1
|
Torre A, Martínez‐Sánchez FD, Narvaez‐Chávez SM, Herrera‐Islas MA, Aguilar‐Salinas CA, Córdova‐Gallardo J. Pirfenidone use in fibrotic diseases: What do we know so far? Immun Inflamm Dis 2024; 12:e1335. [PMID: 38967367 PMCID: PMC11225083 DOI: 10.1002/iid3.1335] [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: 02/04/2024] [Revised: 05/27/2024] [Accepted: 06/19/2024] [Indexed: 07/06/2024] Open
Abstract
BACKGROUND Pirfenidone has demonstrated significant anti-inflammatory and antifibrotic effects in both animal models and some clinical trials. Its potential for antifibrotic activity positions it as a promising candidate for the treatment of various fibrotic diseases. Pirfenidone exerts several pleiotropic and anti-inflammatory effects through different molecular pathways, attenuating multiple inflammatory processes, including the secretion of pro-inflammatory cytokines, apoptosis, and fibroblast activation. OBJECTIVE To present the current evidence of pirfenidone's effects on several fibrotic diseases, with a focus on its potential as a therapeutic option for managing chronic fibrotic conditions. FINDINGS Pirfenidone has been extensively studied for idiopathic pulmonary fibrosis, showing a favorable impact and forming part of the current treatment regimen for this disease. Additionally, pirfenidone appears to have beneficial effects on similar fibrotic diseases such as interstitial lung disease, myocardial fibrosis, glomerulopathies, aberrant skin scarring, chronic liver disease, and other fibrotic disorders. CONCLUSION Given the increasing incidence of chronic fibrotic conditions, pirfenidone emerges as a potential therapeutic option for these patients. However, further clinical trials are necessary to confirm its therapeutic efficacy in various fibrotic diseases. This review aims to highlight the current evidence of pirfenidone's effects in multiple fibrotic conditions.
Collapse
Affiliation(s)
- Aldo Torre
- Metabolic UnitInstituto Nacional de Ciencias Médicas y Nutrición “Salvador Zubiran”Mexico CityMexico
| | - Froylan David Martínez‐Sánchez
- Facultad de MedicinaUniversidad Nacional Autonoma de MexicoMexico CityMexico
- Department of Internal MedicineHospital General “Dr. Manuel Gea González”Mexico CityMexico
| | | | | | | | - Jacqueline Córdova‐Gallardo
- Facultad de MedicinaUniversidad Nacional Autonoma de MexicoMexico CityMexico
- Department of HepatologyHospital General “Dr. Manuel Gea González”Mexico CityMexico
| |
Collapse
|
2
|
Bisen AC, Agrawal S, Rayiti R, Sanap SN, Biswas A, Mishra A, Gupta NM, Bhatta RS. Pirfenidone: A Promising Drug in Ocular Therapeutics. Chem Biodivers 2024; 21:e202301389. [PMID: 38299764 DOI: 10.1002/cbdv.202301389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 01/28/2024] [Accepted: 02/01/2024] [Indexed: 02/02/2024]
Abstract
Pirfenidone, initially indicated for lung fibrosis, has gone beyond its original purpose, and shown promise in eye care. This detailed review tracks its evolution from lung treatment to aiding eye healing as evidenced by published literature. Pirfenidone's multifaceted attributes extend to mitigating corneal fibrosis, inflammation, and trauma. Through rigorous investigations, its efficacy emerges in diabetic retinopathy, macular degeneration, and postoperative glaucoma interventions. As an unheralded protagonist, pirfenidone reshapes ocular care paradigms, inviting renewed research opportunities.
Collapse
Affiliation(s)
- Amol Chhatrapati Bisen
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Sristi Agrawal
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Ramakrishna Rayiti
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Sachin Nashik Sanap
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Arpon Biswas
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Anjali Mishra
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Neeraj Mohan Gupta
- Department of Chemistry, Government P. G. College, Guna, Madhya Pradesh, 473001, India
| | - Rabi Sankar Bhatta
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| |
Collapse
|
3
|
Talpan D, Salla S, Seidelmann N, Walter P, Fuest M. Antifibrotic Effects of Caffeine, Curcumin and Pirfenidone in Primary Human Keratocytes. Int J Mol Sci 2023; 24:ijms24021461. [PMID: 36674976 PMCID: PMC9862324 DOI: 10.3390/ijms24021461] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/09/2023] [Accepted: 01/10/2023] [Indexed: 01/14/2023] Open
Abstract
We evaluated the small molecules (AFM) caffeine, curcumin and pirfenidone to find non-toxic concentrations reducing the transformation of activated human corneal stromal keratocytes (aCSK) to scar-inducing myofibroblasts (MYO-SF). CSK were isolated from 16 human corneas unsuitable for transplantation and expanded for three passages in control medium (0.5% FBS). Then, aCSK were exposed to concentrations of caffeine of 0−500 μM, curcumin of 0−200 μM, pirfenidone of 0−2.2 nM and the profibrotic cytokine TGF-β1 (10 ng/mL) for 48 h. Alterations in viability and gene expression were evaluated by cell viability staining (FDA/PI), real-time polymerase chain reaction (RT-PCR) and immunocytochemistry. We found that all AFMs reduced cell counts at high concentrations. The highest concentrations with no toxic effect were 100 µM of caffeine, 20 µM of curcumin and 1.1 nM of pirfenidone. The addition of TGF-β1 to the control medium effectively transformed aCSK into myofibroblasts (MYO-SF), indicated by a 10-fold increase in α-smooth muscle actin (SMA) expression, a 39% decrease in lumican (LUM) expression and a 98% decrease in ALDH3A1 expression (p < 0.001). The concentrations of 100 µM of caffeine, 20/50 µM of curcumin and 1.1 nM of pirfenidone each significantly reduced SMA expression under TGF-β1 stimulation (p ≤ 0.024). LUM and ALDH3A1 expression remained low under TGF-β1 stimulation, independently of AFM supplementation. Immunocytochemistry showed that 100 µM of caffeine, 20 µM of curcumin and 1.1 nM of pirfenidone reduce the conversion rate of aCSK to SMA+ MYO-SF. In conclusion, in aCSK, 100 µM of caffeine, 20 µM of curcumin and 1.1 nM of pirfenidone significantly reduced SMA expression and MYO-SF conversion under TGF-β1 stimulation, with no influence on cell counts. However, the AFMs were unable to protect aCSK from characteristic marker loss.
