1
|
Russo RC, Ryffel B. The Chemokine System as a Key Regulator of Pulmonary Fibrosis: Converging Pathways in Human Idiopathic Pulmonary Fibrosis (IPF) and the Bleomycin-Induced Lung Fibrosis Model in Mice. Cells 2024; 13:2058. [PMID: 39768150 PMCID: PMC11674266 DOI: 10.3390/cells13242058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 12/06/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic and lethal interstitial lung disease (ILD) of unknown origin, characterized by limited treatment efficacy and a fibroproliferative nature. It is marked by excessive extracellular matrix deposition in the pulmonary parenchyma, leading to progressive lung volume decline and impaired gas exchange. The chemokine system, a network of proteins involved in cellular communication with diverse biological functions, plays a crucial role in various respiratory diseases. Chemokine receptors trigger the activation, proliferation, and migration of lung-resident cells, including pneumocytes, endothelial cells, alveolar macrophages, and fibroblasts. Around 50 chemokines can potentially interact with 20 receptors, expressed by both leukocytes and non-leukocytes such as tissue parenchyma cells, contributing to processes such as leukocyte mobilization from the bone marrow, recirculation through lymphoid organs, and tissue influx during inflammation or immune response. This narrative review explores the complexity of the chemokine system in the context of IPF and the bleomycin-induced lung fibrosis mouse model. The goal is to identify specific chemokines and receptors as potential therapeutic targets. Recent progress in understanding the role of the chemokine system during IPF, using experimental models and molecular diagnosis, underscores the complex nature of this system in the context of the disease. Despite advances in experimental models and molecular diagnostics, discovering an effective therapy for IPF remains a significant challenge in both medicine and pharmacology. This work delves into microarray results from lung samples of IPF patients and murine samples at different stages of bleomycin-induced pulmonary fibrosis. By discussing common pathways identified in both IPF and the experimental model, we aim to shed light on potential targets for therapeutic intervention. Dysregulation caused by abnormal chemokine levels observed in IPF lungs may activate multiple targets, suggesting that chemokine signaling plays a central role in maintaining or perpetuating lung fibrogenesis. The highlighted chemokine axes (CCL8-CCR2, CCL19/CCL21-CCR7, CXCL9-CXCR3, CCL3/CCL4/CCL5-CCR5, and CCL20-CCR6) present promising opportunities for advancing IPF treatment research and uncovering new pharmacological targets within the chemokine system.
Collapse
Affiliation(s)
- Remo Castro Russo
- Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais-UFMG, Belo Horizonte 31270-901, MG, Brazil
| | - Bernhard Ryffel
- Laboratory of Immuno-Neuro Modulation (INEM), UMR7355 Centre National de la Recherche Scientifique (CNRS), University of Orleans, 45071 Orleans, France
| |
Collapse
|
2
|
Juvkam IS, Zlygosteva O, Sitarz M, Sørensen BS, Aass HCD, Edin NJ, Galtung HK, Søland TM, Malinen E. Proton- compared to X-irradiation leads to more acinar atrophy and greater hyposalivation accompanied by a differential cytokine response. Sci Rep 2024; 14:22311. [PMID: 39333378 PMCID: PMC11437014 DOI: 10.1038/s41598-024-73110-7] [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: 03/20/2024] [Accepted: 09/13/2024] [Indexed: 09/29/2024] Open
Abstract
Proton therapy gives less dose to healthy tissue compared to conventional X-ray therapy, but systematic comparisons of normal tissue responses are lacking. The aim of this study was to investigate late tissue responses in the salivary glands following proton- or X-irradiation of the head and neck in mice. Moreover, we aimed at investigating molecular responses by monitoring the cytokine levels in serum and saliva. Female C57BL/6J mice underwent local fractionated irradiation with protons or X-rays to the maximally tolerated acute level. Saliva and serum were collected before and at different time points after irradiation to assess salivary gland function and cytokine expression. To study late responses in the major salivary glands, histological analyses were performed on tissues collected at day 105 after onset of irradiation. Saliva volume after proton and X-irradiation was significantly lower than for controls and remained reduced at all time points after irradiation. Protons caused reduced saliva production and fewer acinar cells in the submandibular glands compared to X-rays at day 105. X-rays induced a stronger inflammatory cytokine response in saliva compared to protons. This work supports previous preclinical findings and indicate that the relative biological effectiveness of protons in normal tissue might be higher than the commonly used value of 1.1.
Collapse
Affiliation(s)
- Inga Solgård Juvkam
- Institute of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway
- Department of Radiation Biology, Institute of Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Olga Zlygosteva
- Department of Physics, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | - Mateusz Sitarz
- Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark
| | - Brita Singers Sørensen
- Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Hans Christian D Aass
- The Blood Cell Research Group, Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| | - Nina Jeppesen Edin
- Department of Physics, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | - Hilde Kanli Galtung
- Institute of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - Tine Merete Søland
- Institute of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway
- Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Eirik Malinen
- Department of Radiation Biology, Institute of Cancer Research, Oslo University Hospital, Oslo, Norway.
- Department of Physics, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway.
| |
Collapse
|
3
|
Hamdi E, Bekhit AA, Higazi A, Ahmed ABF, Hussein Kasem A, Najim MA, Alshammari TM, Thabet K. Interferon-λ3 rs12979860 can regulate inflammatory cytokines production in pulmonary fibrosis. Saudi Pharm J 2023; 31:101816. [PMID: 37876736 PMCID: PMC10590737 DOI: 10.1016/j.jsps.2023.101816] [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: 06/19/2023] [Accepted: 10/01/2023] [Indexed: 10/26/2023] Open
Abstract
Pulmonary fibrosis (PF) is the last phase of interstitial lung diseases (ILDs), which are a collection of pulmonary illnesses marked by parenchymal remodeling and scarring. Treatment can only halt the functional decline of the lung, raising the necessity of identifying the basic processes implicated in lung fibrogenesis. The Interferon lambda-3 (IFNL3) gene variant, rs12979860, was determined to be related to an elevated risk of fibrosis in different organs, but the mechanism through which it mediates fibrogenesis is not clear. In the current research, we aim to figure out some of the mechanistic pathways by which IFN-λ3 mediates ILDs. 100 healthy controls and 74 ILD patients were genotyped for IFNL3 rs12979860. Then the mRNA expression of IFNL3 and some other proinflammatory mediators was examined according to genotype in the peripheral blood mononuclear cells (PBMCs) of ILDs patients. The IFNL3 rs12979860 genotype distribution of healthy individuals and ILDs patients was shown to be in Hardy-Weinberg equilibrium (HWE) with a minor allele frequency (MAF) of 0.293 and 0.326, respectively. Furthermore, the CC genotype was demonstrated to be linked to enhanced IFNL3 expression. Also, the CC genotype was linked to an increase in the mRNA expression of TLR4 (P = 0.03) and the inflammatory cytokines IL-1β and TNF-α (P = 0.01 and 0.04, respectively) and had no effect on the NF-kB level (P = 0.3). From these results, we can deduce that IFN-λ3 may mediate tissue fibrosis via increasing the expression of IFN-λ3 itself and other proinflammatory mediators. This stimulates a self-sustaining loop mechanism which includes a reciprocal production of IFN-λ3, TLR4, IL-1β, and TNF-α leading to persistent inflammation and fibrosis.
Collapse
Affiliation(s)
- Eman Hamdi
- Department of Biochemistry, Faculty of Pharmacy, Minia University, Minia 61519, Egypt
| | - Amany A. Bekhit
- Department of Biochemistry, Faculty of Pharmacy, Minia University, Minia 61519, Egypt
| | - Aliaa Higazi
- Department of Clinical Pathology, Faculty of Medicine, Minia University, Minia 61519, Egypt
| | - Abo Bakr F. Ahmed
- Department of Microbiology and Immunology, Faculty of Pharmacy, Minia University, Minia 61519, Egypt
| | - Ahmed Hussein Kasem
- Department of Chest Diseases, Faculty of Medicine, Minia University, Minia 61519, Egypt
| | - Mustafa A.M. Najim
- Department of Medical Laboratories Technology, Faculty of Applied Medical Sciences, Taibah University, Medina, Saudi Arabia
| | - Thamir M. Alshammari
- Medication Safety Research Chair, King Saud University, Riyadh, Saudi Arabia
- College of pharmacy, Almaarefa University, Riyadh, Saudi Arabia
| | - Khaled Thabet
- Department of Biochemistry, Faculty of Pharmacy, Minia University, Minia 61519, Egypt
| |
Collapse
|
4
|
Mehryab F, Taghizadeh F, Goshtasbi N, Merati F, Rabbani S, Haeri A. Exosomes as cutting-edge therapeutics in various biomedical applications: An update on engineering, delivery, and preclinical studies. Biochimie 2023; 213:139-167. [PMID: 37207937 DOI: 10.1016/j.biochi.2023.05.010] [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: 12/24/2022] [Revised: 04/29/2023] [Accepted: 05/16/2023] [Indexed: 05/21/2023]
Abstract
Exosomes are cell-derived nanovesicles, circulating in different body fluids, and acting as an intercellular mechanism. They can be purified from culture media of different cell types and carry an enriched content of various protein and nucleic acid molecules originating from their parental cells. It was indicated that the exosomal cargo can mediate immune responses via many signaling pathways. Over recent years, the therapeutic effects of various exosome types were broadly investigated in many preclinical studies. Herein, we present an update on recent preclinical studies on exosomes as therapeutic and/or delivery agents for various applications. The exosome origin, structural modifications, natural or loaded active ingredients, size, and research outcomes were summarized for various diseases. Overall, the present article provides an overview of the latest exosome research interests and developments to clear the way for the clinical study design and application.
Collapse
Affiliation(s)
- Fatemeh Mehryab
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Taghizadeh
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nazanin Goshtasbi
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Faezeh Merati
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shahram Rabbani
- Research Center for Advanced Technologies in Cardiovascular Medicine, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Azadeh Haeri
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Protein Technology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Jahankhani K, Ahangari F, Adcock IM, Mortaz E. Possible cancer-causing capacity of COVID-19: Is SARS-CoV-2 an oncogenic agent? Biochimie 2023; 213:130-138. [PMID: 37230238 PMCID: PMC10202899 DOI: 10.1016/j.biochi.2023.05.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 04/24/2023] [Accepted: 05/22/2023] [Indexed: 05/27/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has shown diverse life-threatening effects, most of which are considered short-term. In addition to its short-term effects, which has claimed many millions of lives since 2019, the long-term complications of this virus are still under investigation. Similar to many oncogenic viruses, it has been hypothesized that SARS-CoV-2 employs various strategies to cause cancer in different organs. These include leveraging the renin angiotensin system, altering tumor suppressing pathways by means of its nonstructural proteins, and triggering inflammatory cascades by enhancing cytokine production in the form of a "cytokine storm" paving the way for the emergence of cancer stem cells in target organs. Since infection with SARS-CoV-2 occurs in several organs either directly or indirectly, it is expected that cancer stem cells may develop in multiple organs. Thus, we have reviewed the impact of coronavirus disease 2019 (COVID-19) on the vulnerability and susceptibility of specific organs to cancer development. It is important to note that the cancer-related effects of SARS-CoV-2 proposed in this article are based on the ability of the virus and its proteins to cause cancer but that the long-term consequences of this infection will only be illustrated in the long run.
Collapse
Affiliation(s)
- Kasra Jahankhani
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Ahangari
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ian M Adcock
- Airways Disease, National Heart and Lung Institute, Imperial College London, London, United Kingdom; Immune Health Program at Hunter Medical Research Institute and the College of Health and Medicine at the University of Newcastle, Australia
| | - Esmaeil Mortaz
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
6
|
He X, Jarrell ZR, Smith MR, Ly VT, Hu X, Sueblinvong V, Liang Y, Orr M, Go YM, Jones DP. Low-dose vanadium pentoxide perturbed lung metabolism associated with inflammation and fibrosis signaling in male animal and in vitro models. Am J Physiol Lung Cell Mol Physiol 2023; 325:L215-L232. [PMID: 37310758 PMCID: PMC10396228 DOI: 10.1152/ajplung.00303.2022] [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: 09/22/2022] [Revised: 05/25/2023] [Accepted: 05/30/2023] [Indexed: 06/14/2023] Open
Abstract
Vanadium is available as a dietary supplement and also is known to be toxic if inhaled, yet little information is available concerning the effects of vanadium on mammalian metabolism when concentrations found in food and water. Vanadium pentoxide (V+5) is representative of the most common dietary and environmental exposures, and prior research shows that low-dose V+5 exposure causes oxidative stress measured by glutathione oxidation and protein S-glutathionylation. We examined the metabolic impact of V+5 at relevant dietary and environmental doses (0.01, 0.1, and 1 ppm for 24 h) in human lung fibroblasts (HLFs) and male C57BL/6J mice (0.02, 0.2, and 2 ppm in drinking water for 7 mo). Untargeted metabolomics using liquid chromatography-high-resolution mass spectrometry (LC-HRMS) showed that V+5 induced significant metabolic perturbations in both HLF cells and mouse lungs. We noted 30% of the significantly altered pathways in HLF cells, including pyrimidines and aminosugars, fatty acids, mitochondrial and redox pathways, showed similar dose-dependent patterns in mouse lung tissues. Alterations in lipid metabolism included leukotrienes and prostaglandins involved in inflammatory signaling, which have been associated with the pathogenesis of idiopathic pulmonary fibrosis (IPF) and other disease processes. Elevated hydroxyproline levels and excessive collagen deposition were also present in lungs from V+5-treated mice. Taken together, these results show that oxidative stress from environmental V+5, ingested at low levels, could alter metabolism to contribute to common human lung diseases.NEW & NOTEWORTHY We used relevant dietary and environmental doses of Vanadium pentoxide (V+5) to examine its metabolic impact in vitro and in vivo. Using liquid chromatography-high-resolution mass spectrometry (LC-HRMS), we found significant metabolic perturbations, with similar dose-dependent patterns observed in human lung fibroblasts and male mouse lungs. Alterations in lipid metabolism included inflammatory signaling, elevated hydroxyproline levels, and excessive collagen deposition were present in V+5-treated lungs. Our findings suggest that low levels of V+5 could trigger pulmonary fibrotic signaling.
