remain obscure. Prostaglandin (PG)F2α, despite modulating reproductive physiological processes, may also provoke local pathological collagen deposition (fibrogenesis). Neutrophil extracellular traps (NETs) released during inflammation have been linked to fibrogenesis in several tissues. We have previously shown that enzymes found in NETs increase in vitro collagen production in mare endometrium. In this study, activation of PGF2α-pathway in equine endometrial explants challenged in vitro by enzymes found in NETs is shown. Our results indicate that both endocrine microenvironment (estrous cycle phase) and healthy or pathological conditions of endometrial tissues play an important role in PGF2α-pathway activation. In the endometrium of the follicular phase, we have observed both high production of PGF2α and/or PGF2α receptor gene transcription under the action of enzymes found in NETs, both conditions associated with fibrogenesis in other tissues. Nevertheless, transcription of the PGF2α receptor gene does not appear to be hormone-dependent, albeit their levels seem to be dependent on endometrial category in the mid-luteal phase. This study suggests that enzymes existing in NETs may instigate changes on PGF2α mediators, which may become an additional mechanism of fibrogenesis in mare endometrium. Abstract Endometrosis, a fibrotic disease of mare endometrium, impairs uterine function. Prostaglandins (PG), despite modulating reproductive physiological functions, may also cause local pathological collagen deposition (fibrogenesis). We have previously shown that neutrophil extracellular traps (NETs) may also favor mare endometrosis. The aim of this study was to investigate the effect of enzymes present in NETs on PGF2α-pathway activation. Kenney and Doig’s type I/IIA and IIB/III mare endometria, from follicular phase (FLP) and mid-luteal (MLP) phase, were cultured in vitro in the presence of NETs enzymes (elastase, cathepsin-G or myeloperoxidase). Production of PGF2α (EIA) and transcription (qPCR) of its synthases (PTGS2, AKR1C3) and receptor (PTGFR) genes were evaluated. PGF2α and PTGFR were influenced by endometrial category and estrous cycle phase. In FLP endometrium, NETs enzymes induced both high PGF2α production and/or PTGFR transcription. In MLP type I/IIA tissues, down-regulation of PTGFR transcripts occurred. However, in MLP type IIB/III endometrium, high levels of PTGFR transcripts were induced by NETs enzymes. As PGF2α-pathway activation facilitates fibrogenesis in other tissues, PGF2α may be involved in endometrosis pathogenesis. In the mare, the endocrine microenvironment of healthy and pathological endometrium might modulate the PGF2α pathway, as well as fibrosis outcome on endometrium challenged by NETs enzymes.
Collapse
Affiliation(s)
- Maria Rosa Rebordão
- CIISA—Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, 1300-477 Lisboa, Portugal; (M.R.R.); (A.A.); (C.F.); (E.S.)
- Polytechnic Institute of Coimbra, College of Agriculture, 3045-601 Coimbra, Portugal;
| | - Ana Amaral
- CIISA—Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, 1300-477 Lisboa, Portugal; (M.R.R.); (A.A.); (C.F.); (E.S.)
| | - Carina Fernandes
- CIISA—Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, 1300-477 Lisboa, Portugal; (M.R.R.); (A.A.); (C.F.); (E.S.)
| | - Elisabete Silva
- CIISA—Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, 1300-477 Lisboa, Portugal; (M.R.R.); (A.A.); (C.F.); (E.S.)
| | - Karolina Lukasik
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of PAS, 10-748 Olsztyn, Poland; (K.L.); (A.S.-M.); (A.G.); (D.J.S.)
| | - Anna Szóstek-Mioduchowska
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of PAS, 10-748 Olsztyn, Poland; (K.L.); (A.S.-M.); (A.G.); (D.J.S.)
| | - Pedro Pinto-Bravo
- Polytechnic Institute of Coimbra, College of Agriculture, 3045-601 Coimbra, Portugal;
| | - António Galvão
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of PAS, 10-748 Olsztyn, Poland; (K.L.); (A.S.-M.); (A.G.); (D.J.S.)
| | - Dariusz J. Skarzynski
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of PAS, 10-748 Olsztyn, Poland; (K.L.); (A.S.-M.); (A.G.); (D.J.S.)
| | - Graça Ferreira-Dias
- CIISA—Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, 1300-477 Lisboa, Portugal; (M.R.R.); (A.A.); (C.F.); (E.S.)