Collapse
Affiliation(s)
- Delia Talpan
- Department of Ophthalmology, RWTH Aachen University, 52074 Aachen, Germany
| | - Sabine Salla
- Department of Ophthalmology, RWTH Aachen University, 52074 Aachen, Germany
- Cornea Bank Aachen, RWTH Aachen University, 52074 Aachen, Germany
| | - Nina Seidelmann
- Department of Ophthalmology, RWTH Aachen University, 52074 Aachen, Germany
| | - Peter Walter
- Department of Ophthalmology, RWTH Aachen University, 52074 Aachen, Germany
- Cornea Bank Aachen, RWTH Aachen University, 52074 Aachen, Germany
| | - Matthias Fuest
- Department of Ophthalmology, RWTH Aachen University, 52074 Aachen, Germany
- Cornea Bank Aachen, RWTH Aachen University, 52074 Aachen, Germany
- Correspondence:
| |
Collapse
|
4
|
Gupta V, Hammond CL, Roztocil E, Gonzalez MO, Feldon SE, Woeller CF. Thinking inside the box: Current insights into targeting orbital tissue remodeling and inflammation in thyroid eye disease. Surv Ophthalmol 2022; 67:858-874. [PMID: 34487739 PMCID: PMC8891393 DOI: 10.1016/j.survophthal.2021.08.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 08/24/2021] [Accepted: 08/30/2021] [Indexed: 12/21/2022]
Abstract
Thyroid eye disease (TED) is an autoimmune disorder that manifests in the orbit. In TED, the connective tissue behind the eye becomes inflamed and remodels with increased fat accumulation and/or increased muscle and scar tissue. As orbital tissue expands, patients develop edema, exophthalmos, diplopia, and optic neuropathy. In severe cases vision loss may occur secondary to corneal scarring from exposure or optic nerve compression. Currently there is no cure for TED, and treatments are limited. A major breakthrough in TED therapy occurred with the FDA approval of teprotumumab, a monoclonal insulin-like growth factor 1 receptor (IGF1R) blocking antibody. Yet, teprotumumab therapy has limitations, including cost, infusion method of drug delivery, variable response, and relapse. We describe approaches to target orbital fibroblasts and the complex pathophysiology that underlies tissue remodeling and inflammation driving TED. Further advances in the elucidation of the mechanisms of TED may lead to prophylaxis based upon early biomarkers as well as lead to more convenient, less expensive therapies.
Collapse
Affiliation(s)
- Vardaan Gupta
- Flaum Eye Institute, University of Rochester, 210 Crittenden Boulevard, Rochester, New York 14642, USA
| | - Christine L Hammond
- Flaum Eye Institute, University of Rochester, 210 Crittenden Boulevard, Rochester, New York 14642, USA
| | - Elisa Roztocil
- Flaum Eye Institute, University of Rochester, 210 Crittenden Boulevard, Rochester, New York 14642, USA
| | - Mithra O Gonzalez
- Flaum Eye Institute, University of Rochester, 210 Crittenden Boulevard, Rochester, New York 14642, USA
| | - Steven E Feldon
- Flaum Eye Institute, University of Rochester, 210 Crittenden Boulevard, Rochester, New York 14642, USA
| | - Collynn F Woeller
- Flaum Eye Institute, University of Rochester, 210 Crittenden Boulevard, Rochester, New York 14642, USA.
| |
Collapse
|
5
|
Niklander SE. Inflammatory Mediators in Oral Cancer: Pathogenic Mechanisms and Diagnostic Potential. FRONTIERS IN ORAL HEALTH 2022; 2:642238. [PMID: 35047997 PMCID: PMC8757707 DOI: 10.3389/froh.2021.642238] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 01/13/2021] [Indexed: 12/12/2022] Open
Abstract
Approximately 15% of cancers are attributable to the inflammatory process, and growing evidence supports an association between oral squamous cell carcinoma (OSCC) and chronic inflammation. Different oral inflammatory conditions, such as oral lichen planus (OLP), submucous fibrosis, and oral discoid lupus, are all predisposing for the development of OSCC. The microenvironment of these conditions contains various transcription factors and inflammatory mediators with the ability to induce proliferation, epithelial-to-mesenchymal transition (EMT), and invasion of genetically predisposed lesions, thereby promoting tumor development. In this review, we will focus on the main inflammatory molecules and transcription factors activated in OSCC, with emphasis on their translational potential.