Collapse
Affiliation(s)
- Xiaojia He
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, Georgia, United States
| | - Zachery R Jarrell
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, Georgia, United States
| | - Matthew Ryan Smith
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, Georgia, United States
- Atlanta Department of Veterans Affairs Healthcare System, Decatur, Georgia, United States
| | - ViLinh Thi Ly
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, Georgia, United States
| | - Xin Hu
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, Georgia, United States
| | - Viranuj Sueblinvong
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, Georgia, United States
| | - Yongliang Liang
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, Georgia, United States
| | - Michael Orr
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, Georgia, United States
| | - Young-Mi Go
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, Georgia, United States
| | - Dean P Jones
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, Georgia, United States
| |
Collapse
|
7
|
Hegazy W, Sakr HI, Abdul Hamid M, Abdelaziz MA, Salah M, Abdel Rehiem ES, Abdel Moneim A. Hesperidin Attenuates Hypothyroidism-Induced Lung Damage in Adult Albino Rats by Modulating Oxidative Stress, Nuclear Factor Kappa-B Pathway, Proliferating Cell Nuclear Antigen and Inflammatory Cytokines. Biomedicines 2023; 11:1570. [PMID: 37371665 DOI: 10.3390/biomedicines11061570] [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: 03/23/2023] [Revised: 05/19/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
The occurrence of worsening pulmonary function has been connected to hypothyroidism (HPO). Hesperidin (HES) was suggested to have antioxidant, anti-proliferative, and anti-inflammatory potential. Our study's objective was to determine whether HES could reduce carbimazole (CBZ)-induced lung injury more effectively than Eltroxin (ELT) in adult male albino rats or not. At random, 32 rats were distributed into four groups: Group I: normal control, to induce HPO, the remaining three groups were given CBZ (20 mg/kg/day) dissolved in distilled water for 1 week. They were then split up into three groups. Group II: orally administered CBZ (20 mg/kg b.w in water/day), Group III: HES (200 mg/kg/day) dissolved in 1% carboxymethyl-cellulose + CBZ treated, and Group IV: ELT (0.045 mg/kg/day) dissolved in distilled water + CBZ treated. All treatments were delivered for 12 weeks. Blood was collected to assess thyroid-stimulating hormone (TSH) and thyroid hormones (THs). Lung injury was evaluated based on the pulmonary content of interleukin (IL)-35, IL-6, and tumor necrosis factor-alpha (TNF-α), along with the estimation of lipid peroxidation, catalase, glutathione levels, superoxide dismutase, heme oxygenase-1 (HO-1), and nuclear factor erythroid 2-related factor 2 (Nrf2). The histological, ultrastructural, and immunohistochemical study of nuclear factor Kappa-B (NF-κB) and inducible nitric oxide synthase (iNOS), together with estimating the proliferation of cells using Antigen Ki-67 in lung tissue were performed. HES and ELT primarily suppressed variable lung damage mechanisms by suppressing TSH, the NF-κB/TNF-α pathway, iNOS, lipid peroxidation, Ki-67, and inflammatory mediators. On the other hand, they improved THs, antioxidant parameters, and the Nrf2/HO-1 pathway. HES and ELT exhibited an ameliorative effect that was reflected in the histopathological, immunohistochemical, and ultrastructural results. These results indicate that HES is a pneumoprotective agent that could be a promising treatment for oxidative stress, inflammation, and proliferation.
Collapse
Affiliation(s)
- Walaa Hegazy
- Histology Division, Basic Science Department, Faculty of Physical Therapy, Nahda University, Beni-Suef 62511, Egypt
| | - Hader I Sakr
- Department of Medical Physiology, Faculty of Medicine, Cairo University, Cairo 11562, Egypt
- Department of Medical Physiology, Medicine Program, Batterjee Medical College, Jeddah 21442, Saudi Arabia
| | - Manal Abdul Hamid
- Cell Biology, Histology and Genetics Division, Zoology Department, Faculty of Science, Beni-Suef University, Salah Salem St., Beni-Suef 62511, Egypt
| | - Mohamed A Abdelaziz
- Basic Medical Sciences Department, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
- Medical Physiology Department, Faculty of Medicine, Al-Azhar University, Cairo 11651, Egypt
| | - Marwa Salah
- Cell Biology, Histology and Genetics Division, Zoology Department, Faculty of Science, Beni-Suef University, Salah Salem St., Beni-Suef 62511, Egypt
| | - Eman S Abdel Rehiem
- Molecular Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, Salah Salem St., Beni-Suef 62511, Egypt
| | - Adel Abdel Moneim
- Molecular Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, Salah Salem St., Beni-Suef 62511, Egypt
| |
Collapse
|
8
|
The Global Prevalence of Pulmonary Fibrosis Among Post–COVID-19 Follow-up Patients. INFECTIOUS DISEASES IN CLINICAL PRACTICE 2023. [DOI: 10.1097/ipc.0000000000001190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
9
|
Begum R, Mamun-Or-Rashid ANM, Lucy TT, Pramanik MK, Sil BK, Mukerjee N, Tagde P, Yagi M, Yonei Y. Potential Therapeutic Approach of Melatonin against Omicron and Some Other Variants of SARS-CoV-2. Molecules 2022; 27:6934. [PMID: 36296527 PMCID: PMC9609612 DOI: 10.3390/molecules27206934] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/03/2022] [Accepted: 10/11/2022] [Indexed: 11/06/2022] Open
Abstract
The Omicron variant (B.529) of COVID-19 caused disease outbreaks worldwide because of its contagious and diverse mutations. To reduce these outbreaks, therapeutic drugs and adjuvant vaccines have been applied for the treatment of the disease. However, these drugs have not shown high efficacy in reducing COVID-19 severity, and even antiviral drugs have not shown to be effective. Researchers thus continue to search for an effective adjuvant therapy with a combination of drugs or vaccines to treat COVID-19 disease. We were motivated to consider melatonin as a defensive agent against SARS-CoV-2 because of its various unique properties. Over 200 scientific publications have shown the significant effects of melatonin in treating diseases, with strong antioxidant, anti-inflammatory, and immunomodulatory effects. Melatonin has a high safety profile, but it needs further clinical trials and experiments for use as a therapeutic agent against the Omicron variant of COVID-19. It might immediately be able to prevent the development of severe symptoms caused by the coronavirus and can reduce the severity of the infection by improving immunity.
Collapse
Affiliation(s)
- Rahima Begum
- Department of Microbiology, Gono Bishwabidyalay, Dhaka 1344, Bangladesh
| | - A. N. M. Mamun-Or-Rashid
- Anti-Aging Medical Research Center, Graduate School of Life and Medical Sciences, Doshisha University 1-3 TataraMiyakodani, Kyoto 610-0394, Japan
- Glycative Stress Research Center, Graduate School of Life and Medical Sciences, Doshisha University 1-3 Tatara Miyakodani, Kyoto 610-0394, Japan
- Department of Environmental & Occupational Health, School of Public Health, University of Pittsburgh, 130 De Soto Str., Pittsburgh, PA 15231, USA
| | - Tanzima Tarannum Lucy
- Anti-Aging Medical Research Center, Graduate School of Life and Medical Sciences, Doshisha University 1-3 TataraMiyakodani, Kyoto 610-0394, Japan
- Glycative Stress Research Center, Graduate School of Life and Medical Sciences, Doshisha University 1-3 Tatara Miyakodani, Kyoto 610-0394, Japan
| | - Md. Kamruzzaman Pramanik
- Microbiology and Industrial Irradiation Division, Institute of Food and Radiation Biology, Atomic Energy Research Establishment, Savar 1349, Bangladesh
| | - Bijon Kumar Sil
- Department of Microbiology, Gono Bishwabidyalay, Dhaka 1344, Bangladesh
| | - Nobendu Mukerjee
- Department of Microbiology, Ramakrishna Mission Vivekananda Centenary College, Kolkata 700118, India
- Department of Health Sciences, Novel Global Community Educational Foundation, Sydney 37729, Australia
| | - Priti Tagde
- Patel College of Pharmacy, Madhyanchal Professional University, Bhopal 462044, India
| | - Masayuki Yagi
- Anti-Aging Medical Research Center, Graduate School of Life and Medical Sciences, Doshisha University 1-3 TataraMiyakodani, Kyoto 610-0394, Japan
- Glycative Stress Research Center, Graduate School of Life and Medical Sciences, Doshisha University 1-3 Tatara Miyakodani, Kyoto 610-0394, Japan
| | - Yoshikazu Yonei
- Anti-Aging Medical Research Center, Graduate School of Life and Medical Sciences, Doshisha University 1-3 TataraMiyakodani, Kyoto 610-0394, Japan
- Glycative Stress Research Center, Graduate School of Life and Medical Sciences, Doshisha University 1-3 Tatara Miyakodani, Kyoto 610-0394, Japan
| |
Collapse
|
10
|
Sehgal M, Jakhete SM, Manekar AG, Sasikumar S. Specific epigenetic regulators serve as potential therapeutic targets in idiopathic pulmonary fibrosis. Heliyon 2022; 8:e09773. [PMID: 36061031 PMCID: PMC9434059 DOI: 10.1016/j.heliyon.2022.e09773] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/27/2022] [Accepted: 06/17/2022] [Indexed: 12/15/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF), a disorder observed mostly in older human beings, is characterised by chronic and progressive lung scarring leading to an irreversible decline in lung function. This health condition has a dismal prognosis and the currently available drugs only delay but fail to reverse the progression of lung damage. Consequently, it becomes imperative to discover improved therapeutic compounds and their cellular targets to cure IPF. In this regard, a number of recent studies have targeted the epigenetic regulation by histone deacetylases (HDACs) to develop and categorise antifibrotic drugs for lungs. Therefore, this review focuses on how aberrant expression or activity of Classes I, II and III HDACs alter TGF-β signalling to promote events such as epithelial-mesenchymal transition, differentiation of activated fibroblasts into myofibroblasts, and excess deposition of the extracellular matrix to propel lung fibrosis. Further, this study describes how certain chemical compounds or dietary changes modulate dysregulated HDACs to attenuate five faulty TGF-β-dependent profibrotic processes, both in animal models and cell lines replicating IPF, thereby identifying promising means to treat this lung disorder.
Collapse
Affiliation(s)
- Manas Sehgal
- Genetics and Molecular Biology Research Centre, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune, Maharashtra, PIN - 411033, India
| | - Sharayu Manish Jakhete
- Genetics and Molecular Biology Research Centre, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune, Maharashtra, PIN - 411033, India
| | - Amruta Ganesh Manekar
- Genetics and Molecular Biology Research Centre, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune, Maharashtra, PIN - 411033, India
| | - Satish Sasikumar
- Genetics and Molecular Biology Research Centre, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune, Maharashtra, PIN - 411033, India
| |
Collapse
|
11
|
Pathak E, Atri N, Mishra R. Single-Cell Transcriptome Analysis Reveals the Role of Pancreatic Secretome in COVID-19 Associated Multi-organ Dysfunctions. Interdiscip Sci 2022; 14:863-878. [PMID: 35394619 PMCID: PMC8990272 DOI: 10.1007/s12539-022-00513-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 03/16/2022] [Accepted: 03/17/2022] [Indexed: 01/09/2023]
Abstract
The SARS-CoV-2 infection affects the lungs, heart, kidney, intestine, olfactory epithelia, liver, and pancreas and brings forward multi-organ dysfunctions (MODs). However, mechanistic details of SARS-CoV-2-induced MODs are unclear. Here, we have investigated the role of pancreatic secretory proteins to mechanistically link COVID-19 with MODs using single-cell transcriptome analysis. Secretory proteins were identified using the Human Protein Atlas. Gene ontology, pathway, and disease enrichment analyses were used to highlight the role of upregulated pancreatic secretory proteins (secretome). We show that SARS-CoV-2 infection shifts the expression profile of pancreatic endocrine cells to acinar and ductal cell-specific profiles, resulting in increased expression of acinar and ductal cell-specific genes. Among all the secretory proteins, the upregulated expression of IL1B, AGT, ALB, SPP1, CRP, SERPINA1, C3, TFRC, TNFSF10, and MIF was mainly associated with disease of diverse organs. Extensive literature and experimental evidence are used to validate the association of the upregulated pancreatic secretome with the coagulation cascade, complement activation, renin-angiotensinogen system dysregulation, endothelial cell injury and thrombosis, immune system dysregulation, and fibrosis. Our finding suggests the influence of an upregulated secretome on multi-organ systems such as nervous, cardiovascular, immune, digestive, and urogenital systems. Our study provides evidence that an upregulated pancreatic secretome is a possible cause of SARS-CoV-2-induced MODs. This finding may have a significant impact on the clinical setting regarding the prevention of SARS-CoV-2-induced MODs.
