1
|
Salama MM, Zaghloul RA, Khalil RM, El-Shishtawy MM. Anti-neoplastic activity of celastrol in experimentally-induced mammary adenocarcinoma in mice: targeting wnt/β-catenin signaling pathway. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04148-1. [PMID: 40293499 DOI: 10.1007/s00210-025-04148-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Accepted: 04/04/2025] [Indexed: 04/30/2025]
Abstract
Natural bioactive compounds with anti-neoplastic effects, such as celastrol (CLS), have attracted considerable interest in recent years. The present study aimed to investigate the effect of CLS on wnt/β-catenin signaling, and its potential combination with doxorubicin (Dox) to enhance chemotherapeutic effects. After intramuscular inoculation of Ehrlich tumor cells, tumor-bearing mice received CLS (2 mg/kg, i.p), Dox (5 mg/kg, once/week, i.p), and their combination for 21 days. Treatment with CLS showed showing antioxidant and anti-inflammatory, as evidenced by a significant increase in glutathione content and a significant decrease in the malondialdehyde, interleukin 6, and interleukin 1β concentrations. CLS also inhibited VEGF-mediated angiogenesis. The current study revealed that CLS downregulated β-catenin gene expression with subsequent downstream target genes, such as cyclin-D1, and survivin, which dampens tumor cell proliferation and triggers cell cycle arrest as well as induces apoptosis as indicated by the increased expression of p53, caspase-3. The current study concludes that CLS exerted its anti-neoplastic activity by suppressing the wnt/β-catenin signaling pathway, and opens a new perspective for combining CLS with Dox to enhance its chemotherapeutic effects and reduce the oxidative imbalance and inflammatory responses associated with Dox treatment.
Collapse
Affiliation(s)
- Mohamed M Salama
- Department of Biochemistry, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, 35712, Egypt.
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt.
| | - Randa A Zaghloul
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| | - Rania M Khalil
- Department of Biochemistry, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, 35712, Egypt
| | - Mamdouh M El-Shishtawy
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| |
Collapse
|
2
|
Silva LS, Cavallini E, da Silva RA, Sant’Ana M, Yoshikawa AH, Salomão T, Huang B, Craice P, de Souza Ferreira LP, Della Matta HP, Gil CD, Pereira MDLG, Girol AP. Garcinia brasiliensis Leaves Extracts Inhibit the Development of Ascitic and Solid Ehrlich Tumors. Pharmaceuticals (Basel) 2024; 18:24. [PMID: 39861087 PMCID: PMC11768557 DOI: 10.3390/ph18010024] [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: 10/26/2024] [Revised: 12/18/2024] [Accepted: 12/20/2024] [Indexed: 01/27/2025] Open
Abstract
Background:Garcinia brasiliensis is traditionally known for its medicinal properties. Objectives: Here, we investigated the effects of crude extract (CE) and ethyl acetate fraction (EAF) obtained from G. brasiliensis leaves on the ascitic (EA) and solid (ES) forms of Ehrlich tumors. Methods: Induced and uninduced BALB/c mice were treated intramuscularly, for 7 or 14 days, with saline solution or CE and EAF, both at a 10% concentration, based on in vitro cytotoxicity assessment. Biochemical analyses were also performed to evaluate in vivo cytotoxicity. In relation to tumor-induced animals, morphological changes, plasma enzymes, inflammatory mediators and the induction of apoptosis were analyzed, in addition to histopathological studies, to evaluate the inhibition of tumor growth. Results: Alanine aminotransferase (ALT), aspartate aminotransferase (AST) and gamma glutamyl transferase (GGT) were regulated by CE and EAF administration. Furthermore, both treatments were effective in inhibiting tumor growth in EA and ES by modulating the levels of interleukin (IL)-6 and tumor necrosis factor (TNF)-α, decreasing mast cells numbers and inducing apoptosis. Conclusions: This research indicates that both CE and EAF from G. brasiliensis leaves have potential antitumor effects with low cytotoxicity.
Collapse
Affiliation(s)
- Lucas Sylvestre Silva
- Post Graduate Program in Structural and Functional Biology, Paulista School of Medicine (UNIFESP-EPM), Federal University of São Paulo, São Paulo 04023-062, SP, Brazil; (L.S.S.); (E.C.); (M.S.); (L.P.d.S.F.); (C.D.G.)
| | - Eduardo Cavallini
- Post Graduate Program in Structural and Functional Biology, Paulista School of Medicine (UNIFESP-EPM), Federal University of São Paulo, São Paulo 04023-062, SP, Brazil; (L.S.S.); (E.C.); (M.S.); (L.P.d.S.F.); (C.D.G.)
| | - Rafael André da Silva
- Department of Biology, Institute of Biosciences, Humanities and Exact Sciences (Ibilce), São Paulo State University (UNESP), São José do Rio Preto 15054-000, SP, Brazil;
| | - Monielle Sant’Ana
- Post Graduate Program in Structural and Functional Biology, Paulista School of Medicine (UNIFESP-EPM), Federal University of São Paulo, São Paulo 04023-062, SP, Brazil; (L.S.S.); (E.C.); (M.S.); (L.P.d.S.F.); (C.D.G.)
