1
|
Carter H, Costa RM, Adams TS, Gilchrist TM, Emch CE, Bame M, Oldham JM, Huang SK, Linderholm AL, Noth I, Kaminski N, Moore BB, Gurczynski SJ. CD103+ dendritic cell-fibroblast crosstalk via TLR9, TDO2, and AHR signaling drives lung fibrogenesis. JCI Insight 2025; 10:e177072. [PMID: 39964756 PMCID: PMC11949071 DOI: 10.1172/jci.insight.177072] [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: 11/07/2023] [Accepted: 02/12/2025] [Indexed: 02/20/2025] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is characterized by progressive scarring and loss of lung function. With limited treatment options, patients die from the disease within 2-5 years. The molecular pathogenesis underlying the immunologic changes that occur in IPF is poorly understood. We characterize noncanonical aryl-hydrocarbon receptor (ncAHR) signaling in DCs as playing a role in the production of IL-6 and increased IL-17+ cells, promoting fibrosis. TLR9 signaling in myofibroblasts is shown to regulate production of TDO2, which converts tryptophan into the endogenous AHR ligand kynurenine. Mice with augmented ncAHR signaling were created by crossing mice harboring a floxed AHR exon 2 deletion (AHRΔex2) with mice harboring a CD11c-Cre. Bleomycin (blm) was used to study fibrotic pathogenesis. Isolated CD11c+ cells and primary fibroblasts were treated ex vivo with relevant TLR agonists and AHR-modulating compounds to study how AHR signaling influenced inflammatory cytokine production. Human datasets were also interrogated. Inhibition of all AHR signaling rescued fibrosis; however, AHRΔex2 mice treated with blm developed more fibrosis, and DCs from these mice were hyperinflammatory and profibrotic upon adoptive transfer. Treatment of fibrotic fibroblasts with TLR9 agonist increased expression of TDO2, and fibrotic fibroblasts activated IL-6 production in CD103+ DCs. Study of human samples corroborated the relevance of these findings in patients with IPF. We also show, for the first time to our knowledge, that AHR exon 2 floxed mice retain the capacity for ncAHR signaling.
Collapse
Affiliation(s)
- Hannah Carter
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
| | - Rita Medina Costa
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
| | - Taylor S. Adams
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Talon M. Gilchrist
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
| | - Claire E. Emch
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
| | - Monica Bame
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
| | - Justin M. Oldham
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Steven K. Huang
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Angela L. Linderholm
- Division of Pulmonary and Critical Care Medicine, University of California, Davis, California, USA
| | - Imre Noth
- Division of Pulmonary and Critical Care Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Naftali Kaminski
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Bethany B. Moore
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Stephen J. Gurczynski
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
2
|
Bai L, Zhu J, Ma W, Zhao P, Li F, Zhang C, Zhang S. A novel mouse model of myositis-associated interstitial lung disease was established by using TLR9 agonist combined with muscle homogenate. Clin Exp Immunol 2025; 219:uxae106. [PMID: 39575634 PMCID: PMC11773800 DOI: 10.1093/cei/uxae106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 10/13/2024] [Accepted: 11/20/2024] [Indexed: 01/29/2025] Open
Abstract
Our group previously demonstrated that NETs were involved in interstitial lung diseases (ILD) among patients with idiopathic inflammatory myopathies (IIM) and the experimental autoimmune myositis (EAM) mouse model and that NETs activated lung fibroblasts through the TLR9-miR7-Smad2 axis. This study aimed to establish a novel mouse model of myositis-associated interstitial lung disease (MAILD) by using a TLR9 agonist (ODN2395). ODN2395 and muscle homogenate were used to induce MAILD in BALB/c mice. MAILD was evaluated using histopathology, immunohistochemistry, serum NETs determination, and myositis-specific antibody profile. Furthermore, TLR9 and IRF3 were examined in a lung biopsy tissue from a dermatomyositis patient with ILD. MAILD mice developed inflammatory myopathy with positive expression of myositis-specific antibodies. ILD occurred in all mice of the MAILD group. ODN2395 at doses of 5 μg, 10 μg, or 20 μg induced ILD, with increasing severity as the dose increased, but 20 μg ODN2395 was not recommended due to non-specific damage to the lungs. ILD could occur as early as one week after immunization and was most pronounced by the fourth/fifth week. MAILD process was accompanied by NETs infiltration and TLR9 activation. TLR9 activation was demonstrated in the patient with DM-ILD. Serum levels of Cit-H3 were elevated in the MAILD group. Skeletal muscle homogenate and ODN2395 induced neutrophils to form NETs in vitro. Combined with muscle homogenate, ODN2395 induced a novel MAILD mouse model with NETs infiltration and TLR9 activation, which are similar to pathogenesis of IIM-ILD, suggesting that MAILD model could replace EAM model in IIM-ILD research.