Collapse
Affiliation(s)
- Ekta Pathak
- Banaras Hindu University, Varanasi, Uttar Pradesh, 221005, India.
| | - Neelam Atri
- Bioinformatics Department, MMV, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
- Department of Botany, MMV, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Rajeev Mishra
- Bioinformatics Department, MMV, Institute of Science, Banaras Hindu University, Varanasi, 221005, India.
| |
Collapse
|
12
|
Bergantini L, Mainardi A, d’Alessandro M, Cameli P, Bennett D, Bargagli E, Sestini P. Common Molecular Pathways Between Post-COVID19 Syndrome and Lung Fibrosis: A Scoping Review. Front Pharmacol 2022; 13:748931. [PMID: 35308222 PMCID: PMC8931519 DOI: 10.3389/fphar.2022.748931] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 02/14/2022] [Indexed: 01/18/2023] Open
Abstract
The pathogenetic mechanism of post-Covid-19 pulmonary fibrosis is currently a topic of intense research interest, but still largely unexplored. The aim of this work was to carry out a systematic exploratory search of the literature (Scoping review) to identify and systematize the main pathogenetic mechanisms that are believed to be involved in this phenomenon, in order to highlight the same molecular aspect of the lung. These aims could be essential in the future for therapeutic management. We identified all primary studies involving in post COVID19 syndrome with pulmonary fibrosis as a primary endpoint by performing data searches in various systematic review databases. Two reviewers independently reviewed all abstracts (398) and full text data. The quality of study has been assess through SANRA protocol. A total of 32 studies involving were included, included the possible involvement of inflammatory cytokines, concerned the renin-angiotensin system, the potential role of galectin-3, epithelial injuries in fibrosis, alveolar type 2 involvement, Neutrophil extracellular traps (NETs) and the others implied other specific aspects (relationship with clinical and mechanical factors, epithelial transition mesenchymal, TGF-β signaling pathway, midkine, caspase and macrophages, genetics). In most cases, these were narrative reviews or letters to the editor, except for 10 articles, which presented original data, albeit sometimes in experimental models. From the development of these researches, progress in the knowledge of the phenomenon and hopefully in its prevention and therapy may originate.
Collapse
Affiliation(s)
- Laura Bergantini
- Department of Medical Sciences, Surgery and Neurosciences, Respiratory Disease and Lung Transplant Unit, Respiratory Diseases and Transplant Unit, Siena University, Siena, Italy
| | | | | | | | | | | | | |
Collapse
|
13
|
Salvi S, Ghorpade D, Dhoori S, Dhar R, Dumra H, Chhajed P, Bhattacharya P, Rajan S, Talwar D, Christopher D, Mohan M, Udwadia Z. Role of antifibrotic drugs in the management of post-COVID-19 interstitial lung disease: A review of literature and report from an expert working group. Lung India 2022; 39:177-186. [PMID: 35259802 PMCID: PMC9053913 DOI: 10.4103/lungindia.lungindia_659_21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
|
14
|
Giacomelli C, Piccarducci R, Marchetti L, Romei C, Martini C. Pulmonary fibrosis from molecular mechanisms to therapeutic interventions: lessons from post-COVID-19 patients. Biochem Pharmacol 2021; 193:114812. [PMID: 34687672 PMCID: PMC8546906 DOI: 10.1016/j.bcp.2021.114812] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/14/2021] [Accepted: 10/15/2021] [Indexed: 02/07/2023]
Abstract
Pulmonary fibrosis (PF) is characterised by several grades of chronic inflammation and collagen deposition in the interalveolar space and is a hallmark of interstitial lung diseases (ILDs). Recently, infectious agents have emerged as driving causes for PF development; however, the role of viral/bacterial infections in the initiation and propagation of PF is still debated. In this context, the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the virus responsible for the current coronavirus disease 2019 (COVID-19) pandemic, has been associated with acute respiratory distress syndrome (ARDS) and PF development. Although the infection by SARS-CoV-2 can be eradicated in most cases, the development of fibrotic lesions cannot be precluded; furthermore, whether these lesions are stable or progressive fibrotic events is still unknown. Herein, an overview of the main molecular mechanisms driving the fibrotic process together with the currently approved and newly proposed therapeutic solutions was given. Then, the most recent data that emerged from post-COVID-19 patients was discussed, in order to compare PF and COVID-19-dependent PF, highlighting shared and specific mechanisms. A better understanding of PF aetiology is certainly needed, also to develop effective therapeutic strategies and COVID-19 pathology is offering one more chance to do it. Overall, the work reported here could help to define new approaches for therapeutic intervention in the diversity of the ILD spectrum.
Collapse
Affiliation(s)
- Chiara Giacomelli
- Department of Pharmacy, University of Pisa, Via Bonanno 6, Pisa 56126, Italy
| | - Rebecca Piccarducci
- Department of Pharmacy, University of Pisa, Via Bonanno 6, Pisa 56126, Italy
| | - Laura Marchetti
- Department of Pharmacy, University of Pisa, Via Bonanno 6, Pisa 56126, Italy
| | - Chiara Romei
- Multidisciplinary Team of Interstitial Lung Disease, Radiology Department, Pisa University Hospital, Via Paradisa 2, Pisa 56124, Italy
| | - Claudia Martini
- Department of Pharmacy, University of Pisa, Via Bonanno 6, Pisa 56126, Italy,Corresponding author
| |
Collapse
|
15
|
Hakim A, Hasan MM, Hasan M, Lokman SM, Azim KF, Raihan T, Chowdhury PA, Azad AK. Major Insights in Dynamics of Host Response to SARS-CoV-2: Impacts and Challenges. Front Microbiol 2021; 12:637554. [PMID: 34512561 PMCID: PMC8424194 DOI: 10.3389/fmicb.2021.637554] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 07/28/2021] [Indexed: 01/08/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19), a pandemic declared by the World Health Organization on March 11, 2020, is caused by the infection of highly transmissible species of a novel coronavirus called severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). As of July 25, 2021, there are 194,372,584 cases and 4,167,937 deaths with high variability in clinical manifestations, disease burden, and post-disease complications among different people around the globe. Overall, COVID-19 is manifested as mild to moderate in almost 90% of the cases and only the rest 10% of the cases need hospitalization. However, patients with older age and those having different comorbidities have made worst the pandemic scenario. The variability of pathological consequences and clinical manifestations of COVID-19 is associated with differential host-SARS-CoV-2 interactions, which are influenced by the factors that originated from the SARS-CoV-2 and the host. These factors usually include the genomic attributes and virulent factors of the SARS-CoV-2, the burden of coinfection with other viruses and bacteria, age and gender of the individuals, different comorbidities, immune suppressions/deficiency, genotypes of major histocompatibility complex, and blood group antigens and antibodies. We herein retrieved and reviewed literatures from PubMed, Scopus, and Google relevant to clinical complications and pathogenesis of COVID-19 among people of different age, sex, and geographical locations; genomic characteristics of SARS-CoV-2 including its variants, host response under different variables, and comorbidities to summarize the dynamics of the host response to SARS-CoV-2 infection; and host response toward approved vaccines and treatment strategies against COVID-19. After reviewing a large number of published articles covering different aspects of host response to SARS-CoV-2, it is clear that one aspect from one region is not working with the scenario same to others, as studies have been done separately with a very small number of cases from a particular area/region of a country. Importantly, to combat such a pandemic as COVID-19, a conclusive understanding of the disease dynamics is required. This review emphasizes on the identification of the factors influencing the dynamics of host responses to SARS-CoV-2 and offers a future perspective to explore the molecular insights of COVID-19.
Collapse
Affiliation(s)
- Al Hakim
- Department of Genetic Engineering and Biotechnology, School of Life Sciences, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| | - Md. Mahbub Hasan
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences, University of Chittagong, Chittagong, Bangladesh
- Institute of Pharmaceutical Science, School of Cancer and Pharmaceutical Sciences, King’s College London, Franklin-Wilkins Building, London, United Kingdom
| | - Mahmudul Hasan
- Department of Pharmaceutical and Industrial Biotechnology, Sylhet Agricultural University, Sylhet, Bangladesh
| | - Syed Mohammad Lokman
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences, University of Chittagong, Chittagong, Bangladesh
| | - Kazi Faizul Azim
- Department of Microbial Biotechnology, Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, Bangladesh
| | - Topu Raihan
- Department of Genetic Engineering and Biotechnology, School of Life Sciences, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| | | | - Abul Kalam Azad
- Department of Genetic Engineering and Biotechnology, School of Life Sciences, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| |
Collapse
|
16
|
Bayat M, Asemani Y, Mohammadi MR, Sanaei M, Namvarpour M, Eftekhari R. An overview of some potential immunotherapeutic options against COVID-19. Int Immunopharmacol 2021; 95:107516. [PMID: 33765610 PMCID: PMC7908848 DOI: 10.1016/j.intimp.2021.107516] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 02/15/2021] [Accepted: 02/15/2021] [Indexed: 02/07/2023]
Abstract
After the advent of the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV2) in the late 2019, the resulting severe and pernicious syndrome (COVID-19) immediately was deployed all around the world. To date, despite relentless efforts to control the disease by drug repurposing, there is no approved specific therapy for COVID-19. Given the role of innate and acquired immune components in the control and elimination of viral infections and inflammatory mutilations during SARS-CoV2 pathogenesis, immunotherapeutic strategies appear to be beneficent. Passive immunotherapies such as convalescent plasma, which has received much attention especially in severe cases, as well as suppressing inflammatory cytokines, interferon administration, inhibition of kinases and complement cascade, virus neutralization with key engineered products, cell-based therapies, immunomodulators and anti-inflammatory drugs are among the key immunotherapeutic approaches to deal with COVID-19, which is discussed in this review. Also, details of leading COVID-19 vaccine candidates as the most potent immunotherapy have been provided. However, despite salient improvements, there is still a lack of completely assured vaccines for universal application. Therefore, adopting proper immunotherapies according to the cytokine pattern and involved immune responses, alongside engineered biologics specially ACE2-Fc to curb SARS-CoV2 infection until achieving a tailored vaccine is probably the best strategy to better manage this pandemic. Therefore, gaining knowledge about the mechanism of action, potential targets, as well as the effectiveness of immune-based approaches to confront COVID-19 in the form of a well-ordered review study is highly momentous.
Collapse
Affiliation(s)
- Maryam Bayat
- Department of Immunology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Yahya Asemani
- Department of Immunology, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mohammad Reza Mohammadi
- Department of Bacteriology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mahsa Sanaei
- Department of Environmental, Polymer and Organic Chemistry, School of Chemistry, Damghan University, Damghan, Iran
| | - Mozhdeh Namvarpour
- Department of Immunology, Shahid Sadoughi University of Medical Science and services, Yazd, Iran
| | - Reyhaneh Eftekhari
- Department of Microbiology, Faculty of Biology, Semnan University, Semnan, Iran
| |
Collapse
|
17
|
Alghizzawi M, Ata F, Yousaf Z, Alhiyari M, Bint I Bilal A, Elhiday A, Abdulhadi A. The second wave of desaturation in coronavirus disease 2019. New Microbes New Infect 2021; 41:100866. [PMID: 33758668 PMCID: PMC7972827 DOI: 10.1016/j.nmni.2021.100866] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 03/12/2021] [Accepted: 03/15/2021] [Indexed: 12/01/2022] Open
Abstract
After a complete symptomatic recovery from coronavirus disease 2019 pneumonia, the second phase of desaturation is a new phenomenon that is being increasingly observed. Two possible mechanisms behind it can be a continued subclinical infection and lung fibrosis. We have presented a case with the former mechanism, who responded well to steroids.