- Correspondence: ; Tel.: +351-213-652-859
| |
Collapse
|
9
|
Zhou HM, Ti Y, Wang H, Shang YY, Liu YP, Ni XN, Wang D, Wang ZH, Zhang W, Zhong M. Cell death-inducing DFFA-like effector C/CIDEC gene silencing alleviates diabetic cardiomyopathy via upregulating AMPKa phosphorylation. FASEB J 2021; 35:e21504. [PMID: 33913563 DOI: 10.1096/fj.202002562r] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 02/17/2021] [Accepted: 02/19/2021] [Indexed: 11/11/2022]
Abstract
Cell death-inducing DFFA-like effector C (CIDEC) is responsible for metabolic disturbance and insulin resistance, which are considered to be important triggers in the development of diabetic cardiomyopathy (DCM). To investigate whether CIDEC plays a critical role in DCM, DCM rat model was induced by a high-fat diet and a single injection of low-dose streptozotocin (27.5 mg/kg). DCM rats showed severe metabolic disturbance, insulin resistance, myocardial hypertrophy, interstitial fibrosis, ectopic lipid deposition, inflammation and cardiac dysfunction, accompanied by CIDEC elevation. With CIDEC gene silencing, the above pathophysiological characteristics were significantly ameliorated accompanied by significant improvements in cardiac function in DCM rats. Enhanced AMP-activated protein kinase (AMPK) α activation was involved in the underlying pathophysiological molecular mechanisms. To further explore the underlying mechanisms that CIDEC facilitated collagen syntheses in vitro, insulin-resistant cardiac fibroblast (CF) model was induced by high glucose (15.5 mmol/L) and high insulin (104 μU/mL). We observed that insulin-resistant stimulation dramatically raised CIDEC expression and promoted CIDEC nuclear translocation in CFs. Meanwhile, AMPKα2 was observed to distribute almost completely inside CF nucleus. The results further proved that CIDEC biochemically interacted and co-localized with AMPKα2 rather than AMPKα1 in CF nucleus, which provided a novel mechanism of CIDEC in promoting collagen syntheses. This study suggested that CIDEC gene silencing alleviates DCM via AMPKα signaling both in vivo and in vitro, implicating CIDEC may be a promising target for treatment of human DCM.
Collapse
Affiliation(s)
- Hui-Min Zhou
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yun Ti
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Hui Wang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Geriatric Medicines, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yuan-Yuan Shang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ya-Peng Liu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiao-Ning Ni
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Di Wang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zhi-Hao Wang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Geriatric Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Shandong key Laboratory of Cardiovascular Proteomics, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wei Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ming Zhong
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
10
|
Li K, Zhao J, Wang M, Niu L, Wang Y, Li Y, Zheng Y. The Roles of Various Prostaglandins in Fibrosis: A Review. Biomolecules 2021; 11:biom11060789. [PMID: 34073892 PMCID: PMC8225152 DOI: 10.3390/biom11060789] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/20/2021] [Accepted: 05/12/2021] [Indexed: 02/07/2023] Open
Abstract
Organ fibrosis is a common pathological result of various chronic diseases with multiple causes. Fibrosis is characterized by the excessive deposition of extracellular matrix and eventually leads to the destruction of the tissue structure and impaired organ function. Prostaglandins are produced by arachidonic acid through cyclooxygenases and various prostaglandin-specific synthases. Prostaglandins bind to homologous receptors on adjacent tissue cells in an autocrine or paracrine manner and participate in the regulation of a series of physiological or pathological processes, including fibrosis. This review summarizes the properties, synthesis, and degradation of various prostaglandins, as well as the roles of these prostaglandins and their receptors in fibrosis in multiple models to reveal the clinical significance of prostaglandins and their receptors in the treatment of fibrosis.
Collapse
|
11
|
Beck H, Thaler T, Meibom D, Meininghaus M, Jörißen H, Dietz L, Terjung C, Bairlein M, von Bühler CJ, Anlauf S, Fürstner C, Stellfeld T, Schneider D, Gericke KM, Buyck T, Lovis K, Münster U, Anlahr J, Kersten E, Levilain G, Marossek V, Kast R. Potent and Selective Human Prostaglandin F (FP) Receptor Antagonist (BAY-6672) for the Treatment of Idiopathic Pulmonary Fibrosis (IPF). J Med Chem 2020; 63:11639-11662. [PMID: 32969660 DOI: 10.1021/acs.jmedchem.0c00834] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a rare and devastating chronic lung disease of unknown etiology. Despite the approved treatment options nintedanib and pirfenidone, the medical need for a safe and well-tolerated antifibrotic treatment of IPF remains high. The human prostaglandin F receptor (hFP-R) is widely expressed in the lung tissue and constitutes an attractive target for the treatment of fibrotic lung diseases. Herein, we present our research toward novel quinoline-based hFP-R antagonists, including synthesis and detailed structure-activity relationship (SAR). Starting from a high-throughput screening (HTS) hit of our corporate compound library, multiple parameter improvements-including increase of the relative oral bioavailability Frel from 3 to ≥100%-led to a highly potent and selective hFP-R antagonist with complete oral absorption from suspension. BAY-6672 (46) represents-to the best of our knowledge-the first reported FP-R antagonist to demonstrate in vivo efficacy in a preclinical animal model of lung fibrosis, thus paving the way for a new treatment option in IPF.