Collapse
Affiliation(s)
- Ling Bai
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of Pediatric Cardiology, Kidney Disease and Rheumatology, Gansu Provincial Maternity and Child Care Hospital, Lanzhou, China
| | - Jiarui Zhu
- Department of Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, China
| | - Wenlan Ma
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of Rheumatology, Qinghai University Affiliated Hospital, Xining, China
| | - Peipei Zhao
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Feifei Li
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Cen Zhang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Sigong Zhang
- Department of Rheumatology, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
3
|
Manavi MA, Fathian Nasab MH, Mohammad Jafari R, Dehpour AR. Mechanisms underlying dose-limiting toxicities of conventional chemotherapeutic agents. J Chemother 2024; 36:623-653. [PMID: 38179685 DOI: 10.1080/1120009x.2023.2300217] [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: 09/20/2023] [Revised: 12/19/2023] [Accepted: 12/21/2023] [Indexed: 01/06/2024]
Abstract
Dose-limiting toxicities (DLTs) are severe adverse effects that define the maximum tolerated dose of a cancer drug. In addition to the specific mechanisms of each drug, common contributing factors include inflammation, apoptosis, ion imbalances, and tissue-specific enzyme deficiencies. Among various DLTs are bleomycin-induced pulmonary fibrosis, doxorubicin-induced cardiomyopathy, cisplatin-induced nephrotoxicity, methotrexate-induced hepatotoxicity, vincristine-induced neurotoxicity, paclitaxel-induced peripheral neuropathy, and irinotecan, which elicits severe diarrhea. Currently, specific treatments beyond dose reduction are lacking for most toxicities. Further research on cellular and molecular pathways is imperative to improve their management. This review synthesizes preclinical and clinical data on the pharmacological mechanisms underlying DLTs and explores possible treatment approaches. A comprehensive perspective reveals knowledge gaps and emphasizes the need for future studies to develop more targeted strategies for mitigating these dose-dependent adverse effects. This could allow the safer administration of fully efficacious doses to maximize patient survival.
Collapse
Affiliation(s)
- Mohammad Amin Manavi
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Razieh Mohammad Jafari
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad Reza Dehpour
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
4
|
Xu MX, Xu T, An N. Suppression of miR-17 Alleviates Acute Respiratory Distress-associated Lung Fibrosis by Regulating Mfn2. Curr Med Sci 2024; 44:964-970. [PMID: 39446286 DOI: 10.1007/s11596-024-2940-9] [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: 05/08/2024] [Accepted: 09/13/2024] [Indexed: 10/25/2024]
Abstract
OBJECTIVE Acute respiratory distress syndrome (ARDS) patients currently have relatively high mortality, which is associated with early lung fibrosis. This study aimed to investigate whether miR-17 suppression could alleviate ARDS-associated lung fibrosis by regulating Mfn2. METHODS A mouse model of ARDS-related lung fibrosis was constructed via intratracheal instillation of bleomycin. The expression level of miR-17 in lung tissues was detected via quantitative real time polymerase chain reaction (qRT-PCR). In the ARDS mouse model of lung fibrosis, the mitigating effects of miR-17 interference were evaluated via tail vein injection of the miR negative control or the miR-17 antagomir. The pathological changes in the lung tissue were examined via HE staining and Masson's trichrome staining, and the underlying molecular mechanism was investigated via ELISA, qRT-PCR and Western blotting. RESULTS Bleomycin-induced pulmonary fibrosis significantly increased collagen deposition and the levels of hydroxyproline (HYP) and miR-17. Interfering with miR-17 significantly reduced the levels of HYP and miR-17 and upregulated the expression of Mfn2. The intravenous injection of the miR-17 antagomir alleviated lung inflammation and reduced collagen deposition. In addition, interference with miR-17 could upregulate LC3B expression, downregulate p62 expression, and improve mitochondrial structure. CONCLUSION Interfering with miR-17 can improve pulmonary fibrosis in mice by promoting mitochondrial autophagy via Mfn2.
Collapse
Affiliation(s)
- Mei-Xia Xu
- Department of Critical Care Medicine, Wuhan Fourth Hospital, Wuhan, 430033, China
| | - Tao Xu
- Department of Critical Care Medicine, Wuhan Fourth Hospital, Wuhan, 430033, China.