Collapse
Affiliation(s)
- Sven E Niklander
- Unidad de Patologia y Medicina Oral, Facultad de Odontologia, Universidad Andres Bello, Viña del Mar, Chile
| |
Collapse
|
6
|
Fatty acid nitroalkene reversal of established lung fibrosis. Redox Biol 2021; 50:102226. [PMID: 35150970 PMCID: PMC8844680 DOI: 10.1016/j.redox.2021.102226] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 12/17/2021] [Accepted: 12/27/2021] [Indexed: 02/06/2023] Open
Abstract
Tissue fibrosis occurs in response to dysregulated metabolism, pro-inflammatory signaling and tissue repair reactions. For example, lungs exposed to environmental toxins, cancer therapies, chronic inflammation and other stimuli manifest a phenotypic shift to activated myofibroblasts and progressive and often irreversible lung tissue scarring. There are no therapies that stop or reverse fibrosis. The 2 FDA-approved anti-fibrotic drugs at best only slow the progression of fibrosis in humans. The present study was designed to test whether a small molecule electrophilic nitroalkene, nitro-oleic acid (NO2-OA), could reverse established pulmonary fibrosis induced by the intratracheal administration of bleomycin in C57BL/6 mice. After 14 d of bleomycin-induced fibrosis development in vivo, lungs were removed, sectioned and precision-cut lung slices (PCLS) from control and bleomycin-treated mice were cultured ex vivo for 4 d with either vehicle or NO2-OA (5 μM). Biochemical and morphological analyses showed that over a 4 d time frame, NO2-OA significantly inhibited pro-inflammatory mediator and growth factor expression and reversed key indices of fibrosis (hydroxyproline, collagen 1A1 and 3A1, fibronectin-1). Quantitative image analysis of PCLS immunohistology reinforced these observations, revealing that NO2-OA suppressed additional hallmarks of the fibrotic response, including alveolar epithelial cell loss, myofibroblast differentiation and proliferation, collagen and α-smooth muscle actin expression. NO2-OA also accelerated collagen degradation by resident macrophages. These effects occurred in the absence of the recognized NO2-OA modulation of circulating and migrating immune cell activation. Thus, small molecule nitroalkenes may be useful agents for reversing pathogenic fibrosis of lung and other organs. Small molecule electrophiles, pleiotropic anti-inflammatory and anti-fibrotic drugs. NO2-OA inhibits activated myofibroblasts, induces dedifferentiation to fibroblasts. NO2-OA activates extracellular matrix degradation by macrophages. NO2-OA promotes proliferation of alveolar type 1 and 2 epithelial cells. NO2-OA reverses established lung fibrosis in murine lung slices.
Collapse
|
7
|
Corrie L, Muzaffar-Ur-Rehman MD, Kukatil L, Manasa D, Shirisha A. Antifibrotic Drugs for COVID-19: From Orphan Drugs to Blockbusters? CURRENT RESPIRATORY MEDICINE REVIEWS 2021. [DOI: 10.2174/1573398x17666210304100043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
:
Antifibrotic agents are known to treat idiopathic pulmonary fibrosis. The two antifibrotic
agents approved and in usage are Pirfenidone and Nintedanib granted by the USFDA in 2014.
They are both known to decrease inflammation in the lungs. The fact that COVID-19 has shown to
cause inflammation and fibrosis in the lungs frames the theory of their usage in the treatment of the
disease by reducing lung scaring and allowing faster discharge of patients with post-COVID complications.
The need for them to change their status from orphans to blockbusters has not happened
yet due to fewer data and less research available on them as well as various other economic and patient-
related factors. Since COVID-19 is widespread and causes many complications of the lungs
that are similar to what these two drugs treat. We believe that the status of these drugs could be
changed due to an increase in demand for them.
Collapse
Affiliation(s)
- Leander Corrie
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - MD Muzaffar-Ur-Rehman
- Nalla Narasimha Reddy Educational Society, Chowdariguda, Narapally, Telangana State, 500088, India
| | - Latha Kukatil
- G. Pulla Reddy College of Pharmacy, Pillar No : 23 (PVNR elevated Expressway) Mehdipatnam, Hyderabad, Telangana State, 500028, India
| | - Devasari Manasa
- G. Pulla Reddy College of Pharmacy, Pillar No : 23 (PVNR elevated Expressway) Mehdipatnam, Hyderabad, Telangana State, 500028, India
| | - Adepu Shirisha
- G. Pulla Reddy College of Pharmacy, Pillar No : 23 (PVNR elevated Expressway) Mehdipatnam, Hyderabad, Telangana State, 500028, India
| |
Collapse
|
8
|
Ma Y, Luo L, Liu X, Li H, Zeng Z, He X, Zhan Z, Chen Y. Pirfenidone mediates cigarette smoke extract induced inflammation and oxidative stress in vitro and in vivo. Int Immunopharmacol 2021; 96:107593. [PMID: 33819731 DOI: 10.1016/j.intimp.2021.107593] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 03/09/2021] [Accepted: 03/12/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Antioxidant and anti-inflammatory effects are two main pharmacological mechanisms of pirfenidone (PFD) besides the anti-fibrotic effect. This study aims to investigate whether PFD could mediate cigarette smoke extract (CSE) induced inflammation and oxidative stress in vitro and in vivo. METHODS BALB/C mice and alveolar epithelial (A549) cells treated with CSE were established as disease models in vivo and in vitro. Effects of PFD treatment on disease models were further measured. Hematoxylin and eosin (HE) staining was used to evaluate the pathological changes in lung tissues of mice. CCK-8 assay kit was applied to measure the viability of A549 cells treated by different concentrations of PFD. Inflammation cytokine expression in cell supernatants was measured with ELISA kits. The mRNA and protein levels of inflammation and oxidative stress-related factors were determined by real-time quantitative polymerase chain reaction analysis (RT-qPCR) and Western blotting. Furthermore, myeloperoxidase (MPO), malondialdehyde (MDA), and total antioxidant capacity (T-AOC) were measured to detect the antioxidative activity of lung tissues. Moreover, an assay kit with fluorescent probe 2',7'-dichlorofluorescin diacetate (DCFH-DA) was used to evaluate the intracellular reactive oxygen species (ROS) generation. RESULTS In vitro and in vivo, PFD significantly reversed TNF-α, IL-6, CCL2, SOD1, and CAT mRNA level changes led by CSE; in addition, PFD significantly decreased the ratios of p-p65 to p65, p-ikBα to ikBα and increased Nrf-2 protein level compared with CSE group. In mice, high-dose (100 mg/kg/d) PFD significantly reversed MPO and MDA increases induced by CSE. However, PFD didn't significantly reverse T-AOC decrease induced by CSE. In A549 cell supernatant, PFD dramatically reversed the elevated levels of TNF-α and IL-1β induced by CSE. Furthermore, PFD could significantly reverse the increased level of ROS induced by CSE in A549 cells. CONCLUSION Our study reveals the potential role of PFD in regulating inflammatory response and oxidative stress induced by CSE.