| | - Ariane Harumi Yoshikawa
- Experimental and Clinical Research Center (CEPEC), Padre Albino University Center (UNIFIPA), Catanduva 15809-144, SP, Brazil; (A.H.Y.); (T.S.); (B.H.); (P.C.); (H.P.D.M.)
| | - Thiago Salomão
- Experimental and Clinical Research Center (CEPEC), Padre Albino University Center (UNIFIPA), Catanduva 15809-144, SP, Brazil; (A.H.Y.); (T.S.); (B.H.); (P.C.); (H.P.D.M.)
| | - Bianca Huang
- Experimental and Clinical Research Center (CEPEC), Padre Albino University Center (UNIFIPA), Catanduva 15809-144, SP, Brazil; (A.H.Y.); (T.S.); (B.H.); (P.C.); (H.P.D.M.)
| | - Paula Craice
- Experimental and Clinical Research Center (CEPEC), Padre Albino University Center (UNIFIPA), Catanduva 15809-144, SP, Brazil; (A.H.Y.); (T.S.); (B.H.); (P.C.); (H.P.D.M.)
| | - Luiz Philipe de Souza Ferreira
- Post Graduate Program in Structural and Functional Biology, Paulista School of Medicine (UNIFESP-EPM), Federal University of São Paulo, São Paulo 04023-062, SP, Brazil; (L.S.S.); (E.C.); (M.S.); (L.P.d.S.F.); (C.D.G.)
| | - Heitor Pedro Della Matta
- Experimental and Clinical Research Center (CEPEC), Padre Albino University Center (UNIFIPA), Catanduva 15809-144, SP, Brazil; (A.H.Y.); (T.S.); (B.H.); (P.C.); (H.P.D.M.)
| | - Cristiane Damas Gil
- Post Graduate Program in Structural and Functional Biology, Paulista School of Medicine (UNIFESP-EPM), Federal University of São Paulo, São Paulo 04023-062, SP, Brazil; (L.S.S.); (E.C.); (M.S.); (L.P.d.S.F.); (C.D.G.)
- Department of Biology, Institute of Biosciences, Humanities and Exact Sciences (Ibilce), São Paulo State University (UNESP), São José do Rio Preto 15054-000, SP, Brazil;
| | | | - Ana Paula Girol
- Post Graduate Program in Structural and Functional Biology, Paulista School of Medicine (UNIFESP-EPM), Federal University of São Paulo, São Paulo 04023-062, SP, Brazil; (L.S.S.); (E.C.); (M.S.); (L.P.d.S.F.); (C.D.G.)
- Department of Biology, Institute of Biosciences, Humanities and Exact Sciences (Ibilce), São Paulo State University (UNESP), São José do Rio Preto 15054-000, SP, Brazil;
- Experimental and Clinical Research Center (CEPEC), Padre Albino University Center (UNIFIPA), Catanduva 15809-144, SP, Brazil; (A.H.Y.); (T.S.); (B.H.); (P.C.); (H.P.D.M.)
| |
Collapse
|
3
|
Sant’Ana PGDS, Lima WG, Lopes GFM, de Oliveira SE, da Costa GAF, Lima LARDS, Ferreira EE, dos Santos IC, Damázio LCM, Ribeiro RIMA, Pinto FCH. Ocimum basilicum L. (basil) presents pro-apoptotic activity in an Ehrlich's experimental tumor murine model. Acta Cir Bras 2024; 39:e393924. [PMID: 39082586 PMCID: PMC11285817 DOI: 10.1590/acb393924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 05/22/2024] [Indexed: 08/03/2024] Open
Abstract
PURPOSE This study aimed to evaluate the therapeutic effect of an ethanol extract of Ocimum basilicum L. (EEOb) aerial parts against Ehrlich's experimental tumor (EET) in mice. METHODS Swiss mice were divided into two groups (control and treated; n = 6). On day 21, all mice were inoculated subcutaneously with 2 × 106 (0.05 mL) EET cells in the left paw for solid tumor development. This study lasted 28 days. Treatment began 24 hours after inoculation with EET. Measurements of dorsoplantar thickness were used to assess tumor growth. The paw pad was collected for histopathological analysis and stained using the argyrophilic nucleolar organizing regions (AgNOR) technique and immunohistochemistry for proliferating cell nuclear antigen, Bcl-2 and Bax. RESULTS The treatment of animals with EEOb at 100 mg/kg intraperitoneally was able to reduce the growth (Control = 3.7 ± 0.1 mm vs. EEOb = 5.7 ± 0.2 mm) and the number of AgNORs of solid Ehrlich tumor. The antitumor effect of EEOb was associated with the induction of apoptosis of tumoral cell, as suggested by the reduction of the content of Bcl-2 induced by extract. CONCLUSIONS The study demonstrated that daily administration of EEOb is able to reduce the growth of EET by induce apoptosis of tumoral cells.