| | - Ning An
- Institute of Anesthesiology and Critical Care, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
5
|
Trujillo G, Regueiro-Ren A, Liu C, Hu B, Sun Y, Ahangari F, Fiorini V, Ishikawa G, Al Jumaily K, Khoury J, McGovern J, Lee CJ, Peng XY, Pivarnik T, Sun H, Walia A, Woo S, Yu S, Antin-Ozerkis DE, Sauler M, Kaminski N, Herzog EL, Ryu C. Toll-like Receptor 9 Inhibition Mitigates Fibroproliferative Responses in Translational Models of Pulmonary Fibrosis. Am J Respir Crit Care Med 2024; 211:91-102. [PMID: 39189851 PMCID: PMC11755360 DOI: 10.1164/rccm.202401-0065oc] [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: 01/08/2024] [Accepted: 08/27/2024] [Indexed: 08/28/2024] Open
Abstract
RATIONALE Idiopathic pulmonary fibrosis (IPF) is a fatal lung disease for which current treatment options only slow clinical progression. Previously, we identified a subset of patients with IPF with an accelerated disease course associated with fibroblast expression of Toll-Like Receptor 9 (TLR9) mediated by interactions with its ligand mitochondrial DNA (mtDNA). OBJECTIVES We aimed to show that TLR9 activation induces fibroproliferative responses that are abrogated by its antagonism by using two commercially-available indirect inhibitors and a proprietary, selective direct small molecule inhibitor. METHODS We employed two independent cohorts of patients with IPF, multiple in vitro fibroblast cell culture platforms, an in vivo mouse model, and an ex vivo human precision cut lung slices system to investigate the clinical and biologic significance of TLR9 in this disease. MEASUREMENTS AND MAIN RESULTS In two independent IPF cohorts, plasma mtDNA activates TLR9 in a manner associated with the expression of MCP-1, IL-6, TNFα, and IP-10 and worsened transplant-free survival. Our cell culture platform showed that TLR9 mediates fibroblast activation via TGFβ1 and stiff substrates, and that its antagonism, particularly direct inhibition, ameliorates this process, including production of these TLR9 associated pharmacodynamic endpoints. We further demonstrated that direct TLR9 inhibition mitigates these fibroproliferative responses in our in vivo and ex vivo models of pulmonary fibrosis. CONCLUSIONS In this novel study, we found that direct TLR9 inhibition mitigates fibroproliferative responses in preclinical models of pulmonary fibrosis. Our work demonstrates the therapeutic potential of direct TLR9 antagonism in IPF and related fibrotic lung diseases.
Collapse
Affiliation(s)
- Glenda Trujillo
- Bristol-Myers Squibb Company, New York, New York, United States
| | | | - Chunjian Liu
- Bristol-Myers Squibb Company, New York, New York, United States
| | - Buqu Hu
- Yale School of Medicine, New Haven, Connecticut, United States
| | - Ying Sun
- Yale School of Medicine, New Haven, Connecticut, United States
| | - Farida Ahangari
- Yale School of Medicine, New Haven, Connecticut, United States
| | - Vitoria Fiorini
- Yale School of Medicine, New Haven, Connecticut, United States
| | - Genta Ishikawa
- Yale School of Medicine, New Haven, Connecticut, United States
| | | | - Johad Khoury
- Yale School of Medicine, Pulmonology Division, New Haven, Connecticut, United States
- Lady Davis Carmel Medical Center, , Haifa, Israel
| | - John McGovern
- Yale School of Medicine, New Haven, Connecticut, United States
| | - Chris J Lee
- Yale School of Medicine, New Haven, Connecticut, United States
| | - Xue Yan Peng
- Yale School of Medicine, New Haven, Connecticut, United States
| | - Taylor Pivarnik
- Yale School of Medicine, New Haven, Connecticut, United States
| | - Huanxing Sun
- Yale School of Medicine, New Haven, Connecticut, United States
| | - Anjali Walia
- Yale School of Medicine, New Haven, Connecticut, United States
| | - Samuel Woo
- Yale School of Medicine, New Haven, Connecticut, United States
| | - Sheeline Yu
- Yale School of Medicine, New Haven, Connecticut, United States
| | | | - Maor Sauler
- Yale School of Medicine, New Haven, Connecticut, United States
| | | | - Erica L Herzog
- Yale School of Medicine, New Haven, Connecticut, United States
| | - Changwan Ryu
- Yale University School of Medicine, New Haven, Connecticut, United States;
| |
Collapse
|
6
|
Carter H, Costa RM, Adams TS, Gilchrist T, Emch CE, Bame M, Oldham JM, Linderholm AL, Noth I, Kaminski N, Moore BB, Gurczynski SJ. Dendritic Cell - Fibroblast Crosstalk via TLR9 and AHR Signaling Drives Lung Fibrogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.15.584457. [PMID: 38559175 PMCID: PMC10980010 DOI: 10.1101/2024.03.15.584457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is characterized by progressive scarring and loss of lung function. With limited treatment options, patients succumb to the disease within 2-5 years. The molecular pathogenesis of IPF regarding the immunologic changes that occur is poorly understood. We characterize a role for non-canonical aryl-hydrocarbon receptor signaling (ncAHR) in dendritic cells (DCs) that leads to production of IL-6 and IL-17, promoting fibrosis. TLR9 signaling in myofibroblasts is shown to regulate production of TDO2 which converts tryptophan into the endogenous AHR ligand kynurenine. Mice with augmented ncAHR signaling were created by crossing floxed AHR exon-2 deletion mice (AHR Δex2 ) with mice harboring a CD11c-Cre. Bleomycin was used to study fibrotic pathogenesis. Isolated CD11c+ cells and primary fibroblasts were treated ex-vivo with relevant TLR agonists and AHR modulating compounds to study how AHR signaling influenced inflammatory cytokine production. Human datasets were also interrogated. Inhibition of all AHR signaling rescued fibrosis, however, AHR Δex2 mice treated with bleomycin developed more fibrosis and DCs from these mice were hyperinflammatory and profibrotic upon adoptive transfer. Treatment of fibrotic fibroblasts with TLR9 agonist increased expression of TDO2. Study of human samples corroborate the relevance of these findings in IPF patients. We also, for the first time, identify that AHR exon-2 floxed mice retain capacity for ncAHR signaling.