Collapse
Affiliation(s)
- Yiming Ma
- Department of Respiratory and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lijuan Luo
- Department of Respiratory and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiangming Liu
- Department of Respiratory and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Herui Li
- Department of Respiratory and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zihang Zeng
- Department of Respiratory and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xue He
- Department of Respiratory and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zijie Zhan
- Department of Respiratory and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yan Chen
- Department of Respiratory and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
9
|
Gutiérrez-Cuevas J, Sandoval-Rodríguez A, Monroy-Ramírez HC, Vazquez-Del Mercado M, Santos-García A, Armendáriz-Borunda J. Prolonged-release pirfenidone prevents obesity-induced cardiac steatosis and fibrosis in a mouse NASH model. Cardiovasc Drugs Ther 2020; 35:927-938. [PMID: 32621046 DOI: 10.1007/s10557-020-07014-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE Obesity is associated with systemic insulin resistance and cardiac hypertrophy with fibrosis. Peroxisome proliferator-activated receptors (PPARs) regulate carbohydrate and lipid metabolism, improving insulin sensitivity, triglyceride levels, inflammation, and oxidative stress. We previously demonstrated that prolonged-release pirfenidone (PR-PFD) is an agonistic ligand for Pparα with anti-inflammatory and anti-fibrotic effects, and might be a promising drug for cardiac diseases-treatment. Here, we investigated the effects of PR-PFD in ventricular tissue of mice with nonalcoholic steatohepatitis (NASH) and obesity induced by high-fat/high-carbohydrate (HFHC) diet. METHODS Five male C57BL/6 J mice were fed with normal diet (ND) and ten with HFHC diet for 16 weeks; at 8 weeks of feeding, five mice with HFHC diet were administered PR-PFD (350 mg/kg/day) mixed with HFHC diet. RESULT Systemic insulin resistance, heart weight/body weight ratio, myocardial steatosis with inflammatory foci, hypertrophy, and fibrosis were prevented by PR-PFD. In addition, HFHC mice showed significantly increased desmin, Tgfβ1, Timp1, collagen I (Col I), collagen III (Col III), TNF-α, and Nrf2 mRNA levels, including α-SMA, NF-kB, Nrf2, troponin I, Acox1, Cpt1A, and Lxrα protein levels compared with the ND ventricular tissues. Mechanistically, HFHC mice with PR-PFD treatment significantly decreased these genes overexpressed by HFHC diet. Furthermore, PR-PFD overexpressed the Pgc1a mRNA levels and Pparα, Pparγ, Acox1, and Cpt1A protein levels. CONCLUSIONS The results suggest that PR-PFD could be a promising drug for the prevention and treatment of cardiac steatosis and fibrosis induced by obesity.
Collapse
Affiliation(s)
- Jorge Gutiérrez-Cuevas
- Department of Molecular Biology and Genomics, Institute for Molecular Biology in Medicine and Gene Therapy, University of Guadalajara, CUCS, Guadalajara, Jalisco, México
| | - Ana Sandoval-Rodríguez
- Department of Molecular Biology and Genomics, Institute for Molecular Biology in Medicine and Gene Therapy, University of Guadalajara, CUCS, Guadalajara, Jalisco, México
| | - Hugo Christian Monroy-Ramírez
- Department of Molecular Biology and Genomics, Institute for Molecular Biology in Medicine and Gene Therapy, University of Guadalajara, CUCS, Guadalajara, Jalisco, México
| | | | | | - Juan Armendáriz-Borunda
- Department of Molecular Biology and Genomics, Institute for Molecular Biology in Medicine and Gene Therapy, University of Guadalajara, CUCS, Guadalajara, Jalisco, México. .,Tecnologico de Monterrey, Campus Guadalajara, Guadalajara, Jalisco, México.
| |
Collapse
|
10
|
Lee C, Liao J, Chen S, Yen C, Lee Y, Huang S, Huang S, Lin C, Chang VH. Fluorine-Modified Rutaecarpine Exerts Cyclooxygenase-2 Inhibition and Anti-inflammatory Effects in Lungs. Front Pharmacol 2019; 10:91. [PMID: 30792658 PMCID: PMC6374341 DOI: 10.3389/fphar.2019.00091] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 01/23/2019] [Indexed: 12/14/2022] Open
Abstract
Inflammation is the first step that leads to inflammatory cell migration, cytokine release, and myofibroblast formation. Myofibroblasts can deposit excess amounts of extracellular matrix. Cyclooxygenase (COX) inhibitor exhibits strong anti-inflammatory response; however, this is usually achieved with undesirable side effects. In this study, we demonstrated the effects of the fluorine-modified rutaecarpine (RUT), fluoro-2-methoxyrutaecarpine (F-RUT), in inflammatory damage in the lungs. Based on the results, F-RUT retained anti-inflammatory activity both in vitro and in vivo in lungs. Compared to the parent compound, F-RUT showed better COX-2 suppression as a COX-2-selective inhibitor with lower cytotoxicity, and enhanced molecular reactivity and biological activity. F-RUT was also observed to reduce reactive oxygen species (ROS) generation and inflammatory infiltrating neutrophils in lipopolysaccharide (LPS)-stimulated zebrafish and ovalbumin (OVA)/alum-challenged KLF-10-knockout mouse lungs, respectively. Furthermore, F-RUT ameliorated the respiratory function in OVA/alum-challenged BALB/c mice by maintaining the thickness of the blood-air barrier in mouse lungs. Overall, these data suggest that F-RUT may function as an effective therapeutic agent for inflammation-induced lung dysfunction, and a better selection for pharmaceutical purposes than conventionally used anti-inflammatory agents.