Collapse
Affiliation(s)
- M.I. Alghizzawi
- Department of Internal Medicine, Hamad Medical Corporation, Doha, Qatar
| | - F. Ata
- Department of Internal Medicine, Hamad Medical Corporation, Doha, Qatar
| | - Z. Yousaf
- Department of Internal Medicine, Hamad Medical Corporation, Doha, Qatar
| | - M.A. Alhiyari
- Department of Internal Medicine, Hamad Medical Corporation, Doha, Qatar
| | - A. Bint I Bilal
- Department of Radiology, Hamad Medical Corporation, Doha, Qatar
| | - A. Elhiday
- Department of Internal Medicine, Hamad Medical Corporation, Doha, Qatar
| | - A.S. Abdulhadi
- Department of Internal Medicine, Hamad Medical Corporation, Doha, Qatar
| |
Collapse
|
18
|
Prakash J, Bhattacharya PK, Priye S, Kumar N. Post-COVID-19 Pulmonary Fibrosis: A Lifesaving Challenge. Indian J Crit Care Med 2021; 25:104-105. [PMID: 33603312 PMCID: PMC7874295 DOI: 10.5005/jp-journals-10071-23709] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
How to cite this article: Prakash J, Bhattacharya PK, Priye S, Kumar N. Post-COVID-19 Pulmonary Fibrosis: A Lifesaving Challenge. Indian J Crit Care Med 2021;25(1):104–105.
Collapse
Affiliation(s)
- Jay Prakash
- Department of Critical Care Medicine, Rajendra Institute of Medical Sciences, Ranchi, Jharkhand, India
| | - Pradip K Bhattacharya
- Department of Critical Care Medicine, Rajendra Institute of Medical Sciences, Ranchi, Jharkhand, India
| | - Shio Priye
- Department of Superspeciality Anesthesia, Rajendra Institute of Medical Sciences, Ranchi, Jharkhand, India
| | - Naveen Kumar
- Department of Radiodiagnosis, Rajendra Institute of Medical Sciences, Ranchi, Jharkhand, India
| |
Collapse
|
19
|
COVID-19 Patients with Pulmonary Fibrotic Tissue: Clinical Pharmacological Rational of Antifibrotic Therapy. ACTA ACUST UNITED AC 2020; 2:1709-1712. [PMID: 32875276 PMCID: PMC7452615 DOI: 10.1007/s42399-020-00487-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/24/2020] [Indexed: 02/07/2023]
Abstract
In December 2019, the first data emerged from Wuhan, China, of a serious acute respiratory disease caused by a new coronavirus, SARS-CoV-2 (COVID-19). In a short time, the health emergency became a global pandemic. To date, there are about 18.8 million infected people and about 700,000 deaths. There are currently no effective vaccines, and treatments are mostly experimental. The symptoms associated with COVID-19 are different, ranging from mild upper respiratory tract symptoms to severe acute respiratory distress syndrome (SARS). Data from previous coronavirus outbreaks such as SARS-CoV (2003 outbreak) and emerging epidemiological data from the current global COVID-19 pandemic suggest that there could be substantial tissue fibrotic consequences following SARS-CoV-2 infection, responsible for severe and in some cases fatal lung lesions. Some data show that even patients cured of viral infection have lung fibrotic tissue residues responsible for incorrect respiratory function even after healing. The role of antifibrotic drug therapy in patients with ongoing SARS-CoV-2 infection or in patients cured of residual pulmonary fibrosis is still to be defined and unclear; the scientific rationale for initiating, continuing, or discontinuing therapy is poorly defined. In this article, we describe the advantages of antifibrotic therapy in patients with ongoing SARS-CoV-2 viral infection to prevent the worsening and aggravation of the clinical situation, and the advantages it could have in the role of preventing pulmonary fibrosis after SARS-CoV-2 infection, and in accelerating the complete healing process.
Collapse
|
20
|
Nakanishi T, Sakiyama S, Takashima H, Honda R, Shumba MN, Nakamura Y, Kasahara K, Tamai I. Toxicological implication of prostaglandin transporter SLCO2A1 inhibition by cigarette smoke in exacerbation of lung inflammation. Toxicol Appl Pharmacol 2020; 405:115201. [PMID: 32828905 DOI: 10.1016/j.taap.2020.115201] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 08/11/2020] [Accepted: 08/14/2020] [Indexed: 12/21/2022]
Abstract
We reported that bleomycin (BLM)-induced pulmonary fibrosis was exacerbated in the prostaglandin transporter gene (Slco2a1)-deficient mice (Slco2a1(-/-)). Because cigarette smoke (CS) contributes to creating a profibrotic milieu in the respiratory region, the present study aimed to investigate the impact of CS on SLCO2A1-associated pathogenesis in the lungs of BLM-instilled mice. Bronchoalveolar lavage (BAL) fluid cell analysis indicated more severe inflammation in Slco2a1(-/-) on day 5 after BLM intratracheal instillation, and Slco2a1 deletion increased mRNA expression of pro-inflammatory cytokines (Tnf-α and Il-1β) and chemokine (Ccl5) in BAL cells. Male Slco2a1(-/-) exhibited significantly higher amounts of released Il-1β in BAL fluid, compared with female Slco2a1(-/-), male or female Slco2a1(+/+) group. The amount of PGE2 collected in BAL fluid tended to increase in Slco2a1(-/-) compared with Slco2a1(+/+) group, whereas the PGE2 concentrations in lung tissues were comparable between both groups. Besides, PGE2 accumulated more in BAL fluid of male than that of female mice. Therefore, Slco2a1-deficient male mice were found to be more susceptible to BLM-treatment. Moreover, CS extracts (CSE) significantly reduced initial PGE2 uptake by rat type1 alveolar epithelial cell-like (AT1-L) cells and human SLCO2A1-transfected cells. Exposure of AT1-L cells to CSE resulted in decreased mRNA expression of Slco2a1, suggesting that CS modulates SLCO2A1 function. These results indicate that exacerbated lung inflammation is attributed to an increase in Il-1β peptide and PGE2 accumulation in the alveolar space, which exhibits a male predominance. SLCO2A1 inhibition by CSE is considered to be a new rationale for the lung toxicity of CS.
Collapse
Affiliation(s)
- Takeo Nakanishi
- Faculty of Pharmacy, Takasaki University of Health and Welfare, 60 Nakaorui-machi, Takasaki, Japan.
| | - Shiori Sakiyama
- School of Pharmaceutical Sciences, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Hiroki Takashima
- School of Pharmaceutical Sciences, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Ryokichi Honda
- School of Pharmaceutical Sciences, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Melody N Shumba
- Depatiment of Nutrition, Faculty of Health and Welfare, Takasaki University of Health and Welfare, Takasaki, Japan
| | - Yoshinobu Nakamura
- Faculty of Pharmacy, Takasaki University of Health and Welfare, 60 Nakaorui-machi, Takasaki, Japan
| | - Kazuo Kasahara
- School of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa, 920-0934, Japan
| | - Ikumi Tamai
- School of Pharmaceutical Sciences, Kakuma-machi, Kanazawa 920-1192, Japan
| |
Collapse
|
21
|
Lechowicz K, Drożdżal S, Machaj F, Rosik J, Szostak B, Zegan-Barańska M, Biernawska J, Dabrowski W, Rotter I, Kotfis K. COVID-19: The Potential Treatment of Pulmonary Fibrosis Associated with SARS-CoV-2 Infection. J Clin Med 2020; 9:E1917. [PMID: 32575380 PMCID: PMC7356800 DOI: 10.3390/jcm9061917] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/11/2020] [Accepted: 06/17/2020] [Indexed: 01/08/2023] Open
Abstract
In December 2019, a novel coronavirus, SARS-CoV-2, appeared, causing a wide range of symptoms, mainly respiratory infection. In March 2020, the World Health Organization (WHO) declared Coronavirus Disease 2019 (COVID-19) a pandemic, therefore the efforts of scientists around the world are focused on finding the right treatment and vaccine for the novel disease. COVID-19 has spread rapidly over several months, affecting patients across all age groups and geographic areas. The disease has a diverse course; patients may range from asymptomatic to those with respiratory failure, complicated by acute respiratory distress syndrome (ARDS). One possible complication of pulmonary involvement in COVID-19 is pulmonary fibrosis, which leads to chronic breathing difficulties, long-term disability and affects patients' quality of life. There are no specific mechanisms that lead to this phenomenon in COVID-19, but some information arises from previous severe acute respiratory syndrome (SARS) or Middle East respiratory syndrome (MERS) epidemics. The aim of this narrative review is to present the possible causes and pathophysiology of pulmonary fibrosis associated with COVID-19 based on the mechanisms of the immune response, to suggest possible ways of prevention and treatment.
Collapse
Affiliation(s)
- Kacper Lechowicz
- Department of Anaesthesiology, Intensive Therapy and Acute Intoxications, Pomeranian Medical University, 70-111 Szczecin, Poland; (K.L.); (M.Z.-B.)
| | - Sylwester Drożdżal
- Department of Pharmacokinetics and Monitored Therapy, Pomeranian Medical University, 70-111 Szczecin, Poland;
| | - Filip Machaj
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (F.M.); (J.R.); (B.S.)
| | - Jakub Rosik
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (F.M.); (J.R.); (B.S.)
| | - Bartosz Szostak
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (F.M.); (J.R.); (B.S.)
| | - Małgorzata Zegan-Barańska
- Department of Anaesthesiology, Intensive Therapy and Acute Intoxications, Pomeranian Medical University, 70-111 Szczecin, Poland; (K.L.); (M.Z.-B.)
| | - Jowita Biernawska
- Department of Anaesthesiology and Intensive Therapy, Pomeranian Medical University, 71-242 Szczecin, Poland;
| | - Wojciech Dabrowski
- Department of Anaesthesiology and Intensive Care, Medical University, 20-090 Lublin, Poland;
| | - Iwona Rotter
- Department of Medical Rehabilitation and Clinical Physiotherapy, Pomeranian Medical University, 71-210 Szczecin, Poland;
| | - Katarzyna Kotfis
- Department of Anaesthesiology, Intensive Therapy and Acute Intoxications, Pomeranian Medical University, 70-111 Szczecin, Poland; (K.L.); (M.Z.-B.)
| |
Collapse
|
22
|
Shi Y, Wang G, Cai XP, Deng JW, Zheng L, Zhu HH, Zheng M, Yang B, Chen Z. An overview of COVID-19. J Zhejiang Univ Sci B 2020; 21:343-360. [PMID: 32425000 PMCID: PMC7205601 DOI: 10.1631/jzus.b2000083] [Citation(s) in RCA: 249] [Impact Index Per Article: 49.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 03/09/2020] [Indexed: 01/08/2023]
Abstract
Pneumonia caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection emerged in Wuhan City, Hubei Province, China in December 2019. By Feb. 11, 2020, the World Health Organization (WHO) officially named the disease resulting from infection with SARS-CoV-2 as coronavirus disease 2019 (COVID-19). COVID-19 represents a spectrum of clinical manifestations that typically include fever, dry cough, and fatigue, often with pulmonary involvement. SARS-CoV-2 is highly contagious and most individuals within the population at large are susceptible to infection. Wild animal hosts and infected patients are currently the main sources of disease which is transmitted via respiratory droplets and direct contact. Since the outbreak, the Chinese government and scientific community have acted rapidly to identify the causative agent and promptly shared the viral gene sequence, and have carried out measures to contain the epidemic. Meanwhile, recent research has revealed critical aspects of SARS-CoV-2 biology and disease pathogenesis; other studies have focused on epidemiology, clinical features, diagnosis, management, as well as drug and vaccine development. This review aims to summarize the latest research findings and to provide expert consensus. We will also share ongoing efforts and experience in China, which may provide insight on how to contain the epidemic and improve our understanding of this emerging infectious disease, together with updated guidance for prevention, control, and critical management of this pandemic.
Collapse
Affiliation(s)
- Yu Shi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Gang Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Xiao-Peng Cai
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Jing-Wen Deng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Lin Zheng
- School of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Hai-Hong Zhu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Min Zheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Bo Yang
- School of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Zhi Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
- Health Policy and Hospital Management Research Center, School of Medicine, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
23
|
Lesur O, Chagnon F, Lebel R, Lepage M. In Vivo Endomicroscopy of Lung Injury and Repair in ARDS: Potential Added Value to Current Imaging. J Clin Med 2019; 8:jcm8081197. [PMID: 31405200 PMCID: PMC6723156 DOI: 10.3390/jcm8081197] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 08/06/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Standard clinical imaging of the acute respiratory distress syndrome (ARDS) lung lacks resolution and offers limited possibilities in the exploration of the structure-function relationship, and therefore cannot provide an early and clear discrimination of patients with unexpected diagnosis and unrepair profile. The current gold standard is open lung biopsy (OLB). However, despite being able to reveal precise information about the tissue collected, OLB cannot provide real-time information on treatment response and is accompanied with a complication risk rate up to 25%, making longitudinal monitoring a dangerous endeavor. Intravital probe-based confocal laser endomicroscopy (pCLE) is a developing and innovative high-resolution imaging technology. pCLE offers the possibility to leverage multiple and specific imaging probes to enable multiplex screening of several proteases and pathogenic microorganisms, simultaneously and longitudinally, in the lung. This bedside method will ultimately enable physicians to rapidly, noninvasively, and accurately diagnose degrading lung and/or fibrosis without the need of OLBs. OBJECTIVES AND METHODS To extend the information provided by standard imaging of the ARDS lung with a bedside, high-resolution, miniaturized pCLE through the detailed molecular imaging of a carefully selected region-of-interest (ROI). To validate and quantify real-time imaging to validate pCLE against OLB. RESULTS Developments in lung pCLE using fluorescent affinity- or activity-based probes at both preclinical and clinical (first-in-man) stages are ongoing-the results are promising, revealing correlations with OLBs in problematic ARDS. CONCLUSION It can be envisaged that safe, high-resolution, noninvasive pCLE with activatable fluorescence probes will provide a "virtual optical biopsy" and will provide decisive information in selected ARDS patients at the bedside.