Collapse
|
7
|
Amirkhosravi A, Mirtajaddini Goki M, Heidari MR, Karami-Mohajeri S, Iranpour M, Torshabi M, Mehrabani M, Mandegary A, Mehrabani M. Combination of losartan with pirfenidone: a protective anti-fibrotic against pulmonary fibrosis induced by bleomycin in rats. Sci Rep 2024; 14:8729. [PMID: 38622264 PMCID: PMC11018867 DOI: 10.1038/s41598-024-59395-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 04/10/2024] [Indexed: 04/17/2024] Open
Abstract
Pirfenidone (PFD), one acceptable medication for treating idiopathic pulmonary fibrosis (IPF), is not well tolerated by patients at full doses. Hence, employing of some approaches such as combination therapy may be applicable for increasing therapeutic efficacy of PFD. Losartan (LOS), an angiotensin II receptor antagonist, could be a suitable candidate for combination therapy because of its stabilizing effect on the pulmonary function of IPF patients. Therefore, this study aimed to investigate the effects of LOS in combination with PFD on bleomycin (BLM)-induced lung fibrosis in rats. BLM-exposed rats were treated with LOS alone or in combination with PFD. The edema, pathological changes, level of transforming growth factor-β (TGF-β1), collagen content, and oxidative stress parameters were assessed in the lung tissues. Following BLM exposure, the inflammatory response, collagen levels, and antioxidant markers in rat lung tissues were significantly improved by PFD, and these effects were improved by combination with LOS. The findings of this in vivo study suggest that the combined administration of PFD and LOS may provide more potent protection against IPF than single therapy through boosting its anti-inflammatory, anti-fibrotic, and anti-oxidant effects. These results hold promise in developing a more effective therapeutic strategy for treating of lung fibrosis.
Collapse
Affiliation(s)
- Arian Amirkhosravi
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
| | | | - Mahmoud Reza Heidari
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Somayyeh Karami-Mohajeri
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Maryam Iranpour
- Department of Pathology, Pathology and Stem Cell Research Center, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Maryam Torshabi
- Department of Dental Biomaterials, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mitra Mehrabani
- Herbal and Traditional Medicines Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Ali Mandegary
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| | - Mehrnaz Mehrabani
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
8
|
Saber MM, Monir N, Awad AS, Elsherbiny ME, Zaki HF. TLR9: A friend or a foe. Life Sci 2022; 307:120874. [PMID: 35963302 DOI: 10.1016/j.lfs.2022.120874] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/05/2022] [Accepted: 08/06/2022] [Indexed: 10/15/2022]
Abstract
The innate immune system is a primary protective line in our body. It confers its protection through different pattern recognition receptors (PRRs), especially toll like receptors (TLRs). Toll like receptor 9 (TLR9) is an intracellular TLR, expressed in different immunological and non-immunological cells. Release of cellular components, such as proteins, nucleotides, and DNA confers a beneficial inflammatory response and maintains homeostasis for removing cellular debris during normal physiological conditions. However, during pathological cellular damage and stress signals, engagement between mtDNA and TLR9 acts as an alarm for starting inflammatory and autoimmune disorders. The controversial role of TLR9 in different diseases baffled scientists if it has a protective or deleterious effect after activation during insults. Targeting the immune system, especially the TLR9 needs further investigation to provide a therapeutic strategy to control inflammation and autoimmune disorders.
Collapse
Affiliation(s)
- Mona M Saber
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Giza, Egypt.
| | - Nada Monir
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ahram Canadian University, Giza, Egypt
| | - Azza S Awad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ahram Canadian University, Giza, Egypt
| | - Marwa E Elsherbiny
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ahram Canadian University, Giza, Egypt
| | - Hala F Zaki
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Giza, Egypt
| |
Collapse
|