Collapse
Affiliation(s)
- Chiming Lee
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Jiahnhaur Liao
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Seuhwa Chen
- Department of Anatomy and Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chiaohan Yen
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yuchieh Lee
- Department of Obstetrics and Gynecology, Taipei Medical University Hospital, Taipei, Taiwan
| | - Shihhao Huang
- Department of Food Technology and Marketing Management, Taipei University of Marine Technology, Taipei, Taiwan
| | - Shengtung Huang
- Graduate Institute of Biochemical and Biomedical Engineering, College of Engineering, National Taipei University of Technology, Taipei, Taiwan
| | - Chunmao Lin
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Vincent Hungshu Chang
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
11
|
Berkowski WM, Gibson DJ, Seo S, Proietto LR, Whitley RD, Schultz GS, Plummer CE. Assessment of Topical Therapies for Improving the Optical Clarity Following Stromal Wounding in a Novel Ex Vivo Canine Cornea Model. ACTA ACUST UNITED AC 2018; 59:5509-5521. [DOI: 10.1167/iovs.17-23085] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Affiliation(s)
- William M. Berkowski
- Department of Small Animal Clinical Sciences, University of Florida, Gainesville, Florida, United States
| | - Daniel J. Gibson
- Institute for Wound Research, University of Florida, Gainesville, Florida, United States
| | - SooJung Seo
- Institute for Wound Research, University of Florida, Gainesville, Florida, United States
| | - Laura R. Proietto
- Department of Small Animal Clinical Sciences, University of Florida, Gainesville, Florida, United States
| | - R. David Whitley
- Department of Small Animal Clinical Sciences, University of Florida, Gainesville, Florida, United States
| | - Gregory S. Schultz
- Institute for Wound Research, University of Florida, Gainesville, Florida, United States
| | - Caryn E. Plummer
- Department of Small Animal Clinical Sciences, University of Florida, Gainesville, Florida, United States
| |
Collapse
|
12
|
Kurita Y, Araya J, Minagawa S, Hara H, Ichikawa A, Saito N, Kadota T, Tsubouchi K, Sato N, Yoshida M, Kobayashi K, Ito S, Fujita Y, Utsumi H, Yanagisawa H, Hashimoto M, Wakui H, Yoshii Y, Ishikawa T, Numata T, Kaneko Y, Asano H, Yamashita M, Odaka M, Morikawa T, Nakayama K, Kuwano K. Pirfenidone inhibits myofibroblast differentiation and lung fibrosis development during insufficient mitophagy. Respir Res 2017; 18:114. [PMID: 28577568 PMCID: PMC5457546 DOI: 10.1186/s12931-017-0600-3] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 05/26/2017] [Indexed: 01/23/2023] Open
Abstract
Background Pirfenidone (PFD) is an anti-fibrotic agent used to treat idiopathic pulmonary fibrosis (IPF), but its precise mechanism of action remains elusive. Accumulation of profibrotic myofibroblasts is a crucial process for fibrotic remodeling in IPF. Recent findings show participation of autophagy/mitophagy, part of the lysosomal degradation machinery, in IPF pathogenesis. Mitophagy has been implicated in myofibroblast differentiation through regulating mitochondrial reactive oxygen species (ROS)-mediated platelet-derived growth factor receptor (PDGFR) activation. In this study, the effect of PFD on autophagy/mitophagy activation in lung fibroblasts (LF) was evaluated, specifically the anti-fibrotic property of PFD for modulation of myofibroblast differentiation during insufficient mitophagy. Methods Transforming growth factor-β (TGF-β)-induced or ATG5, ATG7, and PARK2 knockdown-mediated myofibroblast differentiation in LF were used for in vitro models. The anti-fibrotic role of PFD was examined in a bleomycin (BLM)-induced lung fibrosis model using PARK2 knockout (KO) mice. Results We found that PFD induced autophagy/mitophagy activation via enhanced PARK2 expression, which was partly involved in the inhibition of myofibroblast differentiation in the presence of TGF-β. PFD inhibited the myofibroblast differentiation induced by PARK2 knockdown by reducing mitochondrial ROS and PDGFR-PI3K-Akt activation. BLM-treated PARK2 KO mice demonstrated augmentation of lung fibrosis and oxidative modifications compared to those of BLM-treated wild type mice, which were efficiently attenuated by PFD. Conclusions These results suggest that PFD induces PARK2-mediated mitophagy and also inhibits lung fibrosis development in the setting of insufficient mitophagy, which may at least partly explain the anti-fibrotic mechanisms of PFD for IPF treatment.
Collapse
Affiliation(s)
- Yusuke Kurita
- Division of Respiratory Diseases, Department of Internal Medicine, Jikei University School of Medicine, 3-25-8 Nishi-shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Jun Araya
- Division of Respiratory Diseases, Department of Internal Medicine, Jikei University School of Medicine, 3-25-8 Nishi-shimbashi, Minato-ku, Tokyo, 105-8461, Japan.
| | - Shunsuke Minagawa
- Division of Respiratory Diseases, Department of Internal Medicine, Jikei University School of Medicine, 3-25-8 Nishi-shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Hiromichi Hara
- Division of Respiratory Diseases, Department of Internal Medicine, Jikei University School of Medicine, 3-25-8 Nishi-shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Akihiro Ichikawa
- Division of Respiratory Diseases, Department of Internal Medicine, Jikei University School of Medicine, 3-25-8 Nishi-shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Nayuta Saito
- Division of Respiratory Diseases, Department of Internal Medicine, Jikei University School of Medicine, 3-25-8 Nishi-shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Tsukasa Kadota
- Division of Respiratory Diseases, Department of Internal Medicine, Jikei University School of Medicine, 3-25-8 Nishi-shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Kazuya Tsubouchi
- Division of Respiratory Diseases, Department of Internal Medicine, Jikei University School of Medicine, 3-25-8 Nishi-shimbashi, Minato-ku, Tokyo, 105-8461, Japan.,Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Nahoko Sato
- Division of Respiratory Diseases, Department of Internal Medicine, Jikei University School of Medicine, 3-25-8 Nishi-shimbashi, Minato-ku, Tokyo, 105-8461, Japan.,Department of Respiratory Medicine, Faculty of Life Science, Kumamoto University, Kumamoto, Japan
| | - Masahiro Yoshida
- Division of Respiratory Diseases, Department of Internal Medicine, Jikei University School of Medicine, 3-25-8 Nishi-shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Kenji Kobayashi
- Division of Respiratory Diseases, Department of Internal Medicine, Jikei University School of Medicine, 3-25-8 Nishi-shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Saburo Ito
- Division of Respiratory Diseases, Department of Internal Medicine, Jikei University School of Medicine, 3-25-8 Nishi-shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Yu Fujita
- Division of Respiratory Diseases, Department of Internal Medicine, Jikei University School of Medicine, 3-25-8 Nishi-shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Hirofumi Utsumi