Collapse
Affiliation(s)
| | - William Gustavo Lima
- Universidade Federal de Minas Gerais – Faculdade de Farmácia – Belo Horizonte (MG) – Brazil
| | - Gabriela Francine Martins Lopes
- Universidade Federal de São João del-Rei – Departamento de Medicina – Laboratório de Patologia Experimental – Divinópolis (MG) – Brazil
| | - Sabrina Elisa de Oliveira
- Universidade Federal de São João del-Rei – Departamento de Ciências Naturais – Laboratório de Patologia Experimental – São João del-Rei (MG) – Brazil
| | | | | | - Elisângela Elduina Ferreira
- Universidade Federal de São João del-Rei – Departamento de Ciências Naturais – Laboratório de Patologia Experimental – São João del-Rei (MG) – Brazil
| | - Ivan Carlos dos Santos
- Universidade Federal de São João del-Rei – Departamento de Engenharia de Biossistemas – Laboratório de Biomoléculas e Biofármacos – São João del-Rei (MG) – Brazil
| | - Laila Cristina Moreira Damázio
- Universidade Federal de São João del-Rei – Departamento de Medicina – Laboratório de Reabilitação – São João del-Rei (MG) – Brazil
| | - Rosy Iara Maciel Azambuja Ribeiro
- Universidade Federal de São João del-Rei – Departamento de Medicina – Laboratório de Patologia Experimental – Divinópolis (MG) – Brazil
| | - Flávia Carmo Horta Pinto
- Universidade Federal de São João del-Rei – Departamento de Ciências Naturais – Laboratório de Patologia Experimental – São João del-Rei (MG) – Brazil
| |
Collapse
|
4
|
Eltahir Z, Ibrahim M, Mohieldeen MY, Bayoumi A, Ahmed SM. Thymoquinone Nanoparticles (TQ-NPs) in Kidney Toxicity Induced by Ehrlich Ascites Carcinoma (EAC): An In Vivo Study. Can J Kidney Health Dis 2024; 11:20543581241258812. [PMID: 38863503 PMCID: PMC11165950 DOI: 10.1177/20543581241258812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 05/06/2024] [Indexed: 06/13/2024] Open
Abstract
Background Cisplatin (Cis) is potent chemotherapy used to treating already many different types of cancer; however, it is found to correlate with nephrotoxicity and other adverse health consequences. Thymoquinone (TQ) is an antioxidant and anti-inflammatory molecule that may defend against the consequences of different chemotherapies. Thymoquinone uses, although, are negatively impacted by its weak solubility and inadequate biological availability. Objectives This investigation examined the efficacy of a new nanoparticle (NP) absorbing TQ in an Ehrlich Ascites Carcinoma (EAC) mice model to address its low solubility, enhance its bioavailability, and protect against Cis-induced nephrotoxicity. Methods Following 4 treatment groups were included in this research: (1) control, (2) EAC, (3) EAC + Cis + Thymoquinone nanoparticle (TQ-NP) treated, and (4) EAC + Cis-treated. Results The study revealed that TQ-NP was efficacious in avoiding Cis-induced kidney problems in EAC mice, as well as restoring kidney function and pathology. Thymoquinone nanoparticle considerably reduced Cis-induced oxidative damage in renal tissue by augmenting antioxidant levels. According to tumor weight and histological investigation results, TQ-NP did not impair Cis's anticancer efficacy. Conclusion Thymoquinone nanoparticle might be used as a potential drug along with Cis anticancer therapy to reduce nephrotoxicity and other side effects while maintaining Cis anticancer properties.
Collapse
Affiliation(s)
- Zakaria Eltahir
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Science, Taibah University, Medina, Saudi Arabia
- Research Unit, School of Medicine, Taibah University, Medina, Saudi Arabia
| | - Maha Ibrahim
- MSF Medical Scientific Foundation for Research & Development, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Muniera Y. Mohieldeen
- Department of Biochemistry, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Ammar Bayoumi
- MSF Medical Scientific Foundation for Research & Development, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Samia M. Ahmed
- Clinical Biochemistry, Department of Clinical Laboratory Sciences, Faculty of Applied Medical Science, Taibah University, Medina, Saudi Arabia
| |
Collapse
|
5
|
Mohamad EA, Ali AA, Sharaky M, El-Gebaly RH. Niosomes loading N-acetyl-L-cysteine for cancer treatment in vivo study. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:4339-4353. [PMID: 38091079 DOI: 10.1007/s00210-023-02893-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 12/05/2023] [Indexed: 05/23/2024]
Abstract
Scientists are seeking to find an effective treatment for tumors that has no side effects. N-Acetyl-l-cysteine (NAC) is a thiol compound extracted from garlic. Current study explores the potential of NAC-loaded niosomes (NAC-NIO) for tumor treatment in mice. NAC-loaded niosomes' efficiency, morphology, UV absorption, size distribution, zeta potential, release, and FTIR analysis were evaluated. For vivo study, 25 male BALB/c mice were divided to five groups: gp1 negative control (receive saline), gp2 positive control (tumor group), gp3 treated with NAC, gp4 treated with NAC-NIO at the same time of tumor injection, and gp5 treated with NAC-NIO after tumor growth (day 14). The impact of NAC-NIO on the tumor treatment was evaluated by measuring tumor size progress, comet assay, oxidative stress parameters (GSH, nitric oxide, MDA), western blot analysis, and histopathological investigation of tissues. NAC-NIO showed 72 ± 3% encapsulation efficiency and zeta potential - 5.95 mV with spherical shape. It was found that oral administration of NAC-NIO in a dose of 50 mg/kg provided significant protection against tumor cells. Our formulation decreases DNA injury significantly (P < 0.05). It was noticed that NAC-NIO can increase oxidative stress levels in tumor tissue. On the other hand, the caspase 3 and caspase 9 gene expression were upregulated significantly (P < 0.001) in mice administrated NAC-NIO compared with all other groups. Histological studies confirmed the protective effect of NAC-NIO against tumor especially for treatment during tumor growth protocol. The results suggested that oral delivery of NAC-NIO formulation improved antioxidant effect.