Collapse
Affiliation(s)
- Hartmut Beck
- Research & Development, Pharmaceuticals, Bayer AG, 42096 Wuppertal, Germany
| | - Tobias Thaler
- Research & Development, Pharmaceuticals, Bayer AG, 42096 Wuppertal, Germany
| | - Daniel Meibom
- Research & Development, Pharmaceuticals, Bayer AG, 42096 Wuppertal, Germany
| | - Mark Meininghaus
- Research & Development, Pharmaceuticals, Bayer AG, 42096 Wuppertal, Germany
| | - Hannah Jörißen
- Research & Development, Pharmaceuticals, Bayer AG, 42096 Wuppertal, Germany
| | - Lisa Dietz
- Research & Development, Pharmaceuticals, Bayer AG, 42096 Wuppertal, Germany
| | - Carsten Terjung
- Research & Development, Pharmaceuticals, Bayer AG, 42096 Wuppertal, Germany
| | - Michaela Bairlein
- Research & Development, Pharmaceuticals, Bayer AG, 42096 Wuppertal, Germany
| | | | - Sonja Anlauf
- Research & Development, Pharmaceuticals, Bayer AG, 42096 Wuppertal, Germany
| | - Chantal Fürstner
- Research & Development, Pharmaceuticals, Bayer AG, 42096 Wuppertal, Germany
| | - Timo Stellfeld
- Research & Development, Pharmaceuticals, Bayer AG, 13353 Berlin, Germany
| | - Dirk Schneider
- Research & Development, Pharmaceuticals, Bayer AG, 42096 Wuppertal, Germany
| | - Kersten M Gericke
- Research & Development, Pharmaceuticals, Bayer AG, 42096 Wuppertal, Germany
| | - Thomas Buyck
- Research & Development, Pharmaceuticals, Bayer AG, 42096 Wuppertal, Germany
| | - Kai Lovis
- Research & Development, Pharmaceuticals, Bayer AG, 42096 Wuppertal, Germany
| | - Uwe Münster
- Research & Development, Pharmaceuticals, Bayer AG, 42096 Wuppertal, Germany
| | - Johanna Anlahr
- Research & Development, Pharmaceuticals, Bayer AG, 42096 Wuppertal, Germany
| | - Elisabeth Kersten
- Research & Development, Pharmaceuticals, Bayer AG, 42096 Wuppertal, Germany
| | - Guillaume Levilain
- Research & Development, Pharmaceuticals, Bayer AG, 42096 Wuppertal, Germany
| | - Virginia Marossek
- Research & Development, Pharmaceuticals, Bayer AG, 42096 Wuppertal, Germany
| | - Raimund Kast
- Research & Development, Pharmaceuticals, Bayer AG, 42096 Wuppertal, Germany
| |
Collapse
|
12
|
Szóstek-Mioduchowska A, Leciejewska N, Zelmańska B, Staszkiewicz-Chodor J, Ferreira-Dias G, Skarzynski D. Lysophosphatidic acid as a regulator of endometrial connective tissue growth factor and prostaglandin secretion during estrous cycle and endometrosis in the mare. BMC Vet Res 2020; 16:343. [PMID: 32943074 PMCID: PMC7499873 DOI: 10.1186/s12917-020-02562-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 09/08/2020] [Indexed: 12/22/2022] Open
Abstract
Background Equine endometrosis is a chronic degenerative condition, described as endometrial fibrosis that forms in the stroma, under the basement membrane and around the endometrial glands. The role of lysophosphatidic acid (LPA) in the development of tissue fibrosis varies depending on the organ, and its profibrotic role in mare endometrosis remains unclear. The study aimed to establish the endometrial presence of LPA and its receptors (LPAR1–4), together with its effects on connective tissue growth factor (CTGF) and prostaglandins (PG) secretion from equine endometrium under physiological (estrous cycle), or pathological conditions (endometrosis). Mare endometria in the mid-luteal phase (n = 5 for each category I, IIA, IIB, III of Kenney and Doig) and in the follicular phase (n = 5 for each category I, IIA, III and n = 4 for IIB) were used. In experiment 1, the levels of LPA, LPAR1–4 mRNA level and protein abundance were investigated in endometria at different stages of endometrosis. In experiment 2, the in vitro effect of LPA (10− 9 M) on the secretion of CTGF and PGs from endometrial tissue explants at different stages of endometrosis were determined. Results Endometrial LPA concentration was higher in the mid-luteal phase compared to the follicular phase in category I endometrium (P < 0.01). There was an alteration in endometrial concentrations of LPA and LPAR1–4 protein abundance in the follicular phase at different stages of endometrosis (P < 0.05). Additionally, LPA increased the secretion of PGE2 from category I endometrium in both phases of the estrous cycle (P < 0.05). The effect of LPA on the secretion of CTGF and PGF2α from endometrial tissue was altered depending on different stages of endometrosis (P < 0.05). Conclusion Our data indicate that endometrosis disturbs proper endometrial function and is associated with altered endometrial LPA concentration, its receptor expression and protein abundance, PGE2/PGF2α ratio, and CTGF secretion in response to LPA. These changes could influence several physiological events occurring in endometrium in mare during estrous cycle and early pregnancy.
Collapse
Affiliation(s)
- Anna Szóstek-Mioduchowska
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Tuwima-st 10, 10-748, Olsztyn, Poland.