- Division of Respiratory Diseases, Department of Internal Medicine, Jikei University School of Medicine, 3-25-8 Nishi-shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Haruhiko Yanagisawa
- Division of Respiratory Diseases, Department of Internal Medicine, Jikei University School of Medicine, 3-25-8 Nishi-shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Mitsuo Hashimoto
- Division of Respiratory Diseases, Department of Internal Medicine, Jikei University School of Medicine, 3-25-8 Nishi-shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Hiroshi Wakui
- Division of Respiratory Diseases, Department of Internal Medicine, Jikei University School of Medicine, 3-25-8 Nishi-shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Yutaka Yoshii
- Division of Respiratory Diseases, Department of Internal Medicine, Jikei University School of Medicine, 3-25-8 Nishi-shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Takeo Ishikawa
- Division of Respiratory Diseases, Department of Internal Medicine, Jikei University School of Medicine, 3-25-8 Nishi-shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Takanori Numata
- Division of Respiratory Diseases, Department of Internal Medicine, Jikei University School of Medicine, 3-25-8 Nishi-shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Yumi Kaneko
- Division of Respiratory Diseases, Department of Internal Medicine, Jikei University School of Medicine, 3-25-8 Nishi-shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Hisatoshi Asano
- Division of Chest Diseases; Department of Surgery, Jikei University School of Medicine, Tokyo, Japan
| | - Makoto Yamashita
- Division of Chest Diseases; Department of Surgery, Jikei University School of Medicine, Tokyo, Japan
| | - Makoto Odaka
- Division of Chest Diseases; Department of Surgery, Jikei University School of Medicine, Tokyo, Japan
| | - Toshiaki Morikawa
- Division of Chest Diseases; Department of Surgery, Jikei University School of Medicine, Tokyo, Japan
| | - Katsutoshi Nakayama
- Division of Respiratory Diseases, Department of Internal Medicine, Jikei University School of Medicine, 3-25-8 Nishi-shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Kazuyoshi Kuwano
- Division of Respiratory Diseases, Department of Internal Medicine, Jikei University School of Medicine, 3-25-8 Nishi-shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| |
Collapse
|
13
|
Saito Y, Azuma A, Matsuda K, Kamio K, Abe S, Gemma A. Pirfenidone exerts a suppressive effect on CCL18 expression in U937-derived macrophages partly by inhibiting STAT6 phosphorylation. Immunopharmacol Immunotoxicol 2016; 38:464-471. [PMID: 27788604 DOI: 10.1080/08923973.2016.1247852] [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] [Indexed: 10/20/2022]
Abstract
CONTEXT CC chemokine ligand 18 (CCL18) is suggested to play a role in the development of pulmonary fibrosis. Macrophages are thought to be the main source of CCL18, and the effect of pirfenidone, an anti-fibrotic agent for idiopathic pulmonary fibrosis, on the expression of CCL18 in macrophages warrants investigation. OBJECTIVE The purpose of this study was to investigate the effect of pirfenidone on the expression of CCL18 in macrophages. MATERIALS AND METHODS U937 cells were differentiated into macrophages by phorbol myristate acetate and then stimulated with recombinant IL-4 to induce the production of CCL18. The cells were treated with pirfenidone, and the mRNA and protein levels for CCL18 were measured by a reverse transcription-polymerase chain reaction and enzyme-linked immunosorbent assay, respectively. The effects of pirfenidone on the IL-4 receptor (IL-4R) expression and STAT6 activation were investigated and on the JAK kinase activity were measured using the Z'-LYTE™ kinase assay. RESULTS Pirfenidone significantly suppressed the expression of CCL18 when the cells were treated with concentrations of 50-250 μg/mL. Pirfenidone did not affect the expression of the IL-4R components. The selective STAT6 inhibitor AS1517499 suppressed CCL18 expression. Both AS1517499 and pirfenidone suppressed STAT6 phosphorylation (p < .05), although the effect of pirfenidone was less marked than that of AS1517499. The Z'-LYTE™ kinase assay showed a reduction in the activities of JAK1, JAK3 and TYK2 by pirfenidone. CONCLUSION Pirfenidone suppresses CCL18 expression in macrophages and this effect is thought to be attributed partly to the inhibition of STAT6 phosphorylation.
Collapse
Affiliation(s)
- Yoshinobu Saito
- a Department of Pulmonary Medicine and Oncology , Graduate School of Medicine, Nippon Medical School , Tokyo , Japan
| | - Arata Azuma
- a Department of Pulmonary Medicine and Oncology , Graduate School of Medicine, Nippon Medical School , Tokyo , Japan
| | - Kuniko Matsuda
- a Department of Pulmonary Medicine and Oncology , Graduate School of Medicine, Nippon Medical School , Tokyo , Japan
| | - Koichiro Kamio
- a Department of Pulmonary Medicine and Oncology , Graduate School of Medicine, Nippon Medical School , Tokyo , Japan
| | - Shinji Abe
- a Department of Pulmonary Medicine and Oncology , Graduate School of Medicine, Nippon Medical School , Tokyo , Japan
| | - Akihiko Gemma
- a Department of Pulmonary Medicine and Oncology , Graduate School of Medicine, Nippon Medical School , Tokyo , Japan
| |
Collapse
|
14
|
Li H, Yuan Y, Zhang Y, He Q, Xu R, Ge F, Wu C. Celastrol inhibits IL-1β-induced inflammation in orbital fibroblasts through the suppression of NF-κB activity. Mol Med Rep 2016; 14:2799-806. [PMID: 27484716 DOI: 10.3892/mmr.2016.5570] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 06/24/2016] [Indexed: 11/06/2022] Open
Abstract
Graves' disease is an autoimmune disease of the thyroid gland, which is characterized by hyperthyroidism, diffuse goiter and Graves' ophthalmopathy (GO). Although several therapeutic strategies for the treatment of GO have been developed, the effectiveness and the safety profile of these therapies remain to be fully elucidated. Therefore, examination of novel GO therapies remains an urgent requirement. Celastrol, a triterpenoid isolated from traditional Chinese medicine, is a promising drug for the treatment of various inflammatory and autoimmune diseases. CCK‑8 and apoptosis assays were performed to investigate cytotoxicity of celastrol and effect on apoptosis on orbital fibroblasts. Reverse transcription‑polymerase chain reaction, western blotting and ELISAs were performed to examine the effect of celastrol on interleukin (IL)‑1β‑induced inflammation in orbital fibroblasts from patients with GO. The results demonstrated that celastrol significantly attenuated the expression of IL‑6, IL‑8, cyclooxygenase (COX)‑2 and intercellular adhesion molecule‑1 (ICAM‑1), and inhibited IL‑1β‑induced increases in the expression of IL‑6, IL‑8, ICAM‑1 and COX‑2. The levels of prostaglandin E2 in orbital fibroblasts induced by IL‑1β were also suppressed by celastrol. Further investigation revealed that celastrol suppressed the IL‑1β‑induced inflammatory responses in orbital fibroblasts through inhibiting the activation of nuclear factor (NF)‑κB. Taken together, these results suggested that celastrol attenuated the IL‑1β‑induced pro‑inflammatory pathway in orbital fibroblasts from patients with GO, which was associated with the suppression of NF-κB activation.