Collapse
Affiliation(s)
- Ebtesam A Mohamad
- Department of Biophysics, Faculty of Science, Cairo University, 12613, Giza, Egypt
- College of Applied Medical Sciences, Prince Sattam Bin Abdul-Aziz University, Al-Kharj, 11942, Kingdom of Saudi Arabia
| | - Abeer A Ali
- Department of Biophysics, Faculty of Science, Cairo University, 12613, Giza, Egypt.
| | - Marwa Sharaky
- Department of Cancer Biology, Pharmacology Unit, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Reem H El-Gebaly
- Department of Biophysics, Faculty of Science, Cairo University, 12613, Giza, Egypt
| |
Collapse
|
6
|
Alves MG, Cabral LGS, Totti PGF, Azarias FR, Pomini KT, Rici REG, Laiso RAN, Maria DA. 2-Aminoethyl Dihydrogen Phosphate (2-AEH2P) Associated with Cell Metabolism-Modulating Drugs Presents a Synergistic and Pro-Apoptotic Effect in an In Vitro Model of the Ascitic Ehrlich Tumor. Biomedicines 2024; 12:109. [PMID: 38255214 PMCID: PMC10813795 DOI: 10.3390/biomedicines12010109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 12/11/2023] [Accepted: 12/14/2023] [Indexed: 01/24/2024] Open
Abstract
The progression and maintenance of cancer characteristics are associated with cellular components linked to the tumor and non-cellular components with pro-tumoral properties. Pharmacological association with antagonists of the cellular components of the tumor, such as anti- and pro-apoptotic drugs, represents a novel adjuvant strategy. In this study, the antiproliferative, pro-apoptotic, and pharmacological effects of the combination of monophosphoester 2-AEH2P with Simvastatin, Coenzyme Q10, the chemotherapeutic drug paclitaxel, and colony-stimulating factor (GM-CSF) were evaluated. Tests were conducted to determine cytotoxic activity using the MTT method, cell cycle phases, and fragmented DNA by flow cytometry, mitochondrial membrane potential, expression of cell markers Bcl2, TNF-α/DR-4, Cytochrome c, caspase 3, and P53, and analysis of drug combination profiles using Synergy Finder 2.0 Software. The results showed a synergistic effect among the combinations, compared to individual treatments with the monophosphoester and other drugs. In addition, there was modulation of marker expression, indicating a pro-apoptotic and immunomodulatory effect of 2-AEH2P. Pharmacological analysis revealed that tumor cells treated with GM-CSF + 2-AEH2P exhibited a synergistic effect, while groups of tumor cells treated with paclitaxel, Coenzyme Q10, and Simvastatin showed additive effects. Furthermore, treatment with the paclitaxel + 2-AEH2P combination (12 h) resulted in a significant reduction in mitochondrial membrane potential. Pharmacological combinations for normal cells did not exhibit deleterious effects compared to mammary carcinomatosis tumor (EAT) cells.
Collapse
Affiliation(s)
- Monique G. Alves
- Development and Innovation Laboratory, Butantan Institute, São Paulo 05359-900, Brazil; (L.G.S.C.); (F.R.A.)
- Graduate Program in Medical Sciences, College of Medicine, University of São Paulo, São Paulo 05508-220, Brazil
| | - Laertty G. S. Cabral
- Development and Innovation Laboratory, Butantan Institute, São Paulo 05359-900, Brazil; (L.G.S.C.); (F.R.A.)
- Graduate Program in Medical Sciences, College of Medicine, University of São Paulo, São Paulo 05508-220, Brazil
| | - Paulo G. F. Totti
- Graduate Program in Structural and Functional Interactions in Rehabilitation, Postgraduate Department, University of Marília (UNIMAR), Marília 17525-902, Brazil (R.E.G.R.)
| | - Felipe R. Azarias
- Development and Innovation Laboratory, Butantan Institute, São Paulo 05359-900, Brazil; (L.G.S.C.); (F.R.A.)
| | - Karine T. Pomini
- Graduate Program in Structural and Functional Interactions in Rehabilitation, Postgraduate Department, University of Marília (UNIMAR), Marília 17525-902, Brazil (R.E.G.R.)
| | - Rose E. G. Rici
- Graduate Program in Structural and Functional Interactions in Rehabilitation, Postgraduate Department, University of Marília (UNIMAR), Marília 17525-902, Brazil (R.E.G.R.)
- Graduate Program in Anatomy of Domestic and Wild Animals, College of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo 05508-220, Brazil
| | - Rosa A. N. Laiso
- Development and Innovation Laboratory, Butantan Institute, São Paulo 05359-900, Brazil; (L.G.S.C.); (F.R.A.)
| | - Durvanei A. Maria
- Development and Innovation Laboratory, Butantan Institute, São Paulo 05359-900, Brazil; (L.G.S.C.); (F.R.A.)