| | - Natalia Leciejewska
- Department of Animal Physiology and Biochemistry and Biostructure, Faculty of Veterinary Medicine and Animal Science, Poznan University of Life Sciences, Poznan, Poland
| | - Beata Zelmańska
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Tuwima-st 10, 10-748, Olsztyn, Poland
| | - Joanna Staszkiewicz-Chodor
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Tuwima-st 10, 10-748, Olsztyn, Poland
| | - Graça Ferreira-Dias
- CIISA, Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Lisbon, Portugal
| | - Dariusz Skarzynski
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Tuwima-st 10, 10-748, Olsztyn, Poland
| |
Collapse
|
13
|
Szóstek-Mioduchowska AZ, Baclawska A, Rebordão MR, Ferreira-Dias G, Skarzynski DJ. Prostaglandins effect on matrix metallopeptidases and collagen in mare endometrial fibroblasts. Theriogenology 2020; 153:74-84. [PMID: 32442743 DOI: 10.1016/j.theriogenology.2020.04.040] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 04/09/2020] [Accepted: 04/30/2020] [Indexed: 01/24/2023]
Abstract
An increasing number of studies have shown that prostaglandins (PGs) exert multiple regulatory actions in the processes associated to tissue remodeling and fibrosis. Extracellular matrix (ECM) turnover is mediated by matrix metallopeptidases (MMPs). The knowledge about the regulation of their expression in mare endometrium is still limited. Thus, the aim of this study was to investigate whether: (i) profibrotic transforming growth factor (TGF)-β1 modulates PG production in equine endometrium; and (ii) PGE2 and PGF2α modulate MMPs, their tissue inhibitors (TIMPs), and collagen 1 (COL1) expression. In experiment 1, the effect of TGF-β1 (5 ng/mL) on PG secretion and PG synthases mRNA transcription, after 24 and 48 h treatment of mare endometrial fibroblast and epithelial cells was investigated using ELISA and qPCR. In experiment 2, the effects of PGE2 and PGF2α in doses 10-7M and 10-8M on secretion and MMP1, 2, 9, 13, TIMP1, 2, and COL1A1 mRNA transcription in mare endometrial fibroblasts were assessed. Transforming growth factor-β1 treatment decreased secretion of PGF2α by endometrial fibroblasts (P < 0.05) and PGF2α and PGE2 by endometrial epithelial cells (P < 0.05). Prostaglandin E2 increased MMP-2 and MMP-9, and decreased MMP-13 secretion by endometrial fibroblasts (P < 0.05). Additionally, PGF2α treatment increased MMP-2, MMP-13 and COL1, but decreased MMP-1 secretion by endometrial fibroblasts (P < 0.05). Prostaglandins may be involved in the processes associated to pathological endometrial remodeling by their effect on MMP expression. The effect of PGF2α on COL1 secretion from fibroblasts suggests its profibrotic role in pathological endometrial remodeling.
Collapse
Affiliation(s)
- Anna Zuzanna Szóstek-Mioduchowska
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima Str 10, 10-748, Olsztyn, Poland.
| | - Agnieszka Baclawska
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima Str 10, 10-748, Olsztyn, Poland
| | - Maria Rosa Rebordão
- CIISA - Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Lisbon, Portugal; Department of Animal Sciences, Coimbra College of Agriculture, Polytechnic Institute of Coimbra, Coimbra, Portugal
| | - Graca Ferreira-Dias
- CIISA - Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Lisbon, Portugal
| | - Dariusz Jan Skarzynski
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima Str 10, 10-748, Olsztyn, Poland
| |
Collapse
|
14
|
Neutrophils, monocytes and other immune components in the equine endometrium: Friends or foes? Theriogenology 2020; 150:150-157. [PMID: 31973963 DOI: 10.1016/j.theriogenology.2020.01.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 01/11/2020] [Indexed: 02/06/2023]
Abstract
The innate and adaptive immune mechanisms are key components of regulation of reproductive physiological function and uterine disorders in equine uterus. The predominant immunological response in equine endometrium, characterized by an innate immune response, occurs under estrogens influence, in the follicular phase. Although, the increase in immune-related genes in equine endometrium during estrus has been suggested to play a role in uterine clearance after mating, immune cells and their product, i.e. cytokines play also mandatory role in the luteal development and maintenance, regression of equine corpus luteum, as well as in early pregnancy. Innate immune response is nonspecific and acts as the first line of defense against pathogens, foreign stimuli that include constituents of seminal fluid and local infections (endometritis). It has been recently established that a phagocytosis-independent mechanism to restrain bacteria, by means of neutrophil extracellular traps (NETs) formation, is involved in pathogenesis of in mare endometrial fibrosis (endometrosis). Moreover, persistent macrophages and mast cell activation could also have pro-fibrotic roles by secreting great amounts of pro-fibrotic factors and lead to fibrosis. This review will highlight the involvement of immune key components of the innate and adaptive immune system and their products in equine uterus and their contribution to reproductive physiological function and uterine disorders.
Collapse
|
15
|
Domokos D, Ducza E, Gáspár R. RhoA and Rho-kinase inhibitors modulate cervical resistance: The possible role of RhoA/Rho-kinase signalling pathway in cervical ripening and contractility. Eur J Pharmacol 2019; 843:27-33. [DOI: 10.1016/j.ejphar.2018.11.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 11/11/2018] [Accepted: 11/12/2018] [Indexed: 12/14/2022]
|
16
|
Sharif NA, Klimko PG. Prostaglandin FP receptor antagonists: discovery, pharmacological characterization and therapeutic utility. Br J Pharmacol 2018; 176:1059-1078. [PMID: 29679483 DOI: 10.1111/bph.14335] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Revised: 03/19/2018] [Accepted: 03/28/2018] [Indexed: 12/19/2022] Open
Abstract
In contrast to the availability of potent and selective antagonists of several prostaglandin receptor types (including DP1 , DP2 , EP and TP receptors), there has been a paucity of well-characterized, selective FP receptor antagonists. The earliest ones included dimethyl amide and dimethyl amine derivatives of PGF2α , but these have failed to gain prominence. The fluorinated PGF2α analogues, AL-8810 and AL-3138, were subsequently discovered as competitive and non-competitive FP receptor antagonists respectively. Non-prostanoid structures, such as the thiazolidinone AS604872, the D-amino acid-based oligopeptide PDC31 and its peptidomimic analogue PDC113.824 came next, but the latter two are allosteric inhibitors of FP receptor signalling. AL-8810 has a sub-micromolar in vitro potency and ≥2 log unit selectivity against most other PG receptors when tested in several cell- and tissue-based functional assays. Additionally, AL-8810 has demonstrated therapeutic efficacy as an FP receptor antagonist in animal models of stroke, traumatic brain injury, multiple sclerosis, allodynia and endometriosis. Consequently, it appears that AL-8810 has become the FP receptor antagonist of choice. LINKED ARTICLES: This article is part of a themed section on Eicosanoids 35 years from the 1982 Nobel: where are we now? To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.8/issuetoc.