Collapse
Affiliation(s)
- Olivier Lesur
- Intensive Care and Pneumology Departments, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC J1H 5N4, Canada.
- Sherbrooke Molecular Imaging Center (CIMS), Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC J1H 5N4, Canada.
| | - Frédéric Chagnon
- Intensive Care and Pneumology Departments, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Réjean Lebel
- Sherbrooke Molecular Imaging Center (CIMS), Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
- Nuclear Medicine and Radiobiology Departments, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Martin Lepage
- Sherbrooke Molecular Imaging Center (CIMS), Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
- Nuclear Medicine and Radiobiology Departments, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| |
Collapse
|
24
|
Chen C, Yun XJ, Liu LZ, Guo H, Liu LF, Chen XL. Exogenous nitric oxide enhances the prophylactic effect of aminoguanidine, a preferred iNOS inhibitor, on bleomycin-induced fibrosis in the lung: Implications for the direct roles of the NO molecule in vivo. Nitric Oxide 2017; 70:31-41. [PMID: 28757441 DOI: 10.1016/j.niox.2017.07.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 06/12/2017] [Accepted: 07/25/2017] [Indexed: 11/15/2022]
Abstract
OBJECTIVE Inducible nitric oxide synthase (iNOS) aggravates and endothelial nitric oxide synthase (eNOS) ameliorates fibrosis in the lung. Our previous study demonstrated that aminoguanidine (AG), a preferred iNOS inhibitor, prevents bleomycin-induced injury and fibrosis in the lung. The diethylenetriamine nitric oxide adduct (DETA/NO) is a slow-release NO donor. Here, to clarify the exact role of the nitric oxide (NO) molecule in the pathogenesis of pulmonary fibrosis in vivo, we observed the effects of inhalation of aerosolized DETA/NO on fibrosis in the lungs of bleomycin-exposed rats with AG treatment, including the effects on the myofibroblast number, collagen deposition, peroxynitrite anion (ONOO-) formation, and injury in the lung. DESIGN AND METHODS Rats received a single intratracheal instillation of bleomycin or normal saline (NS) on day 0, followed by a daily intraperitoneal injection of AG or NS from day 1 to day 13. Each group was additionally given a daily inhalation of DETA/NO or placebo from day 1 to day 13. On day 14, half of the rats in each group was euthanized, and plasma nitrite and nitrate (NOx), myofibroblasts, type I collagen, ONOO- and injury in the lung were estimated by the Griess reaction, western blotting, immunohistochemical staining, sirius red staining, and hematoxylin and eosin (HE) staining, respectively. On day 28, the other half of the rats in each group was euthanized, and the total collagen of the lung was evaluated by hydroxyproline assay. RESULTS ① At the day 14 time point, AG reduced the plasma NOx level in bleomycin rats, while this drug had no significant effect on sham rats. Inhalation of aerosolized DETA/NO increased the plasma NOx level of bleomycin + AG rats, sham rats and sham + AG rats. However, due to large areas of airspace obliteration in the lungs of bleomycin rats, DETA/NO inhalation had no significant effect on the plasma NOx level in these rats. ② At the day 14 time point, AG reduced ONOO- formation (marked by nitrotyrosine, NT), injury, myofibroblast number, and type I collagen deposition in the lungs of bleomycin rats, while this drug had no significant impact on the above parameters in the lungs of sham rats. Interestingly, DETA/NO inhalation enhanced the preventive effects afforded by AG on myofibroblast number and type I collagen deposition, but had no significant impact on ONOO- and injury in lung. ③ At the day 28 time point, because rats were not exposed to DETA/NO after day 13, there was no significant difference of the plasma NOx level in sham rats, sham + AG rats, bleomycin rats, and bleomycin + AG rats between DETA/NO inhalation and placebo inhalation. Interestingly, rats administered both DETA/NO and AG still showed a reduction in total collagen of the entire lung compared to rats administered AG alone at this time point. CONCLUSIONS Exogenous NO enhances the prophylactic effect afforded by AG on the myofibroblast number and collagen deposition in the lungs of bleomycin-treated rats in vivo. These results suggest that NO has a direct antifibrotic effect in lungs, except for the formation of ONOO- in the development of pulmonary fibrosis in vivo.
Collapse
Affiliation(s)
- Chao Chen
- Department of Pathophysiology, Institute of Basic Medicine, Hebei Medical University, 361 Zongshan East Road, Shijiazhuang, Hebei, 050017, PR China; Department of Cardiology, Cangzhou Central Hospital, Cangzhou, Hebei, 061014, PR China
| | - Xiao-Jing Yun
- Department of Pathophysiology, Institute of Basic Medicine, Hebei Medical University, 361 Zongshan East Road, Shijiazhuang, Hebei, 050017, PR China
| | - Li-Ze Liu
- Department of Pathophysiology, Institute of Basic Medicine, Hebei Medical University, 361 Zongshan East Road, Shijiazhuang, Hebei, 050017, PR China
| | - Hong Guo
- Department of Pathophysiology, Institute of Basic Medicine, Hebei Medical University, 361 Zongshan East Road, Shijiazhuang, Hebei, 050017, PR China
| | - Lian-Feng Liu
- Department of Pathophysiology, Institute of Basic Medicine, Hebei Medical University, 361 Zongshan East Road, Shijiazhuang, Hebei, 050017, PR China
| | - Xiao-Ling Chen
- Department of Pathophysiology, Institute of Basic Medicine, Hebei Medical University, 361 Zongshan East Road, Shijiazhuang, Hebei, 050017, PR China.
| |
Collapse
|
25
|
Penetration of Ciprofloxacin and Amikacin into the Alveolar Epithelial Lining Fluid of Rats with Pulmonary Fibrosis. Antimicrob Agents Chemother 2017; 61:AAC.01936-16. [PMID: 28115351 DOI: 10.1128/aac.01936-16] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 01/15/2017] [Indexed: 01/29/2023] Open
Abstract
We determined the concentration-time profiles of ciprofloxacin and amikacin in serum and alveolar epithelial lining fluid (ELF) of rats with or without pulmonary fibrosis and investigated the effect of pulmonary fibrosis on the capacity for penetration of antimicrobials into the ELF of rats. Pulmonary fibrosis was induced in rats with a single intratracheal instillation of bleomycin. After intravenous injection of ciprofloxacin or amikacin, blood and bronchoalveolar lavage fluid samples were collected. Urea concentrations in serum and lavage fluid were determined using an enzymatic assay. Ciprofloxacin and amikacin concentrations were determined by high-performance liquid chromatography and liquid chromatography-tandem mass spectrometry, respectively. The mean ratio of ELF to plasma concentrations of ciprofloxacin at each time point in the normal group did not significantly differ from that in the pulmonary fibrosis group. However, the ratio of the ciprofloxacin area under the concentration-time curve from 0 to 24 h (AUC0-24) in ELF to the AUC0-24 in plasma was 1.02 in the normal group and 0.76 in the pulmonary fibrosis group. The mean ELF-to-plasma concentration ratios of amikacin at each time point in the normal group were higher than those in the pulmonary fibrosis group, reaching a statistically significant difference at 1, 2, and 4 h. The ratio of the AUC0-24 in ELF to the AUC0-24 in plasma was 0.49 in the normal group and 0.27 in the pulmonary fibrosis group. In conclusion, pulmonary fibrosis can influence the penetration of antimicrobials into the ELF of rats and may have a marked effect on the penetration of amikacin than that of ciprofloxacin.
Collapse
|
26
|
Ulker OC, Yucesoy B, Durucu M, Karakaya A. Neopterin as a marker for immune system activation in coal workers' pneumoconiosis. Toxicol Ind Health 2016; 23:155-60. [DOI: 10.1177/0748233707083527] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Coal workers' pneumoconiosis (CWP) is an occupational pulmonary disease that occurs by chronic inhalation of coal dust. Coal workers' pneumoconiosis is divided into two categories depending on the extent of the disease as simple pneumoconiosis (SP) and progressive massive fibrosis (PMF). Development of CWP is associated with the activation of the immune system. Neopterin is a predictive biochemical marker of cell-mediated immune activation and elevated levels of neopterin are detected in body fluids of patients with immune-related diseases. The present study was aimed to investigate whether increased serum, urine and bronchoalveolar lavage (BAL) fluid levels of neopterin is associated with the development and/or severity of CWP. Mean serum neopterin levels in SP and PMF patients (10.72 ± 0.98 nmol/L; 14.08 ± 3.86 nmol/L, respectively) were significantly higher than those of control group (5.30 ± 0.47nmol/L) ( P < 0.05). Although urinary neopterin levels were also increased in SP and PMF patients (235.17 ± 7.40 μmol/mol creatinine; 256.05 ± 9.43 μmol/mol creatinine, respectively) as compared with the control group (140.00 ± 5.43 μmol/mol creatinine) ( P < 0.01), they were within the normal concentration range. No significant difference was observed between serum and urinary neopterin levels of SP and PMF patients. A correlation was observed between serum and urinary neopterin levels of all subjects ( r = 0.525, P < 0.01). Bronchoalveolar lavage fluid neopterin levels were significantly higher in patients with SP and PMF (22.67 ± 2.9 nmol/L; 41.67 ± 8.68nmol/L, respectively) compared with control subjects (6.264 ± 1.74 nmol/L) ( P < 0.05, P < 0.01, respectively). The levels of neopterin in BAL fluid were also significantly higher in patients with PMF than in those with SP ( P < 0.05). These findings indicate that elevated serum and BAL levels of neopterin may be considered as a suitable biomarker for the assessment of CWP. Toxicology and Industrial Health 2007; 23: 155—160.
Collapse
Affiliation(s)
- Ozge C. Ulker
- Department of Toxicology, Ankara University, Ankara, Turkey
| | - Berran Yucesoy
- Department of Toxicology, Ankara University, Ankara, Turkey
| | - Murat Durucu
- Zonguldak Chest and Occupational Disease Hospital, Zonguldak, Turkey
| | - Asuman Karakaya
- Department of Toxicology, Ankara University, Ankara, Turkey,
| |
Collapse
|
27
|
Patel A, Sabbineni H, Clarke A, Somanath PR. Novel roles of Src in cancer cell epithelial-to-mesenchymal transition, vascular permeability, microinvasion and metastasis. Life Sci 2016; 157:52-61. [PMID: 27245276 DOI: 10.1016/j.lfs.2016.05.036] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 05/25/2016] [Accepted: 05/27/2016] [Indexed: 12/21/2022]
Abstract
The Src-family kinases (SFKs), an intracellularly located group of non-receptor tyrosine kinases are involved in oncogenesis. The importance of SFKs has been implicated in the promotion of tumor cell motility, proliferation, inhibition of apoptosis, invasion and metastasis. Recent evidences indicate that specific effects of SFKs on epithelial-to-mesenchymal transition (EMT) as well as on endothelial and stromal cells in the tumor microenvironment can have profound effects on tumor microinvasion and metastasis. Although, having been studied extensively, these novel features of SFKs may contribute to greater understanding of benefits from Src inhibition in various types of cancers. Here we review the novel role of SFKs, particularly c-Src in mediating EMT, modulation of tumor endothelial-barrier, transendothelial migration (microinvasion) and metastasis of cancer cells, and discuss the utility of Src inhibitors in vascular normalization and cancer therapy.