- Graduate Program in Medical Sciences, College of Medicine, University of São Paulo, São Paulo 05508-220, Brazil
| |
Collapse
|
7
|
Sarhan MO, Haffez H, Elsayed NA, El-Haggar RS, Zaghary WA. New phenothiazine conjugates as apoptosis inducing agents: Design, synthesis, In-vitro anti-cancer screening and 131I-radiolabeling for in-vivo evaluation. Bioorg Chem 2023; 141:106924. [PMID: 37871390 DOI: 10.1016/j.bioorg.2023.106924] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 10/06/2023] [Accepted: 10/16/2023] [Indexed: 10/25/2023]
Abstract
Phenothiazines (PTZs) are a group of compounds characterized by the presence of the 10H-dibenzo-[b,e]-1,4-thiazine system. PTZs used in clinics as antipsychotic drugs with other diverse biological activities. The current aim of the study is to investigate and understand the effect of potent PTZs compounds using a group of In-vitro and In-vivo assays. A total of seventeen novel phenothiazine derivatives have been designed, synthesized, and evaluated primarily in-vitro for their ability to inhibit proliferation activity against NCI-60 cancer cell lines, including several multi-drug resistant (MDR) tumor cell lines. Almost all compounds were active and displayed promising cellular activities with GI50 values in the sub-micromolar range. Four of the most promising derivatives (4b, 4h, 4g and 6e) have been further tested against two selected sensitive cancer cell lines (colon cancer; HCT-116 and breast cancer; MDA-MB231). The apoptosis assay showed that all the selected compounds were able to induce early apoptosis and compound 6e was able to induce additional cellular necrosis. Cell cycle assay showed all selected compounds were able to induce cell cycle arrest at sub-molecular phase of G0-G1 with compound 6e induced cell cycle arrest at G2M in HCT-116 cells. Accordingly, the apoptotic effect of the selected compounds was extensively investigated on genetic level and Casp-3, Casp-9 and Bax gene were up-regulated with down-regulation of Bcl-2 gene suggesting the activation of both intrinsic and extrinsic pathways. In-vivo evaluation of the antitumor activity of compound 4b in solid tumor bearing mice showed promising therapeutic effect with manifestation of dose and time dependent toxic effects at higher doses. For better evaluation of the degree of localization of 4b, its 131I-congener (131I-4b) was injected intravenously in Ehrlich solid tumor bearing mice that showed good localization at tumor site with rapid distribution and clearance from the blood. In-silico study suggested NADPH oxidases (NOXs) as potential molecular target. The compounds introduced in the current study work provided a cutting-edge phenothiazine hybrid scaffold with promising anti-proliferation action that may suggest their anti-cancer activity.
Collapse
Affiliation(s)
- Mona O Sarhan
- Labelled Compounds Department, Hot Lab Centre, Egyptian Atomic Energy Authority, Egypt
| | - Hesham Haffez
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Helwan University, 11795 Cairo, Egypt; Center of Scientific Excellence "Helwan Structural Biology Research, (HSBR)", Helwan University, 11795 Cairo, Egypt.
| | - Nosaiba A Elsayed
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Helwan University, 11795 Cairo, Egypt
| | - Radwan S El-Haggar
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Helwan University, 11795 Cairo, Egypt
| | - Wafaa A Zaghary
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Helwan University, 11795 Cairo, Egypt.
| |
Collapse
|
8
|
Viana Barbosa LG, Silva de Jesus EN, Botelho Jerônimo L, Silva da Costa J, Cunha Silva R, Setzer WN, R da Silva JK, da Silva Freitas JJ, B Figueiredo PL. Siparuna guianensis Essential Oil Antitumoral Activity on Ehrlich Model and Its Effect on Oxidative Stress. Chem Biodivers 2023; 20:e202301120. [PMID: 37691004 DOI: 10.1002/cbdv.202301120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 09/08/2023] [Accepted: 09/10/2023] [Indexed: 09/12/2023]
Abstract
This work aims to evaluate the chemical composition, in vitro antioxidant capacity, and in vivo antitumoral activity of S. guianensis essential oil against Ehrlich's ascitic carcinoma and the effects on oxidative stress. The animals (Mus musculus) received a daily dose of S. guianensis oil orally (100 mg/kg) for 9 days. The main constituents of essential oil were curzerenone (16.4±1.5 %), drimenol (13.7±0.2 %), and spathulenol (12.4±0.8 %). S. guianensis oil showed antioxidant activity, inhibiting 11.1 % of DPPH radicals (95.7 mgTE/g); and 15.5 % of the β-carotene peroxidation. The group treated with S. guianensis showed a significant reduction in tumor cells (59.76±12.33) compared to the tumor group (96.88±19.15). Essential oil of S. guianensis decreased MDA levels and increased SOD levels in liver tissue. The essential oil of S. guianensis reduced oxidative stress, and showed antitumor and antioxidant activity, being characterized as a new chemical profile in the investigation of pathologies such as cancer.