Collapse
Affiliation(s)
| | - Peter G Klimko
- Novartis Pharmaceuticals Corporation, Fort Worth, TX, 76134, USA
| |
Collapse
|
17
|
Palmer DJ, Scheman A, Tanna AP, Bueno A. The effect of cutaneous prostaglandin application on nail growth, nail brittleness, and intraocular pressure. J Cosmet Dermatol 2017; 17:263-267. [PMID: 28681570 DOI: 10.1111/jocd.12372] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/26/2017] [Indexed: 11/28/2022]
Abstract
BACKGROUND Bimatoprost has known adnexal activity and was observed to increase nail growth at two clinical centers. OBJECTIVES In this randomized, double-blinded, placebo-controlled pilot study, we examine the effect of bimatoprost (Lumigan 0.01%), applied bid to the proximal nail fold on nail growth, nail brittleness, and intraocular pressure. METHODS Bimatoprost drops were placed on the proximal nail folds of 45 subjects on one hand (medication group) and vehicle drops to the other hand (control group). Baseline and final nail growth measurements, Goldmann applanation tensions of both eyes, and photos at 30 days were performed. Nail brittleness was subjectively graded. RESULTS For the 38 subjects completing the study, the final mean nail growth of the hands, the net individual nail growth of the digits (excluding chipped nails), nail brittleness, and eye pressure readings were NS at P<.05. Photos revealed no increased hirsutism, but one subject with increased skin pigmentation. The drops were well tolerated without adverse effects. Nail chipping was a limitation of the study. CONCLUSIONS Despite the negative results in this pilot study on nail growth and brittleness, further studies with higher bimatoprost concentration (0.03%) are warranted. We recommend monitoring nail growth by etching or marking the nail rather than measuring the full nail length due to our chipped nail findings.
Collapse
Affiliation(s)
- David J Palmer
- Department of Ophthalmology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Andrew Scheman
- Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Angelo P Tanna
- Department of Ophthalmology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Alex Bueno
- University of Illinois at Urbana-Champaign, Urbana, IL, USA
| |
Collapse
|
18
|
El-Yazbi AF, Ibrahim KS, El-Gowelli HM, El-Deeb NM, El-Mas MM. Modulation by NADPH oxidase of the chronic cardiovascular and autonomic interaction between cyclosporine and NSAIDs in female rats. Eur J Pharmacol 2017; 806:96-104. [DOI: 10.1016/j.ejphar.2017.04.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Revised: 04/11/2017] [Accepted: 04/13/2017] [Indexed: 12/23/2022]
|
19
|
Roshandel D, Klein R, Klein BEK, Wolffenbuttel BHR, van der Klauw MM, van Vliet-Ostaptchouk JV, Atzmon G, Ben-Avraham D, Crandall JP, Barzilai N, Bull SB, Canty AJ, Hosseini SM, Hiraki LT, Maynard J, Sell DR, Monnier VM, Cleary PA, Braffett BH, Paterson AD. New Locus for Skin Intrinsic Fluorescence in Type 1 Diabetes Also Associated With Blood and Skin Glycated Proteins. Diabetes 2016; 65:2060-71. [PMID: 27207532 PMCID: PMC4915582 DOI: 10.2337/db15-1484] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 04/06/2016] [Indexed: 12/27/2022]
Abstract
Skin fluorescence (SF) noninvasively measures advanced glycation end products (AGEs) in the skin and is a risk indicator for diabetes complications. N-acetyltransferase 2 (NAT2) is the only known locus influencing SF. We aimed to identify additional genetic loci influencing SF in type 1 diabetes (T1D) through a meta-analysis of genome-wide association studies (N = 1,359) including Diabetes Control and Complications Trial/Epidemiology of Diabetes Interventions and Complications (DCCT/EDIC) and Wisconsin Epidemiologic Study of Diabetic Retinopathy (WESDR). A locus on chromosome 1, rs7533564 (P = 1.9 × 10(-9)), was associated with skin intrinsic fluorescence measured by SCOUT DS (excitation 375 nm, emission 435-655 nm), which remained significant after adjustment for time-weighted HbA1c (P = 1.7 × 10(-8)). rs7533564 was associated with mean HbA1c in meta-analysis (P = 0.0225), mean glycated albumin (P = 0.0029), and glyoxal hydroimidazolones (P = 0.049), an AGE measured in skin biopsy collagen, in DCCT. rs7533564 was not associated with diabetes complications in DCCT/EDIC or with SF in subjects without diabetes (nondiabetic [ND]) (N = 8,721). In conclusion, we identified a new locus associated with SF in T1D subjects that did not show similar effect in ND subjects, suggesting a diabetes-specific effect. This association needs to be investigated in type 2 diabetes.