Collapse
Affiliation(s)
- Ami Patel
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA, United States
| | - Harika Sabbineni
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA, United States; Charlie Norwood VA Medical Center, Augusta, GA, United States
| | - Andrea Clarke
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA, United States
| | - Payaningal R Somanath
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA, United States; Charlie Norwood VA Medical Center, Augusta, GA, United States; Department of Medicine, Vascular Biology Center and Cancer Center, Augusta University, Augusta, GA, United States.
| |
Collapse
|
28
|
Zhou Y, Liao S, Zhang Z, Wang B, Wan L. Astragalus injection attenuates bleomycin-induced pulmonary fibrosis via down-regulating Jagged1/Notch1 in lungs. ACTA ACUST UNITED AC 2016; 68:389-96. [PMID: 26817817 DOI: 10.1111/jphp.12518] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 12/13/2015] [Indexed: 02/05/2023]
Abstract
OBJECTIVES Inhibition of Notch signalling is a potential therapeutic strategy for pulmonary fibrosis. This study was designed to investigate the antifibrosis effects and possible mechanism of astragalus injection (AI) on bleomycin (BLM)-induced pulmonary fibrosis in rats. METHODS Pulmonary fibrosis was induced by intratracheal instillation of bleomycin (5 mg/kg) in male SD rats. All rats received daily intraperitoneally administration of dexamethasone (DEX, 3 mg/kg), astragalus injection (AI, 8 g/kg) or saline 1 day after bleomycin instillation daily for 28 days. Histological changes in the lung were evaluated by haematoxylin and eosin and Masson's trichrome staining. The expression of α-smooth muscle protein (α-SMA) was assayed by immunohistochemical (IHC). The mRNA and protein level of Jagged1, Notch1 and transforming growth factor-β1 (TGF-β1) was analysed by qPCR and Western blot. KEY FINDINGS BLM-induced severe alveolitis and pulmonary fibrosis; together with significant elevation of α-SMA, TGF-β1, Jagged1 and Notch1. Astragalus injection (AI, 8 g/kg) administration notably attenuated the degree of alveolitis and lung fibrosis, and markedly reduced the elevated levels of α-SMA, TGF-β1, Jagged1 and Notch1 in lungs. CONCLUSIONS Astragalus injection (AI, 8 g/kg) may exert protective effects on bleomycin-induced pulmonary fibrosis via downregulating Jagged1/Notch1 in lung.
Collapse
Affiliation(s)
- Yan Zhou
- Department of Intensive Care Unit, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Shiping Liao
- Functional Laboratory, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Zhongwei Zhang
- Department of Intensive Care Unit, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Bo Wang
- Department of Intensive Care Unit, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lihong Wan
- Department of Pharmacology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China.,Sichuan University 985 project - Science and Technology Innovation Platform for Novel Drug Development', Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
29
|
Li L, Li D, Xu L, Zhao P, Deng Z, Mo X, Li P, Qi L, Li J, Gao J. Total extract of Yupingfeng attenuates bleomycin-induced pulmonary fibrosis in rats. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2015; 22:111-119. [PMID: 25636879 DOI: 10.1016/j.phymed.2014.10.011] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 09/06/2014] [Accepted: 10/26/2014] [Indexed: 06/04/2023]
Abstract
Yupingfeng is a Chinese herbal compound used efficaciously to treat respiratory tract diseases. Total glucosides of Yupingfeng have been proven effective in anti-inflammation and immunoregulation. Nevertheless, the role of total extract of Yupingfeng (YTE) in pulmonary fibrosis (PF), a severe lung disease with no substantial therapies, remains unknown. Present study was conducted to elucidate the anti-fibrotic activity of YTE. The rat PF model was induced by intratracheal administration of bleomycin (BLM, 5 mg/kg), and YTE (12 mg/kg/d) was gavaged from the second day. At 14 and 28 days, the lungs were harvested and stained with H&E and Masson's trichrome. The content of hydroxyproline (HYP) and type I collagen (Col-I) were detected, while the protein expression of high-mobility group box 1 (HMGB1), transforming growth factor-beta 1 (TGF-β1), Col-I and α-smooth muscle actin (α-SMA) were analyzed by immunohistochemistry or Western blot. As observed, YTE treatment attenuated the alveolitis and fibrosis induced by BLM, reduced the loss of body weight and increase of lung coefficient. Meanwhile, YTE strongly decreased the levels of HYP and Col-I, and reduced the over-expression of HMGB1, TGF-β1, Col-I and α-SMA. In conclusion, YTE could ameliorate BLM-induced lung fibrosis by alleviating HMGB1 activity and TGF-β1 activation, suggesting therapeutic potential for PF.
Collapse
Affiliation(s)
- Liucheng Li
- School of Pharmacy (Anhui Key Laboratory of Bioactivity of Natural Products), Anhui Medical University, Hefei 230032, China; Pharmaceutical Preparation Section (Third-Grade Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine (TCM-2009-202)), The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Delin Li
- Pharmaceutical Preparation Section (Third-Grade Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine (TCM-2009-202)), The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China; School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230038, China
| | - Liang Xu
- Pharmaceutical Preparation Section (Third-Grade Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine (TCM-2009-202)), The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China; School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230038, China
| | - Ping Zhao
- Pharmaceutical Preparation Section (Third-Grade Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine (TCM-2009-202)), The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China; School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230038, China
| | - Ziyu Deng
- School of Pharmacy (Anhui Key Laboratory of Bioactivity of Natural Products), Anhui Medical University, Hefei 230032, China; The Second Affiliated Hospital of Anhui Medical University, Hefei 230012, China
| | - Xiaoting Mo
- School of Pharmacy (Anhui Key Laboratory of Bioactivity of Natural Products), Anhui Medical University, Hefei 230032, China; Pharmaceutical Preparation Section (Third-Grade Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine (TCM-2009-202)), The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Ping Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Lianwen Qi
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Jun Li
- School of Pharmacy (Anhui Key Laboratory of Bioactivity of Natural Products), Anhui Medical University, Hefei 230032, China.
| | - Jian Gao
- Pharmaceutical Preparation Section (Third-Grade Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine (TCM-2009-202)), The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China.
| |
Collapse
|
30
|
Mostaço-Guidolin LB, Ko ACT, Wang F, Xiang B, Hewko M, Tian G, Major A, Shiomi M, Sowa MG. Collagen morphology and texture analysis: from statistics to classification. Sci Rep 2014; 3:2190. [PMID: 23846580 PMCID: PMC3709165 DOI: 10.1038/srep02190] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Accepted: 06/07/2013] [Indexed: 02/08/2023] Open
Abstract
In this study we present an image analysis methodology capable of quantifying morphological changes in tissue collagen fibril organization caused by pathological conditions. Texture analysis based on first-order statistics (FOS) and second-order statistics such as gray level co-occurrence matrix (GLCM) was explored to extract second-harmonic generation (SHG) image features that are associated with the structural and biochemical changes of tissue collagen networks. Based on these extracted quantitative parameters, multi-group classification of SHG images was performed. With combined FOS and GLCM texture values, we achieved reliable classification of SHG collagen images acquired from atherosclerosis arteries with >90% accuracy, sensitivity and specificity. The proposed methodology can be applied to a wide range of conditions involving collagen re-modeling, such as in skin disorders, different types of fibrosis and muscular-skeletal diseases affecting ligaments and cartilage.
Collapse
Affiliation(s)
- Leila B Mostaço-Guidolin
- National Research Council Canada, Medical Devices Portfolio 435 Ellice Avenue, Winnipeg, MB, Canada R3B 1Y6
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Objective assessment of endogenous collagen in vivo during tissue repair by laser induced fluorescence. PLoS One 2014; 9:e98609. [PMID: 24874229 PMCID: PMC4038633 DOI: 10.1371/journal.pone.0098609] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 05/06/2014] [Indexed: 01/31/2023] Open
Abstract
Collagen, a triple helical protein with the primary role of mechanical function, provides tensile strength to the skin, and plays a pivotal task in tissue repair. During tissue regeneration, collagen level increases gradually and therefore, monitoring of such changes in vivo by laser induced fluorescence was the main objective behind the present study. In order to accomplish this, 15 mm diameter excisional wounds were created on six to eight week old Swiss albino mice. The collagen deposition accelerated upon irradiation of single exposure of 2 J/cm2 He-Ne laser dose immediately after wounding was recorded by laser induced autofluorescence in vivo along with un-illuminated and un-wounded controls. Autofluorescence spectra were recorded for each animal of the experimental groups on 0, 5, 10, 30, 45 and 60 days post-wounding, by exciting the granulation tissue/skin with 325 nm He-Cd laser. The variations in the average collagen intensities from the granulation tissue/skin of mice were inspected as a function of age and gender. Further, the spectral findings of the collagen synthesis in wound granulation tissue/un-wounded skin tissues were validated by Picro-Sirius red- polarized light microscopy in a blinded manner through image analysis of the respective collagen birefringence. The in vivo autofluorescence studies have shown a significant increase in collagen synthesis in laser treated animals as compared to the un-illuminated controls. Image analysis of the collagen birefringence further authenticated the ability of autofluorescence in the objective monitoring of collagen in vivo. Our results clearly demonstrate the potential of laser induced autofluorescence in the monitoring of collegen synthesis during tissue regeneration, which may have clinical implications.
Collapse
|
32
|
Evaluating the Ameliorative Potential of Quercetin against the Bleomycin-Induced Pulmonary Fibrosis in Wistar Rats. Pulm Med 2013; 2013:921724. [PMID: 24396596 PMCID: PMC3875129 DOI: 10.1155/2013/921724] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Revised: 08/15/2013] [Accepted: 10/10/2013] [Indexed: 02/07/2023] Open
Abstract
The current study deals with the effect of a dietary flavanoid quercetin on fibrotic lung tissue in rats. Bleomycin was administered by single intratracheal instillation to Wistar rats to induce lung fibrosis. The pathologies associated with this included significantly reduced antioxidant capacity, ultimately leading to protracted inflammation of the lung tissue. The hallmark of this induced fibrosis condition was an excessive collagen deposition in peribronchial and perialveolar regions of the lung. Oral quercetin treatment over a period of twenty days resulted in significant reversal of the pathologies. The antioxidant defense in lung tissue was revived. Moreover, activity of the collagenase MMP-7, which was high in fibrotic tissue, was seen restored after quercetin administration. Trichome staining of lung tissue sections showed high collagen deposition in fibrotic rats, which may be a direct result of increased mobilization of collagen by MMP-7. This was appreciably reduced in quercetin treated animals. These results point towards an important protective role of quercetin against idiopathic lung fibrosis, which remains a widely prevalent yet incurable condition in the present times.
Collapse
|
33
|
Verma R, Brahmankar M, Kushwah L, Suresh B. Evaluating the inhibitory potential of sulindac against the bleomycin-induced pulmonary fibrosis in wistar rats. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2013; 36:769-778. [PMID: 23958969 DOI: 10.1016/j.etap.2013.07.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 07/15/2013] [Indexed: 06/02/2023]
Abstract
The present study examined the protective effect of sulindac on bleomycin-induced lung fibrosis in rats. Animals were divided into saline group, bleomycin group (single intra-tracheal instillation of bleomycin) and bleomycin+sulindac (orally from day 1 to day 20). Bleomycin administration reduced the body weight, altered antioxidant status (such as superoxide dismutase, catalase, glutathione peroxidase, glutathione reductase and glutathione) while it increased the lung weight, hydroxyproline content, collagen deposition and lipid peroxidation. However, simultaneous administration of sulindac improved the body weight, antioxidant status and decreased the collagen deposition in lungs. Moreover, the levels of inflammatory cytokine tumour necrosis factor-α increased in bleomycin-induced group, whereas, on treatment with sulindac the levels of tumour necrosis factor-α were found reduced. Finally, histological evidence also supported the ability of sulindac to inhibit bleomycin-induced lung fibrosis. The results of the present study indicate that sulindac can be used as an agent against bleomycin-induced pulmonary fibrosis.
Collapse
Affiliation(s)
- Ramesh Verma
- Department of Toxicology, Jai Research Foundation, Valvada 396108, Gujarat, India
| | | | | | | |
Collapse
|
34
|
Johnson A, DiPietro LA. Apoptosis and angiogenesis: an evolving mechanism for fibrosis. FASEB J 2013; 27:3893-901. [PMID: 23783074 PMCID: PMC4046186 DOI: 10.1096/fj.12-214189] [Citation(s) in RCA: 122] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Accepted: 05/28/2013] [Indexed: 12/11/2022]
Abstract
Fibrosis, seen in the liver, lung, heart, kidney, and skin, is a significant global disease burden. Currently, therapeutic treatment is limited, and the number of cases continues to grow. Apoptosis has been identified as a potential initiator and propagator of fibrosis. This review specifically examines the correlation between the presence of apoptotic cells and their effect on fibroblast phenotype and collagen metabolism in several different experimental models of fibrosis. Fibrosis in these models is generally preceded by robust angiogenesis and vascular regression, suggesting that the vascular apoptotic burden may be important to fibrotic outcomes. This review considers the emerging evidence that angiogenesis or vascular regression contributes to fibrosis and identifies initial vascular outgrowth or vascular apoptotic cell presence as possible regulators of fibrosis. A further understanding of the cellular mechanisms of fibrosis may suggest novel methods for the reduction of the fibrotic response and promotion of regeneration.