Collapse
Affiliation(s)
- Lucas Gabriel Viana Barbosa
- Laboratório de Química dos Produtos Naturais, Universidade do Estado do Pará, Belém, 66087-662, Brazil
- Laboratório de Morfofisiologia Aplicada a Saúde, Departamento de Morfologia e Ciências Fisiológicas, Universidade do Estado do Pará, Belém, 66087-662, Brazil
| | - Ellen Nayara Silva de Jesus
- Laboratório de Química dos Produtos Naturais, Universidade do Estado do Pará, Belém, 66087-662, Brazil
- Laboratório de Morfofisiologia Aplicada a Saúde, Departamento de Morfologia e Ciências Fisiológicas, Universidade do Estado do Pará, Belém, 66087-662, Brazil
| | - Lucas Botelho Jerônimo
- Laboratório de Química dos Produtos Naturais, Universidade do Estado do Pará, Belém, 66087-662, Brazil
| | - Jamile Silva da Costa
- Laboratório de Química dos Produtos Naturais, Universidade do Estado do Pará, Belém, 66087-662, Brazil
| | - Renata Cunha Silva
- Laboratório de Morfofisiologia Aplicada a Saúde, Departamento de Morfologia e Ciências Fisiológicas, Universidade do Estado do Pará, Belém, 66087-662, Brazil
| | - William N Setzer
- Aromatic Plant Research Center, 230 N 1200 E, Suite 100, Lehi, UT, 84043, USA
| | - Joyce Kelly R da Silva
- Aromatic Plant Research Center, 230 N 1200 E, Suite 100, Lehi, UT, 84043, USA
- Programa de Pós-Graduação em Biotecnologia, Universidade Federal do Pará, Belém, 66075-900, Brazil
| | - Jofre Jacob da Silva Freitas
- Laboratório de Morfofisiologia Aplicada a Saúde, Departamento de Morfologia e Ciências Fisiológicas, Universidade do Estado do Pará, Belém, 66087-662, Brazil
| | - Pablo Luis B Figueiredo
- Laboratório de Química dos Produtos Naturais, Universidade do Estado do Pará, Belém, 66087-662, Brazil
- Programa de Pós-graduação em Ciências Farmacêuticas, Instituto de Ciências da Saúde, Universidade Federal do Pará, Belem, 66075-110, Brazil
| |
Collapse
|
9
|
Ghambashidze K, Chikhladze R, Saladze T, Hoopes PJ, Shubitidze F. E. coli Phagelysate: A Primer to Enhance Nanoparticles and Drug Deliveries in Tumor. Cancers (Basel) 2023; 15:cancers15082315. [PMID: 37190243 DOI: 10.3390/cancers15082315] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/05/2023] [Accepted: 04/12/2023] [Indexed: 05/17/2023] Open
Abstract
The tumor microenvironment (TME), where cancer cells reside, plays a crucial role in cancer progression and metastasis. It maintains an immunosuppressive state in many tumors and regulates the differentiation of precursor monocytes into M1 (anti-tumor)- and M2 (pro-tumor)-polarized macrophages, and greatly reduces anticancer drug and nanoparticle delivery. As a result, the effectiveness of recently developed chemo- and/or nanotechnology-mediated immune and magnetic nanoparticle hyperthermia (mNPH) therapies is inhibited significantly. One of the ways to overcome this limitation is to use E. coli phagelysate as a primer to modify the tumor microenvironment by switching tumor-associated M2 macrophages to anti-tumor M1 macrophages, and initiate the infiltration of tumor-associated macrophages (TAMs). Recently, bacteriophages and phage-induced lysed bacteria (bacterial phagelysates-BPLs) have been shown to be capable of modifying the tumor-associated environment. Phage/BPL-coated proteins tend to elicit strong anti-tumor responses from the innate immune system, prompting phagocytosis and cytokine release. It has also been reported that the microenvironments of bacteriophage- and BPL-treated tumors facilitate the conversion of M2-polarized TAMS to a more M1-polarized (tumoricidal) environment post-phage treatment. This paper demonstrates the feasibility and enhanced efficacy of combining E. coli phagelysate (EcPHL) and mNPH, a promising technology for treating cancers, in a rodent model. Specifically, we illustrate the EcPHL vaccination effect on the TME and mNP distribution in Ehrlich adenocarcinoma tumors by providing the tumor growth dynamics and histology (H&E and Prussian blue) distribution of mNP in tumor and normal tissue.