Collapse
Affiliation(s)
- Delnaz Roshandel
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Ronald Klein
- Department of Ophthalmology and Visual Sciences, University of Wisconsin, Madison, WI
| | - Barbara E K Klein
- Department of Ophthalmology and Visual Sciences, University of Wisconsin, Madison, WI
| | - Bruce H R Wolffenbuttel
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Melanie M van der Klauw
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Jana V van Vliet-Ostaptchouk
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Gil Atzmon
- Departments of Medicine and Genetics, Institute for Aging Research and the Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY Department of Natural Science, University of Haifa, Haifa, Israel
| | - Danny Ben-Avraham
- Departments of Medicine and Genetics, Institute for Aging Research and the Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY
| | - Jill P Crandall
- Departments of Medicine and Genetics, Institute for Aging Research and the Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY
| | - Nir Barzilai
- Departments of Medicine and Genetics, Institute for Aging Research and the Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY
| | - Shelley B Bull
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Angelo J Canty
- Department of Mathematics and Statistics, McMaster University, Hamilton, Ontario, Canada
| | - S Mohsen Hosseini
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Linda T Hiraki
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | | | - David R Sell
- Department of Pathology, Case Western Reserve University, Cleveland, OH
| | - Vincent M Monnier
- Department of Pathology, Case Western Reserve University, Cleveland, OH Department of Biochemistry, Case Western Reserve University, Cleveland, OH
| | | | | | | | - Andrew D Paterson
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
20
|
Li Y, Han L, Ding WY, Ti Y, Li YH, Tang MX, Wang ZH, Zhang Y, Zhang W, Zhong M. Prostaglandin F2α receptor silencing attenuates vascular remodeling in rats with type 2 diabetes. Exp Mol Pathol 2015; 99:517-23. [DOI: 10.1016/j.yexmp.2015.09.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 09/11/2015] [Accepted: 09/21/2015] [Indexed: 12/20/2022]
|
21
|
Dissecting fibrosis: therapeutic insights from the small-molecule toolbox. Nat Rev Drug Discov 2015; 14:693-720. [PMID: 26338155 DOI: 10.1038/nrd4592] [Citation(s) in RCA: 171] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Fibrosis, which leads to progressive loss of tissue function and eventual organ failure, has been estimated to contribute to ~45% of deaths in the developed world, and so new therapeutics to modulate fibrosis are urgently needed. Major advances in our understanding of the mechanisms underlying pathological fibrosis are supporting the search for such therapeutics, and the recent approval of two anti-fibrotic drugs for idiopathic pulmonary fibrosis has demonstrated the tractability of this area for drug discovery. This Review examines the pharmacology and structural information for small molecules being evaluated for lung, liver, kidney and skin fibrosis. In particular, we discuss the insights gained from the use of these pharmacological tools, and how these entities can inform, and probe, emerging insights into disease mechanisms, including the potential for future drug combinations.
Collapse
|
22
|
Lee YK, Lee JY, Moon JI, Park MH. Effectiveness of the ICare rebound tonometer in patients with overestimated intraocular pressure due to tight orbit syndrome. Jpn J Ophthalmol 2014; 58:496-502. [DOI: 10.1007/s10384-014-0343-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Accepted: 07/10/2014] [Indexed: 11/30/2022]
|
23
|
Li L, Li N, Pang W, Zhang X, Hammock BD, Ai D, Zhu Y. Opposite effects of gene deficiency and pharmacological inhibition of soluble epoxide hydrolase on cardiac fibrosis. PLoS One 2014; 9:e94092. [PMID: 24718617 PMCID: PMC3981766 DOI: 10.1371/journal.pone.0094092] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2013] [Accepted: 03/10/2014] [Indexed: 01/09/2023] Open
Abstract
Arachidonic acid-derived epoxyeicosatrienoic acids (EETs) are important regulators of cardiac remodeling; manipulation of their levels is a potentially useful pharmacological strategy. EETs are hydrolyzed by soluble epoxide hydrolase (sEH) to form the corresponding diols, thus altering and reducing the activity of these oxylipins. To better understand the phenotypic impact of sEH disruption, we compared the effect of EPHX2 gene knockout (EPHX2-/-) and sEH inhibition in mouse models. Measurement of plasma oxylipin profiles confirmed that the ratio of EETs/DHETs was increased in EPHX2-/- and sEH-inhibited mice. However, plasma concentrations of 9, 11, 15, 19-HETE were elevated in EPHX2-/- but not sEH-inhibited mice. Next, we investigated the role of this difference in cardiac dysfunction induced by Angiotensin II (AngII). Both EPHX2 gene deletion and inhibition protected against AngII-induced cardiac hypertrophy. Interestingly, cardiac dysfunction was attenuated by sEH inhibition rather than gene deletion. Histochemical staining revealed that compared with pharmacological inhibition, EPHX2 deletion aggravated AngII-induced myocardial fibrosis; the mRNA levels of fibrotic-related genes were increased. Furthermore, cardiac inflammatory response was greater in EPHX2-/- than sEH-inhibited mice with AngII treatment, as evidenced by increased macrophage infiltration and expression of MCP-1 and IL-6. In vitro, AngII-upregulated MCP-1 and IL-6 expression was significantly attenuated by sEH inhibition but promoted by EPHX2 deletion in cardiofibroblasts. Thus, compared with pharmacological inhibition of sEH, EPHX2 deletion caused the shift in arachidonic acid metabolism, which may led to pathological cardiac remodeling, especially cardiac fibrosis.