Collapse
Affiliation(s)
- Hong Li
- Department of Endocrinology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, P.R. China
| | - Yifei Yuan
- Department of Ophthalmology, Eye Ear Nose and Throat Hospital, Fudan University, Shanghai 200031, P.R. China
| | - Yali Zhang
- Department of Endocrinology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, P.R. China
| | - Qianwen He
- Longua Medical School, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Rongjuan Xu
- Department of Endocrinology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, P.R. China
| | - Fangfang Ge
- Department of Endocrinology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, P.R. China
| | - Chen Wu
- Department of Endocrinology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, P.R. China
| |
Collapse
|
15
|
Mediavilla-Varela M, Boateng K, Noyes D, Antonia SJ. The anti-fibrotic agent pirfenidone synergizes with cisplatin in killing tumor cells and cancer-associated fibroblasts. BMC Cancer 2016; 16:176. [PMID: 26935219 PMCID: PMC4776434 DOI: 10.1186/s12885-016-2162-z] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 02/10/2016] [Indexed: 01/03/2023] Open
Abstract
Background Anti-fibrotic drugs such as pirfenidone have been developed for the treatment of idiopathic pulmonary fibrosis. Because activated fibroblasts in inflammatory conditions have similar characteristics as cancer-associated fibroblasts (CAFs) and CAFs contribute actively to the malignant phenotype, we believe that anti-fibrotic drugs have the potential to be repurposed as anti-cancer drugs. Methods The effects of pirfenidone alone and in combination with cisplatin on human patient-derived CAF cell lines and non-small cell lung cancer (NSCLC) cell lines were examined. The impact on cell death in vitro as well as tumor growth in a mouse model was determined. Annexin V/PI staining and Western blot analysis were used to characterize cell death. Synergy was assessed with the combination index method using Calcusyn software. Results Pirfenidone alone induced apoptotic cell death in lung CAFs at a high concentration (1.5 mg/mL). However, co-culture in vitro experiments and co-implantation in vivo experiments showed that the combination of low doses of cisplatin (10 μM) and low doses of pirfenidone (0.5 mg/mL), in both CAFs and tumors, lead to increased cell death and decreased tumor progression, respectively. Furthermore, the combination of cisplatin and pirfenidone in NSCLC cells (A549 and H157 cells) leads to increased apoptosis and synergistic cell death. Conclusions Our studies reveal for the first time that the combination of cisplatin and pirfenidone is active in preclinical models of NSCLC and therefore may be a new therapeutic approach in this disease. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2162-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Melanie Mediavilla-Varela
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Kingsley Boateng
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - David Noyes
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Scott J Antonia
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA. .,Thoracic Oncology Department, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA.
| |
Collapse
|
16
|
Yao Y, Chen L, Xiao J, Wang C, Jiang W, Zhang R, Hao J. Chrysin protects against focal cerebral ischemia/reperfusion injury in mice through attenuation of oxidative stress and inflammation. Int J Mol Sci 2014; 15:20913-26. [PMID: 25402649 PMCID: PMC4264203 DOI: 10.3390/ijms151120913] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 10/19/2014] [Accepted: 11/03/2014] [Indexed: 12/29/2022] Open
Abstract
Inflammation and oxidative stress play an important part in the pathogenesis of focal cerebral ischemia/reperfusion (I/R) injury, resulting in neuronal death. The signaling pathways involved and the underlying mechanisms of these events are not fully understood. Chrysin, which is a naturally occurring flavonoid, exhibits various biological activities. In this study, we investigated the neuroprotective properties of chrysin in a mouse model of middle cerebral artery occlusion (MCAO). To this end, male C57/BL6 mice were pretreated with chrysin once a day for seven days and were then subjected to 1 h of middle cerebral artery occlusion followed by reperfusion for 24 h. Our data show that chrysin successfully decreased neurological deficit scores and infarct volumes, compared with the vehicle group. The increases in glial cell numbers and proinflammatory cytokine secretion usually caused by ischemia/reperfusion were significantly ameliorated by chrysin pretreatment. Moreover, chrysin also inhibited the MCAO-induced up-regulation of nuclear factor-kappa B (NF-κB), cyclooxygenase-2 (COX-2), and inducible nitric oxide synthase (iNOS), compared with the vehicle. These results suggest that chrysin could be a potential prophylactic agent for cerebral ischemia/reperfusion (I/R) injury mediated by its anti-inflammatory and anti-oxidative effects.