Collapse
Affiliation(s)
- Ketevan Ghambashidze
- Department of Pathophysiology, Tbilisi State Medical University, Tbilisi 0177, Georgia
| | - Ramaz Chikhladze
- Department of Anatomic Pathology, Tbilisi State Medical University, Tbilisi 0177, Georgia
| | - Tamar Saladze
- Department of Anatomic Pathology, Tbilisi State Medical University, Tbilisi 0177, Georgia
| | - P Jack Hoopes
- Geisel School of Medicine at Dartmouth College, Hanover, NH 03755, USA
- Thayer School of Engineering at Dartmouth College, Hanover, NH 03755, USA
| | - Fridon Shubitidze
- Geisel School of Medicine at Dartmouth College, Hanover, NH 03755, USA
| |
Collapse
|
10
|
α-Hederin Saponin Augments the Chemopreventive Effect of Cisplatin against Ehrlich Tumors and Bioinformatic Approach Identifying the Role of SDF1/CXCR4/p-AKT-1/NFκB Signaling. Pharmaceuticals (Basel) 2023; 16:ph16030405. [PMID: 36986504 PMCID: PMC10056433 DOI: 10.3390/ph16030405] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/21/2023] [Accepted: 02/22/2023] [Indexed: 03/10/2023] Open
Abstract
Stromal cell-derived factor-1 (SDF1) and its C-X-C chemokine receptor type 4 receptor (CXCR4) are significant mediators for cancer cells’ proliferation, and we studied their expression in Ehrlich solid tumors (ESTs) grown in mice. α-Hederin is a pentacyclic triterpenoid saponin found in Hedera or Nigella species with biological activity that involves suppression of growth of breast cancer cell lines. The aim of this study was to explore the chemopreventive activity of α-hederin with/without cisplatin; this was achieved by measuring the reduction in tumor masses and the downregulation in SDF1/CXCR4/pAKT signaling proteins and nuclear factor kappa B (NFκB). Ehrlich carcinoma cells were injected in four groups of Swiss albino female mice (Group1: EST control group, Group2: EST + α-hederin group, Group3: EST + cisplatin group, and Group4: EST+α-hederin/cisplatin treated group). Tumors were dissected and weighed, one EST was processed for histopathological staining with hematoxylin and eosin (HE), and the second MC was frozen and processed for estimation of signaling proteins. Computational analysis for these target proteins interactions showed direct-ordered interactions. The dissected solid tumors revealed decreases in tumor masses (~21%) and diminished viable tumor regions with significant necrotic surrounds, particularly with the combination regimens. Immunohistochemistry showed reductions (~50%) in intratumoral NFκβ in the mouse group that received the combination therapy. The combination treatment lowered the SDF1/CXCR4/p-AKT proteins in ESTs compared to the control. In conclusion, α-hederin augmented the chemotherapeutic potential of cisplatin against ESTs; this effect was at least partly mediated through suppressing the chemokine SDF1/CXCR4/p-AKT/NFκB signaling. Further studies are recommended to verify the chemotherapeutic potential of α-hederin in other breast cancer models.
Collapse
|
11
|
Mangueira VM, de Sousa TKG, Batista TM, de Abrantes RA, Moura APG, Ferreira RC, de Almeida RN, Braga RM, Leite FC, Medeiros KCDP, Cavalcanti MAT, Moura RO, Silvestre GFG, Batista LM, Sobral MV. A 9-aminoacridine derivative induces growth inhibition of Ehrlich ascites carcinoma cells and antinociceptive effect in mice. Front Pharmacol 2022; 13:963736. [PMID: 36324671 PMCID: PMC9618857 DOI: 10.3389/fphar.2022.963736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 09/29/2022] [Indexed: 11/16/2022] Open
Abstract
Acridine derivatives have been found with anticancer and antinociceptive activities. Herein, we aimed to evaluate the toxicological, antitumor, and antinociceptive actions of N’-(6-chloro-2-methoxyacridin-9-yl)-2-cyanoacetohydrazide (ACS-AZ), a 9-aminoacridine derivative with antimalarial activity. The toxicity was assessed by acute toxicity and micronucleus tests in mice. The in vivo antitumor effect of ACS-AZ (12.5, 25, or 50 mg/kg, intraperitoneally, i.p.) was determined using the Ehrlich tumor model, and toxicity. The antinociceptive efficacy of the compound (50 mg/kg, i.p.) was investigated using formalin and hot plate assays in mice. The role of the opioid system was also investigated. In the acute toxicity test, the LD50 (lethal dose 50%) value was 500 mg/kg (i.p.), and no detectable genotoxic effect was observed. After a 7-day treatment, ACS-AZ significantly (p < 0.05) reduced tumor cell viability and peritumoral microvessels density, suggesting antiangiogenic action. In addition, ACS-AZ reduced (p < 0.05) IL-1β and CCL-2 levels, which may be related to the antiangiogenic effect, while increasing (p < 0.05) TNF-α and IL-4 levels, which are related to its direct cytotoxicity. ACS-AZ also decreased (p < 0.05) oxidative stress and nitric oxide (NO) levels, both of which are crucial mediators in cancer known for their angiogenic action. Moreover, weak toxicological effects were recorded after a 7-day treatment (biochemical, hematological, and histological parameters). Concerning antinociceptive activity, ACS-AZ was effective on hotplate and formalin (early and late phases) tests (p < 0.05), characteristic of analgesic agents with central action. Through pretreatment with the non-selective (naloxone) and μ1-selective (naloxonazine) opioid antagonists, we observed that the antinociceptive effect of ACS-AZ is mediated mainly by μ1-opioid receptors (p < 0.05). In conclusion, ACS-AZ has low toxicity and antitumoral activity related to cytotoxic and antiangiogenic actions that involve the modulation of reactive oxygen species, NO, and cytokine levels, in addition to antinociceptive properties involving the opioid system.