Collapse
Affiliation(s)
- Lijuan Li
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China
| | - Nan Li
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China
| | - Wei Pang
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China
| | - Xu Zhang
- Department of Physiology, Tianjin Medical University, Tianjin, China
| | - Bruce D. Hammock
- Department of Entomology and Comprehensive Cancer Center, University of California Davis, Davis, California, United States of America
| | - Ding Ai
- Department of Physiology, Tianjin Medical University, Tianjin, China
- * E-mail: (YZ); (DA)
| | - Yi Zhu
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China
- Department of Physiology, Tianjin Medical University, Tianjin, China
- * E-mail: (YZ); (DA)
| |
Collapse
|
24
|
Ding WY, Liu L, Wang ZH, Tang MX, Ti Y, Han L, Zhang L, Zhang Y, Zhong M, Zhang W. FP-receptor gene silencing ameliorates myocardial fibrosis and protects from diabetic cardiomyopathy. J Mol Med (Berl) 2014; 92:629-40. [PMID: 24500109 DOI: 10.1007/s00109-013-1119-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Revised: 12/02/2013] [Accepted: 12/06/2013] [Indexed: 11/25/2022]
Abstract
UNLABELLED Prostaglandin F2(α)-F-prostanoid (PGF2(α)-FP) receptor is closely related to insulin resistance, which plays a causal role in the pathogenesis of diabetic cardiomyopathy (DCM). We sought to reveal whether PGF2(α)-FP receptor plays an important part in modulating DCM and the mechanisms involved. We established the type 2 diabetes rat model by high-fat diet and low-dose streptozotocin (STZ) and then evaluated its characteristics by metabolite tests, Western blot analysis for FP-receptor expression, histopathologic analyses of cardiomyocyte density and fibrosis area. Next, we used gene silencing to investigate the role of FP receptor in the pathophysiologic features of DCM. Our study showed elevated cholesterol, triglyceride, glucose, and insulin levels, severe insulin resistance, and FP-receptor overexpression in diabetic rats. The collagen volume fraction (CVF) and perivascular collagen area/luminal area (PVCA/LA) were higher in the diabetic group than the control group (CVF% 10.99 ± 0.99 vs 1.59 ± 0.18, P < 0.05; PVCA/LA% 17.07 ± 2.61 vs 2.86 ± 0.69, P < 0.05). We found that the silencing of FP receptor decreased cholesterol, triglyceride, glucose, and insulin levels and ameliorated insulin resistance. The CVF and PVCF/LA were significantly downregulated in FP-receptor short hairpin RNA (shRNA) treatment group (FP-receptor shRNA group vs vehicle group: CVF% 5.59 ± 0.92 vs 10.97 ± 1.33, P < 0.05, PVCA/LA% 4.74 ± 1.57 vs 14.79 ± 2.22, P < 0.05; FP-receptor shRNA + PGF2(α) group vs vehicle group : CVF% 5.19 ± 0.79 vs 10.97 ± 1.33, P < 0.05, PVCA/LA% 5.96 ± 1.15 vs 14.79 ± 2.22, P < 0.05, respectively). Furthermore, with FP-receptor gene silencing, the activated protein kinase C (PKC) and Rho kinase were significantly decreased, and the blunted phosphorylation of Akt was restored. FP-receptor gene silencing may exert a protective effect on DCM by improving myocardial fibrosis, suggesting a new therapeutic approach for human DCM. KEY MESSAGES FP-receptor gene silencing improves glucose tolerance and insulin resistance in type 2 diabetes (T2D). FP-receptor gene silencing modulates the activities of PKC/Rho and Akt signaling pathways in T2D. FP-receptor gene silencing decreases collagen expression and ameliorates myocardial fibrosis in T2D. FP-receptor gene silencing protects from diabetic cardiomyopathy in T2D.