Collapse
Affiliation(s)
- Ariel Johnson
- 1University of Illinois at Chicago, College of Dentistry, Center for Wound Healing and Tissue Regeneration (MC 859), 801 S. Paulina, Rm. 401B, Chicago, IL 60612-7211, USA.
| | | |
Collapse
|
35
|
Kim DS, Kang SI, Lee SY, Noh KT, Kim EC. Involvement of SDF-1 and monocyte chemoattractant protein-1 in hydrogen peroxide-induced extracellular matrix degradation in human dental pulp cells. Int Endod J 2013; 47:298-308. [DOI: 10.1111/iej.12147] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Accepted: 06/03/2013] [Indexed: 01/20/2023]
Affiliation(s)
- D.-S. Kim
- Department of Conservative Dentistry; School of Dentistry and Institute of Oral Biology; Kyung Hee University; Seoul Korea
| | - S. I. Kang
- Department of Maxillofacial Tissue Regeneration; School of Dentistry; Kyung Hee University; Seoul Korea
| | - S.-Y. Lee
- Department of Maxillofacial Tissue Regeneration; School of Dentistry; Kyung Hee University; Seoul Korea
| | - K.-T. Noh
- Department of Prosthodontics; School of Dentistry; Kyung Hee University; Seoul Korea
| | - E.-C. Kim
- Department of Maxillofacial Tissue Regeneration; School of Dentistry; Kyung Hee University; Seoul Korea
| |
Collapse
|
36
|
Sahin H, Wasmuth HE. Chemokines in tissue fibrosis. Biochim Biophys Acta Mol Basis Dis 2012; 1832:1041-8. [PMID: 23159607 DOI: 10.1016/j.bbadis.2012.11.004] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Revised: 11/06/2012] [Accepted: 11/08/2012] [Indexed: 12/20/2022]
Abstract
Fibrosis or scarring of diverse organs and tissues is considered as a pathologic consequence of a chronically altered wound healing response which is tightly linked to inflammation and angiogenesis. The recruitment of immune cells, local proliferation of fibroblasts and the consecutive accumulation of extracellular matrix proteins are common pathophysiological hallmarks of tissue fibrosis, irrespective of the organ involved. Chemokines, a family of chemotactic cytokines, appear to be central mediators of the initiation as well as progression of these biological processes. Traditionally chemokines have only been considered to play a critical role in orchestrating the influx of immune cells to sites of tissue injury. However, within the last years, further aspects of chemokine biology including fibroblast activation and angiogenesis have been deciphered in tissue fibrosis of many different organs. Interestingly, certain chemokines appear to mediate common effects in liver, kidney, lung, and skin of various animal models, while others mediate tissue specific effects. These aspects have to be kept in mind when extrapolating data of animal studies to early human trials. Nevertheless, the further understanding of chemokine effects in tissue fibrosis might be an attractive approach for identifying novel therapeutic targets in chronic organ damage associated with high morbidity and mortality. This article is part of a Special Issue entitled: Fibrosis: Translation of basic research to human disease.
Collapse
Affiliation(s)
- Hacer Sahin
- Medical Department III, University Hospital Aachen, RWTH Aachen, Germany
| | | |
Collapse
|
37
|
Lagares D, Busnadiego O, García-Fernández RA, Lamas S, Rodríguez-Pascual F. Adenoviral gene transfer of endothelin-1 in the lung induces pulmonary fibrosis through the activation of focal adhesion kinase. Am J Respir Cell Mol Biol 2012; 47:834-42. [PMID: 22962065 DOI: 10.1165/rcmb.2011-0446oc] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Endothelin-1 (ET-1) has been implicated in the development of pulmonary fibrosis, based on its capacity in vitro to promote extracellular matrix (ECM) production and contraction, and on studies showing elevated expression of ET-1 and its receptors in patients with pulmonary fibrosis. However, the in vivo fibrogenic effect of ET-1 is not well characterized. We used the adenoviral-mediated gene transfer of ET-1 to overexpress ET-1 transiently in murine lungs by intratracheal administration. An increased expression of ET-1 for 3 to 10 days after injection resulted in a moderate but reversible fibrotic response, peaking on Day 14 after infection and characterized by the deposition of ECM components, myofibroblast formation, and a significant inflammatory infiltrate, mainly in the peribronchiolar/perivascular region. Adenoviral-mediated ET-1 overexpression activated focal adhesion kinase (FAK) both in vitro, using primary murine lung fibroblasts, and in vivo, intratracheally administered in the lungs of mice. The inhibition of FAK with the compound PF-562,271 prevented ET-1-mediated collagen deposition and myofibroblast formation, thereby preventing the development of lung fibrosis. In conclusion, we demonstrate that the overexpression of ET-1 directly in the lungs of mice can initiate a fibrogenic response characterized by increased ECM deposition and myofibroblast formation, and that this effect of ET-1 can be prevented by inhibition of FAK. Our data suggest that the ET-1/FAK axis may contribute importantly to the pathogenesis of fibrotic disorders, and highlight FAK as a potential therapeutic target in these devastating diseases.
Collapse
Affiliation(s)
- David Lagares
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain
| | | | | | | | | |
Collapse
|
38
|
Gao J, Feng LJ, Huang Y, Li P, Xu DJ, Li J, Wu Q. Total glucosides of Danggui Buxue Tang attenuates bleomycin-induced pulmonary fibrosis via inhibition of extracellular matrix remodelling. ACTA ACUST UNITED AC 2012; 64:811-20. [PMID: 22571259 DOI: 10.1111/j.2042-7158.2012.01490.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
OBJECTIVES This study was designed to investigate the antifibrosis effects and possible mechanism of action of total glucosides of Danggui Buxue Tang (DBTG) on bleomycin-induced pulmonary fibrosis in rats. METHODS DBTG was extracted from Radix Astragali and Radix Angelicae Sinensis. Pulmonary fibrosis was induced by intratracheal instillation of bleomycin (5 mg/kg) in Wistar rats. Subsequently, the rats received daily intragastric administration of DBTG (16, 32 or 64 mg/kg per day) or cortisone (3 mg/kg) 1 day after bleomycin instillation for 4 weeks. Histological changes in the lung were evaluated by hematoxylin and eosin and Masson's trichrome staining. Markers of fibrosis in serum were determined by radioimmunoassay. The mRNA expression of metalloproteinases 1 and 9 (MMP-1, MMP-9) and tissue inhibitor of metalloproteinase 1 (TIMP-1) in lung tissue were detected by reverse transcription PCR. KEY FINDINGS DBTG administration attenuated the degree of alveolitis and lung fibrosis, and markedly reduced the elevated levels of hyaluronic acid, laminin, type III procollagen and type IV collagen in serum. DBTG decreased the mRNA levels of MMP-9 and TIMP-1. MMP-1 expression was only moderately decreased by DBTG. CONCLUSIONS DBTG had an inhibitory effect on bleomycin-induced pulmonary fibrosis and its effect may be associated with the ability of DBTG to inhibit the synthesis of extracellular matrix and balance the MMP/TIMP-1 system.
Collapse
Affiliation(s)
- Jian Gao
- Pharmaceutical Preparation Section, Third-Grade Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine-TCM-2009-202, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
| | | | | | | | | | | | | |
Collapse
|
39
|
Ghosh MC, Makena PS, Gorantla V, Sinclair SE, Waters CM. CXCR4 regulates migration of lung alveolar epithelial cells through activation of Rac1 and matrix metalloproteinase-2. Am J Physiol Lung Cell Mol Physiol 2012; 302:L846-56. [PMID: 22345572 DOI: 10.1152/ajplung.00321.2011] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Restoration of the epithelial barrier following acute lung injury is critical for recovery of lung homeostasis. After injury, alveolar type II epithelial (ATII) cells spread and migrate to cover the denuded surface and, eventually, proliferate and differentiate into type I cells. The chemokine CXCL12, also known as stromal cell-derived factor 1α, has well-recognized roles in organogenesis, hematopoiesis, and immune responses through its binding to the chemokine receptor CXCR4. While CXCL12/CXCR4 signaling is known to be important in immune cell migration, the role of this chemokine-receptor interaction has not been studied in alveolar epithelial repair mechanisms. In this study, we demonstrated that secretion of CXCL12 was increased in the bronchoalveolar lavage of rats ventilated with an injurious tidal volume (25 ml/kg). We also found that CXCL12 secretion was increased by primary rat ATII cells and a mouse alveolar epithelial (MLE12) cell line following scratch wounding and that both types of cells express CXCR4. CXCL12 significantly increased ATII cell migration in a scratch-wound assay. When we treated cells with a specific antagonist for CXCR4, AMD-3100, cell migration was significantly inhibited. Knockdown of CXCR4 by short hairpin RNA (shRNA) caused decreased cell migration compared with cells expressing a nonspecific shRNA. Treatment with AMD-3100 decreased matrix metalloproteinase-14 expression, increased tissue inhibitor of metalloproteinase-3 expression, decreased matrix metalloproteinase-2 activity, and prevented CXCL12-induced Rac1 activation. Similar results were obtained with shRNA knockdown of CXCR4. These findings may help identify a therapeutic target for augmenting epithelial repair following acute lung injury.
Collapse
Affiliation(s)
- Manik C Ghosh
- Department of Physiology, Univ. of Tennessee Health Science Center, Memphis, TN 38163, USA
| | | | | | | | | |
Collapse
|
40
|
Daidzein attenuates inflammation and exhibits antifibrotic effect against Bleomycin-induced pulmonary fibrosis in Wistar rats. ACTA ACUST UNITED AC 2011. [DOI: 10.1016/j.bionut.2011.09.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
41
|
Taguchi T, Nazneen A, Al-Shihri AA, A. Turkistani K, Razzaque MS. Heat shock protein 47: a novel biomarker of phenotypically altered collagen-producing cells. Acta Histochem Cytochem 2011; 44:35-41. [PMID: 21614164 PMCID: PMC3096080 DOI: 10.1267/ahc.11001] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2010] [Accepted: 02/18/2011] [Indexed: 01/15/2023] Open
Abstract
Heat shock protein 47 (HSP47) is a collagen-specific molecular chaperone that helps the molecular maturation of various types of collagens. A close association between increased expression of HSP47 and the excessive accumulation of collagens is found in various human and experimental fibrotic diseases. Increased levels of HSP47 in fibrotic diseases are thought to assist in the increased assembly of procollagen, and thereby contribute to the excessive deposition of collagens in fibrotic areas. Currently, there is not a good universal histological marker to identify collagen-producing cells. Identifying phenotypically altered collagen-producing cells is essential for the development of cell-based therapies to reduce the progression of fibrotic diseases. Since HSP47 has a single substrate, which is collagen, the HSP47 cellular expression provides a novel universal biomarker to identify phenotypically altered collagen-producing cells during wound healing and fibrosis. In this brief article, we explained why HSP47 could be used as a universal marker for identifying phenotypically altered collagen-producing cells.
Collapse
Affiliation(s)
- Takashi Taguchi
- Department of Pathology, Nagasaki University Graduate School of Biomedical Sciences
| | - Arifa Nazneen
- Department of Pathology, Nagasaki University Graduate School of Biomedical Sciences
| | - Abdulmonem A. Al-Shihri
- Department of Restorative Dentistry and Biomaterials Sciences, Harvard School of Dental Medicine
| | | | - Mohammed S. Razzaque
- Department of Pathology, Nagasaki University Graduate School of Biomedical Sciences
- Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine
| |
Collapse
|
42
|
Buckley ST, Medina C, Ehrhardt C. Differential susceptibility to epithelial-mesenchymal transition (EMT) of alveolar, bronchial and intestinal epithelial cells in vitro and the effect of angiotensin II receptor inhibition. Cell Tissue Res 2010; 342:39-51. [PMID: 20848133 DOI: 10.1007/s00441-010-1029-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2010] [Accepted: 07/23/2010] [Indexed: 01/14/2023]
Abstract
The generation of myofibroblasts via epithelial-mesenchymal transition (EMT), a process through which epithelial cells lose their polarity and become motile mesenchymal cells, is a proposed contributory factor in fibrosis of a number of organs. Currently, it remains unclear to what extent epithelia of the upper airways and large intestine are susceptible to this process. Herein, we investigated the ability of model cell lines of alveolar (A549), bronchial (Calu-3) and colonic (Caco-2) epithelial cells to undergo EMT when challenged with transforming growth factor-β1 (TGF-β1) and other pro-inflammatory cytokines. Western blot and immunofluorescence microscopy demonstrated that A549 cells readily underwent EMT, as evidenced by a spindle-like morphology, increase in the mesenchymal marker, vimentin, and down-regulation of E-cadherin, an epithelial marker. In contrast, neither Calu-3 nor Caco-2 cells exhibited morphological changes nor alterations in marker expression associated with EMT. Moreover, whilst stimulation of A549 cells enhanced migration and reduced their proliferative capacity, no such effect was observed in epithelial cell lines of the bronchus or colon. In addition, concomitant treatment of A549 cells with telmisartan, an angiotensin II receptor antagonist with antifibrotic properties, was found to reduce cytokine-induced collagen I production and cell migration, although expression levels of vimentin and E-cadherin remained unaltered. Mechanistically, telmisartan failed to inhibit phosphorylation of Smad2/3. Together, these results, using representative in vitro models of the alveolus, bronchus and colon, tentatively suggest that epithelial cell plasticity and susceptibility to EMT may differ depending on its tissue origin. Furthermore, our investigations point to the beneficial effect of telmisartan in partial abrogation of alveolar EMT.