Collapse
Affiliation(s)
- Yang Yao
- Department of Neurology and Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China.
| | - Li Chen
- Department of Neurology and Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China.
| | - Jinting Xiao
- Department of Neurology and Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China.
| | - Chunyang Wang
- Department of Neurology and Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China.
| | - Wei Jiang
- Department of Neurology and Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China.
| | - Rongxin Zhang
- Laboratory of Immunology and Inflammation, Research Center of Basic Medical Science, Tianjin Medical University, Tianjin 300070, China.
| | - Junwei Hao
- Department of Neurology and Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China.
| |
Collapse
|
17
|
Markers of inflammation and fibrosis in the orbital fat/connective tissue of patients with Graves' orbitopathy: clinical implications. Mediators Inflamm 2014; 2014:412158. [PMID: 25309050 PMCID: PMC4182072 DOI: 10.1155/2014/412158] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 08/06/2014] [Accepted: 08/09/2014] [Indexed: 11/18/2022] Open
Abstract
Purpose. To assess FGF-β, TGF-β, and COX2 expression and immunocompetent cells in the orbital tissue of patients with severe and mild Graves' orbitopathy. Patients and Methods. Orbital tissue was taken from 27 patients with GO: (1) severe GO (n = 18), the mean clinical activity score (CAS) being 8.5 (SD 2.5); and (2) mild GO (n = 9), the mean CAS being 2.2 (SD 0.8), and from 10 individuals undergoing blepharoplasty. The expression of CD4+, CD8+, CD20+, and CD68 and FGF-β, TGF-β, and COX2 in the orbital tissue was evaluated by immunohistochemical methods. Results. We demonstrated predominant CD4+ T cells in severe GO. CD68 expression was observed in the fibrous connective area of mild GO and was robust in severe GO, while the prominent TGF-β expression was seen in all GO. Increased FGF-β expression was observed in the fibroblasts and adipocytes of severe GO. No expression of COX2 was found in patients with GO. Conclusions. Macrophages and CD4 T lymphocytes are both engaged in the active/severe and long stage of inflammation in the orbital tissue. FGF-β and TGF-β expression may contribute to tissue remodeling, fibrosis, and perpetuation of inflammation in the orbital tissue of GO especially in severe GO.
Collapse
|
18
|
Carotenoids, inflammation, and oxidative stress--implications of cellular signaling pathways and relation to chronic disease prevention. Nutr Res 2014; 34:907-29. [PMID: 25134454 DOI: 10.1016/j.nutres.2014.07.010] [Citation(s) in RCA: 410] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 06/24/2014] [Accepted: 07/14/2014] [Indexed: 12/31/2022]
Abstract
Several epidemiologic studies have shown that diets rich in fruits and vegetables reduce the risk of developing several chronic diseases, such as type 2 diabetes, atherosclerosis, and cancer. These diseases are linked with systemic, low-grade chronic inflammation. Although controversy persists on the bioactive ingredients, several secondary plant metabolites have been associated with these beneficial health effects. Carotenoids represent the most abundant lipid-soluble phytochemicals, and in vitro and in vivo studies have suggested that they have antioxidant, antiapoptotic, and anti-inflammatory properties. Recently, many of these properties have been linked to the effect of carotenoids on intracellular signaling cascades, thereby influencing gene expression and protein translation. By blocking the translocation of nuclear factor κB to the nucleus, carotenoids are able to interact with the nuclear factor κB pathway and thus inhibit the downstream production of inflammatory cytokines, such as interleukin-8 or prostaglandin E2. Carotenoids can also block oxidative stress by interacting with the nuclear factor erythroid 2-related factor 2 pathway, enhancing its translocation into the nucleus, and activating phase II enzymes and antioxidants, such as glutathione-S-transferases. In this review, which is organized into in vitro, animal, and human investigations, we summarized current knowledge on carotenoids and metabolites with respect to their ability to modulate inflammatory and oxidative stress pathways and discuss potential dose-health relations. Although many pathways involved in the bioactivity of carotenoids have been revealed, future research should be directed toward dose-response relations of carotenoids, their metabolites, and their effect on transcription factors and metabolism.
Collapse
|
19
|
Liu Z, Duan ZJ, Chang JY, Zhang ZF, Chu R, Li YL, Dai KH, Mo GQ, Chang QY. Sinomenine sensitizes multidrug-resistant colon cancer cells (Caco-2) to doxorubicin by downregulation of MDR-1 expression. PLoS One 2014; 9:e98560. [PMID: 24901713 PMCID: PMC4047020 DOI: 10.1371/journal.pone.0098560] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Accepted: 05/05/2014] [Indexed: 12/28/2022] Open
Abstract
Chemoresistance in multidrug-resistant (MDR) cells over expressing P-glycoprotein (P-gp) encoded by the MDR1 gene, is a major obstacle to successful chemotherapy for colorectal cancer. Previous studies have indicated that sinomenine can enhance the absorption of various P-gp substrates. In the present study, we investigated the effect of sinomenine on the chemoresistance in colon cancer cells and explored the underlying mechanism. We developed multidrug-resistant Caco-2 (MDR-Caco-2) cells by exposure of Caco-2 cells to increasing concentrations of doxorubicin. We identified overexpression of COX-2 and MDR-1 genes as well as activation of the NF-κB signal pathway in MDR-Caco-2 cells. Importantly, we found that sinomenine enhances the sensitivity of MDR-Caco-2 cells towards doxorubicin by downregulating MDR-1 and COX-2 expression through inhibition of the NF-κB signaling pathway. These findings provide a new potential strategy for the reversal of P-gp-mediated anticancer drug resistance.
Collapse
Affiliation(s)
- Zhen Liu
- Department of Gastroenterology, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Zhi-Jun Duan
- Department of Gastroenterology, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- * E-mail: (ZJD); (QYC)
| | - Jiu-Yang Chang
- Department of Gastroenterology, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Zhi-feng Zhang
- Department of Gastroenterology, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Rui Chu
- Department of Gastroenterology, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Yu-Ling Li
- Department of Gastroenterology, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Ke-Hang Dai
- Department of Gastroenterology, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Guang-quan Mo
- Department of Gastroenterology, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Qing-Yong Chang
- Department of Neurosurgery, Zhongshan Affiliated Hospital of Dalian University, Dalian, Liaoning, China
- * E-mail: (ZJD); (QYC)
| |
Collapse
|