Collapse
Affiliation(s)
- Wen-yuan Ding
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital of Shandong University, Jinan, 250012, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Regulation of prostaglandin F2α against β amyloid clearance and its inflammation induction through LXR/RXR heterodimer antagonism in microglia. Prostaglandins Other Lipid Mediat 2013; 106:45-52. [DOI: 10.1016/j.prostaglandins.2013.09.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Revised: 09/04/2013] [Accepted: 09/17/2013] [Indexed: 12/13/2022]
|
26
|
Bagnato G, Bitto A, Pizzino G, Irrera N, Sangari D, Cinquegrani M, Roberts WN, Matucci Cerinic M, Squadrito F, Altavilla D, Bagnato G, Saitta A. Simvastatin attenuates the development of pulmonary and cutaneous fibrosis in a murine model of systemic sclerosis. Rheumatology (Oxford) 2013; 52:1377-86. [DOI: 10.1093/rheumatology/ket144] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
27
|
Bagnato G, Bitto A, Irrera N, Pizzino G, Sangari D, Cinquegrani M, Roberts WN, Atteritano M, Altavilla D, Squadrito F, Bagnato G, Saitta A. Propylthiouracil prevents cutaneous and pulmonary fibrosis in the reactive oxygen species murine model of systemic sclerosis. Arthritis Res Ther 2013; 15:R120. [PMID: 24286160 PMCID: PMC3978728 DOI: 10.1186/ar4300] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Accepted: 09/16/2013] [Indexed: 12/16/2022] Open
Abstract
INTRODUCTION Recent advances suggest that the cellular redox state may play a significant role in the progression of fibrosis in systemic sclerosis (SSc). Another, and as yet poorly accounted for, feature of SSc is its overlap with thyroid abnormalities. Previous reports demonstrate that hypothyroidism reduces oxidant stress. The aim of this study was therefore to evaluate the effect of propylthiouracil (PTU), and of the hypothyroidism induced by it, on the development of cutaneous and pulmonary fibrosis in the oxidant stress murine model of SSc. METHODS Chronic oxidant stress SSc was induced in BALB/c mice by daily subcutaneous injections of hypochlorous acid (HOCl) for 6 weeks. Mice (n = 25) were randomized into three arms: HOCl (n = 10), HOCl plus PTU (n = 10) or vehicle alone (n = 5). PTU administration was initiated 30 minutes after HOCl subcutaneous injection and continued daily for 6 weeks. Skin and lung fibrosis were evaluated by histologic methods. Immunohistochemical staining for alpha-smooth muscle actin (α-SMA) in cutaneous and pulmonary tissues was performed to evaluate myofibroblast differentiation. Lung and skin concentrations of vascular endothelial growth factor (VEGF), extracellular signal-related kinase (ERK), rat sarcoma protein (Ras), Ras homolog gene family (Rho), and transforming growth factor (TGF) β were analyzed by Western blot. RESULTS Injections of HOCl induced cutaneous and lung fibrosis in BALB/c mice. PTU treatment prevented both dermal and pulmonary fibrosis. Myofibroblast differentiation was also inhibited by PTU in the skin and lung. The increase in cutaneous and pulmonary expression of VEGF, ERK, Ras, and Rho in mice treated with HOCl was significantly prevented in mice co-administered with PTU. CONCLUSIONS PTU, probably through its direct effect on reactive oxygen species or indirectly through thyroid function inhibition, prevents the development of cutaneous and pulmonary fibrosis by blocking the activation of the Ras-ERK pathway in the oxidant-stress animal model of SSc.
Collapse
Affiliation(s)
- Gianluca Bagnato
- Department of Clinical and Experimental Medicine, Division of Internal
Medicine, University of Messina, Via Consolare Valeria n°1, 98100,
Messina, Italy
| | - Alessandra Bitto
- Department of Clinical and Experimental Medicine, Division of Pharmacology,
University of Messina, Via Consolare Valeria n°1, 98100, Messina,
Italy
| | - Natasha Irrera
- Department of Clinical and Experimental Medicine, Division of Pharmacology,
University of Messina, Via Consolare Valeria n°1, 98100, Messina,
Italy
| | - Gabriele Pizzino
- Department of Clinical and Experimental Medicine, Division of Pharmacology,
University of Messina, Via Consolare Valeria n°1, 98100, Messina,
Italy
| | - Donatella Sangari
- Department of Clinical and Experimental Medicine, Division of Rheumatology,
University of Messina, Via Consolare Valeria n°1, 98100, Messina,
Italy
| | - Maurizio Cinquegrani
- Department of Clinical and Experimental Medicine, Division of Internal
Medicine, University of Messina, Via Consolare Valeria n°1, 98100,
Messina, Italy
| | - William Neal Roberts
- Department of Internal Medicine, Division of Rheumatology, University of
Louisville, Louisville, KY 40292, Kentucky, USA
| | - Marco Atteritano
- Department of Clinical and Experimental Medicine, Division of Rheumatology,
University of Messina, Via Consolare Valeria n°1, 98100, Messina,
Italy
| | - Domenica Altavilla
- Department of Clinical and Experimental Medicine, Division of Pharmacology,
University of Messina, Via Consolare Valeria n°1, 98100, Messina,
Italy
| | - Francesco Squadrito
- Department of Clinical and Experimental Medicine, Division of Pharmacology,
University of Messina, Via Consolare Valeria n°1, 98100, Messina,
Italy
| | - Gianfilippo Bagnato
- Department of Clinical and Experimental Medicine, Division of Rheumatology,
University of Messina, Via Consolare Valeria n°1, 98100, Messina,
Italy
| | - Antonino Saitta
- Department of Clinical and Experimental Medicine, Division of Internal
Medicine, University of Messina, Via Consolare Valeria n°1, 98100,
Messina, Italy
| |
Collapse
|
28
|
Oga T, Handa T, Mishima M, Chin K, Narumiya S. Roles of eicosanoids in pulmonary fibrosis. Inflamm Regen 2013. [DOI: 10.2492/inflammregen.33.109] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|