Collapse
Affiliation(s)
- Stephen T Buckley
- School of Pharmacy and Pharmaceutical Sciences, Panoz Institute, Trinity College Dublin, Dublin 2, Ireland
| | | | | |
Collapse
|
43
|
Russo RC, Alessandri AL, Garcia CC, Cordeiro BF, Pinho V, Cassali GD, Proudfoot AEI, Teixeira MM. Therapeutic effects of evasin-1, a chemokine binding protein, in bleomycin-induced pulmonary fibrosis. Am J Respir Cell Mol Biol 2010; 45:72-80. [PMID: 20833968 DOI: 10.1165/rcmb.2009-0406oc] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
CC chemokines play an important role in the pathogenesis of idiopathic pulmonary fibrosis. Few studies have evaluated the efficacy of therapeutically targeting CC chemokines and their receptors during interstitial lung diseases. In the present study, the therapeutic effects of Evasin-1, a tick-derived chemokine-binding protein that has high affinity for CCL3/microphage inflammatory protein (MIP)-1α, was investigated in a murine model of bleomycin-induced lung fibrosis. CCL3/MIP-1α concentrations in lung homogenates increased significantly with time after bleomycin challenge, and this was accompanied by increased number of leukocytes and elevated levels of CCL2/monocyte chemoattractant protein (MCP)-1, CCL5/regulated upon activation, normal T cell expressed and secreted, TNF-α and transforming growth factor-β(1), and pulmonary fibrosis. Administration of evasin-1 on a preventive (from the day of bleomycin administration) or therapeutic (from Day 8 after bleomycin) schedule decreased number of leukocytes in the lung, reduced levels of TNF-α and transforming growth factor-β(1), and attenuated lung fibrosis. These protective effects were similar to those observed in CCL3/MIP-1α-deficient mice. In conclusion, targeting CCL3/MIP-1α by treatment with evasin-1 is beneficial in the context of bleomycin-induced lung injury, even when treatment is started after the fibrogenic insult. Mechanistically, evasin-1 treatment was associated with decreased recruitment of leukocytes and production of fibrogenic cytokines. Modulation of CCL3/MIP-1α function by evasin-1 could be useful for the treatment of idiopathic pulmonary fibrosis.
Collapse
Affiliation(s)
- Remo C Russo
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas-Universidade Federal de Minas Gerais, Avenida Antônio Carlos, 6627, CEP 30882-650-Pampulha, Belo Horizonte, MG, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Swigris JJ, Brown KK. The role of endothelin-1 in the pathogenesis of idiopathic pulmonary fibrosis. BioDrugs 2010; 24:49-54. [PMID: 20055532 DOI: 10.2165/11319550-000000000-00000] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The endothelin system participates in a number of critical biologic pathways, including normal wound healing. In addition, emerging basic science, and animal and human data all suggest that endothelin-1 (EDN1, also known as ET-1) is a potentially important contributor in the pathobiology of fibrosing disorders, including those that affect the lung. For example, EDN1 drives fibroblast activation, proliferation, as well as differentiation into myofibroblasts - processes that lead to excessive collagen deposition. Patients with idiopathic pulmonary fibrosis (IPF) have increased levels of EDN1 in both their bronchoalveolar lavage fluid and lung tissue. Beyond this, rodent models suggest that endothelin receptor antagonists can limit bleomycin-induced lung fibrosis. This suggests a biologic rationale for the blockade of EDN1 to limit the evolution of lung fibrosis in humans. Initial results from a trial examining the efficacy of a dual endothelin receptor antagonist suggest that this approach may delay disease progression in a subset of patients with IPF.
Collapse
Affiliation(s)
- Jeffrey J Swigris
- Interstitial Lung Disease Program, National Jewish Health, Denver, Colorado 80206, USA.
| | | |
Collapse
|
45
|
Lee JS, Shin JH, Lee JO, Lee WJ, Hwang JH, Kim JH, Choi BS. Blood Levels of IL-Iβ, IL-6, IL-8, TNF-α, and MCP-1 in Pneumoconiosis Patients Exposed to Inorganic Dusts. Toxicol Res 2009; 25:217-224. [PMID: 32038841 PMCID: PMC7006313 DOI: 10.5487/tr.2009.25.4.217] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2009] [Revised: 11/05/2009] [Accepted: 11/16/2009] [Indexed: 11/29/2022] Open
Abstract
Inhaled inorganic dusts such as coal can cause inflammation and fibrosis in the lung called pneumoconiosis. Chronic inflammatory process in the lung is associated with various cytokines and reactive oxygen species (ROS) formation. Expression of some cytokines mediates inflammation and leads to tissue damage or fibrosis. The aim of the present study was to compare the levels of blood cytokines interleukin (IL)-1β, IL-6, IL-8, tumor necrosis factor (TNF)-α and monocyte chemoattractant protein (MCP)-1 among 124 subjects (control 38 and pneumoconiosis patient 86) with category of chest x-ray according to International Labor Organization (ILO) classification. The levels of serum IL-8 (p = 0.003), TNF-α (p = 0.026), and MCP-1 (p = 0.010) of pneumoconiosis patients were higher than those of subjects with the control. The level of serum IL-8 in the severe group with the small opacity (ILO category II or III) was higher than that of the control (p = 0.035). There was significant correlation between the profusion of radiological findings with small opacity and serum levels of IL-1β (rho = 0.218, p < 0.05), IL-8 (rho = 0.224, p < 0.05), TNF-α (rho = 0.306, p < 0.01), and MCP-1 (rho = 0.213, p < 0.01). The serum levels of IL-6 and IL-8, however, did not show significant difference between pneumoconiosis patients and the control. There was no significant correlation between serum levels of measured cytokines and other associated variables such as lung function, age, BMI, and exposure period of dusts. Future studies will be required to investigate the cytokine profile that is present in pneumoconiosis patient using lung specific specimens such as bronchoalveolar lavage fluid (BALF), exhaled breath condensate, and lung tissue.
Collapse
Affiliation(s)
- Jong Seong Lee
- Center for Occupational Lung Diseases, KWAMCO, 95, II-dong, Sangnok-gu, Ansan-si, Gyeonggi-do, 426-858 Korea
| | - Jae Hoon Shin
- Center for Occupational Lung Diseases, KWAMCO, 95, II-dong, Sangnok-gu, Ansan-si, Gyeonggi-do, 426-858 Korea
| | - Joung Oh Lee
- Center for Occupational Lung Diseases, KWAMCO, 95, II-dong, Sangnok-gu, Ansan-si, Gyeonggi-do, 426-858 Korea
| | - Won-Jeong Lee
- Center for Occupational Lung Diseases, KWAMCO, 95, II-dong, Sangnok-gu, Ansan-si, Gyeonggi-do, 426-858 Korea
| | - Joo-Hwan Hwang
- Center for Occupational Lung Diseases, KWAMCO, 95, II-dong, Sangnok-gu, Ansan-si, Gyeonggi-do, 426-858 Korea
| | - Ji Hong Kim
- Ansan Choongang General Hospital, KWAMCO, Ansan, 426-858 Korea
| | - Byung-Soon Choi
- Center for Occupational Lung Diseases, KWAMCO, 95, II-dong, Sangnok-gu, Ansan-si, Gyeonggi-do, 426-858 Korea
| |
Collapse
|
46
|
Abstract
Severe acute respiratory syndrome (SARS) is an acute infectious disease with significant mortality. A novel coronavirus (SARS-CoV) has been shown to be the causative agent of SARS. The typical clinical feature associated with SARS is diffuse alveolar damage in lung, and lung fibrosis is evident in patients who died from this disease. The mechanisms by which SARS-CoV infection causes lung fibrosis are not fully understood, but transforming growth factor-β (TGF-β) and angiotensin-converting enzyme 2 (ACE2)-mediated lung fibrosis are among the most documented ones. The activation of the TGF-β/Smad pathway is critical to lung fibrosis. SARS-CoV infection not only enhances the expression of TGF-β, but also facilitates its signaling activity. The SARS-CoV receptor ACE2 is a negative regulator of lung fibrosis, and SARS-CoV infection decreases ACE2 expression. Therefore, SARS-CoV infection may lead to lung fibrosis through multiple signaling pathways and TGF-β activation is one of the major contributors.
Collapse
|
47
|
Ji WJ, Zhou X, Zeng S, Wang SX. ATTENUATION OF SILICA-INDUCED PULMONARY FIBROBLASTS PROLIFERATION BY TAURINE AND NIACIN IN VITRO. Exp Lung Res 2009; 35:198-209. [DOI: 10.1080/01902140802499401] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
48
|
Russo RC, Guabiraba R, Garcia CC, Barcelos LS, Roffê E, Souza ALS, Amaral FA, Cisalpino D, Cassali GD, Doni A, Bertini R, Teixeira MM. Role of the chemokine receptor CXCR2 in bleomycin-induced pulmonary inflammation and fibrosis. Am J Respir Cell Mol Biol 2008; 40:410-21. [PMID: 18836137 DOI: 10.1165/rcmb.2007-0364oc] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Pulmonary fibrosis is characterized by chronic inflammation and excessive collagen deposition. Neutrophils are thought to be involved in the pathogenesis of lung fibrosis. We hypothesized that CXCR2-mediated neutrophil recruitment is essential for the cascade of events leading to bleomycin-induced pulmonary fibrosis. CXCL1/KC was detected as early as 6 hours after bleomycin instillation and returned to basal levels after Day 8. Neutrophils were detected in bronchoalveolar lavage and interstitium from 12 hours and peaked at Day 8 after instillation. Treatment with the CXCR2 receptor antagonist, DF2162, reduced airway neutrophil transmigration but led to an increase of neutrophils in lung parenchyma. There was a significant reduction in IL-13, IL-10, CCL5/RANTES, and active transforming growth factor (TGF)-beta(1) levels, but not on IFN-gamma and total TGF-beta(1,) and enhanced granulocyte macrophage-colony-stimulating factor production in DF2162-treated animals. Notably, treatment with the CXCR2 antagonist led to an improvement of the lung pathology and reduced collagen deposition. Using a therapeutic schedule, DF2162 administered from Days 8 to 16 after bleomycin reduced pulmonary fibrosis and levels of active TGF-beta(1) and IL-13. DF2162 treatment reduced bleomycin-induced expression of von Willebrand Factor, a marker of angiogenesis, in the lung. In vitro, DF2162 reduced the angiogenic activity of IL-8 on human umbilical vein endothelial cells. In conclusion, we show that CXCR2 plays an important role in mediating fibrosis after bleomycin instillation. The compound blocks angiogenesis and the production of pro-angiogenic cytokines, and decreases IL-8-induced endothelial cell activation. An effect on neutrophils does not appear to account for the major effects of the blockade of CXCR2 in the system.
Collapse
Affiliation(s)
- Remo C Russo
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Avenida Antônio Carlos 6627, CEP 30882-650-Pampulha, Belo Horizonte, MG, Brazil
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
|
50
|
Antao-Menezes A, Turpin EA, Bost PC, Ryman-Rasmussen JP, Bonner JC. STAT-1 signaling in human lung fibroblasts is induced by vanadium pentoxide through an IFN-beta autocrine loop. THE JOURNAL OF IMMUNOLOGY 2008; 180:4200-7. [PMID: 18322232 DOI: 10.4049/jimmunol.180.6.4200] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The inhalation of vanadium pentoxide (V(2)O(5)) results in bronchitis and airway fibrosis. The lung fibrotic response to V(2)O(5) partially resolves where fibroblasts first proliferate and deposit collagen, but then undergo growth arrest and apoptosis. STAT-1 mediates fibroblast growth arrest and apoptosis. We previously reported that STAT-1 is a protective factor and mice lacking STAT-1 are more susceptible to lung fibrosis. We also reported that V(2)O(5)-induced STAT-1 phosphorylation in lung fibroblasts requires H(2)O(2) and de novo protein synthesis. In this study, we identified IFN-beta as the protein that mediates STAT-1 activation by V(2)O(5) in normal human lung fibroblasts and identified NADPH and xanthine oxidase systems as sources of H(2)O(2) that drive IFN-beta gene expression. STAT-1 phosphorylation was decreased with neutralizing Abs to IFN-beta as well as an inhibitor of JAK. V(2)O(5) also increased transcription of an IFN-inducible and STAT-1-dependent chemokine, CXCL10. Inhibition of H(2)O(2)-generating enzyme systems NADPH oxidase by apocynin and xanthine oxidase by allopurinol individually reduced STAT-1 phosphorylation. Apocynin and allopurinol also decreased V(2)O(5)-induced IFN-beta mRNA levels and CXCL10 expression. IFN-alpha transcription was inhibited only by allopurinol. Taken together, these data indicate that fibroblasts play a role in the innate immune response to vanadium-induced oxidative stress by synthesizing IFN-beta and activating STAT-1 to cause growth arrest and increase levels of CXCL10, a potent antifibrotic factor. This mechanism is postulated to counterbalance profibrogenic mechanisms that follow V(2)O(5) injury.
Collapse
|