Collapse
Affiliation(s)
- Vivianne M. Mangueira
- Post Graduation Program in Bioactive Natural and Synthetic Products, Federal University of Paraíba, João Pessoa, Brazil
| | - Tatyanna K. G. de Sousa
- Post Graduation Program in Bioactive Natural and Synthetic Products, Federal University of Paraíba, João Pessoa, Brazil
| | - Tatianne M. Batista
- Post Graduation Program in Bioactive Natural and Synthetic Products, Federal University of Paraíba, João Pessoa, Brazil
| | - Renata A. de Abrantes
- Post Graduation Program in Bioactive Natural and Synthetic Products, Federal University of Paraíba, João Pessoa, Brazil
| | - Ana Paula G. Moura
- Post Graduation Program in Bioactive Natural and Synthetic Products, Federal University of Paraíba, João Pessoa, Brazil
| | - Rafael C. Ferreira
- Post Graduation Program in Bioactive Natural and Synthetic Products, Federal University of Paraíba, João Pessoa, Brazil
| | - Reinaldo N. de Almeida
- Post Graduation Program in Bioactive Natural and Synthetic Products, Federal University of Paraíba, João Pessoa, Brazil
| | - Renan M. Braga
- Post Graduation Program in Bioactive Natural and Synthetic Products, Federal University of Paraíba, João Pessoa, Brazil
| | - Fagner Carvalho Leite
- Post Graduation Program in Bioactive Natural and Synthetic Products, Federal University of Paraíba, João Pessoa, Brazil
| | | | - Misael Azevedo T. Cavalcanti
- Drug Development and Synthesis Laboratory, Department of Pharmacy, State University of Paraíba, João Pessoa, Brazil
| | - Ricardo O. Moura
- Drug Development and Synthesis Laboratory, Department of Pharmacy, State University of Paraíba, João Pessoa, Brazil
| | - Geovana F. G. Silvestre
- Post Graduation Program in Bioactive Natural and Synthetic Products, Federal University of Paraíba, João Pessoa, Brazil
| | - Leônia M. Batista
- Post Graduation Program in Bioactive Natural and Synthetic Products, Federal University of Paraíba, João Pessoa, Brazil
- Department of Pharmaceutical Sciences, Federal University of Paraíba, João Pessoa, Brazil
| | - Marianna V. Sobral
- Post Graduation Program in Bioactive Natural and Synthetic Products, Federal University of Paraíba, João Pessoa, Brazil
- Department of Pharmaceutical Sciences, Federal University of Paraíba, João Pessoa, Brazil
- *Correspondence: Marianna V. Sobral,
| |
Collapse
|
12
|
Salama MM, Zaghloul RA, Khalil RM, El-Shishtawy MM. Sitagliptin Potentiates the Anti-Neoplastic Activity of Doxorubicin in Experimentally-Induced Mammary Adenocarcinoma in Mice: Implication of Oxidative Stress, Inflammation, Angiogenesis, and Apoptosis. Sci Pharm 2022; 90:42. [DOI: 10.3390/scipharm90030042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2022] Open
Abstract
Sitagliptin (STG) is a highly selective dipeptidyl peptidase-4 inhibitor recently used in the treatment of type 2 diabetes. The current study aimed to investigate the anti-neoplastic effect of STG alone and in combination with Doxorubicin (Dox), a known chemotherapeutic agent but with ominous side effects. After intramuscular inoculation of 2 × 106 Ehrlich tumor cells, Female Swiss mice were divided into tumor-bearing control, STG-treated, Dox-treated, and a combination of STG and Dox-treated groups. The results showed a significant reduction in the tumor growth of the treated animals in comparison with those of the positive control group with a more prominent effect in the co-treated group. Where, the anti-proliferative and apoptotic effect of STG, and its chemo-sensitizing ability, when used in combination with Dox, was mediated by modulation of oxidative stress (MDA and GSH), attenuation of tumor inflammation (IL-6 and IL-1β), and angiogenesis (VEGF), suppressing proliferation (β-catenin and cyclin-D1) and enhancement of apoptosis (survivin, p53, caspase 3). Thus, in conclusion, STG as adjunctive therapy for Dox could be a strategy for the treatment of breast cancer patients, by their ability in hindering cell proliferation and minimizing the associated oxidative and inflammatory adverse reactions.
Collapse
Affiliation(s)
- Mohamed M. Salama
- Department of Biochemistry, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa 35712, Egypt
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Randa A. Zaghloul
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Rania M. Khalil
- Department of Biochemistry, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa 35712, Egypt
| | | |
Collapse
|
13
|
Amer ME, Amer MA, Othman AI, Elsayed DA, El-Missiry MA, Ammar OA. Silymarin inhibits the progression of Ehrlich solid tumor via targeting molecular pathways of cell death, proliferation, angiogenesis, and metastasis in female mice. Mol Biol Rep 2022; 49:4659-4671. [PMID: 35305227 DOI: 10.1007/s11033-022-07315-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 03/01/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Plant-derived phytochemicals have been reported to exert anticancer activity. This study investigated the antitumor role of silymarin (Silybum marianum) (SMN) and its molecular targets in Ehrlich solid tumor xenografts in vivo. METHODS AND RESULTS Female Swiss albino mice were divided into three groups (of five animals each) that were engrafted with Ehrlich tumor (ET) cells with or without SMN treatment. The 3rd groups treated with DMSO only vehicle control group. A significant reduction in animal body mass and tumor volume/weight were observed in xenografted mice treated with SMN. SMN modulated oxidative stress in tumors while enhancing the antioxidant levels in mouse serum. SMN activated both mitochondrial and death receptor-related apoptosis pathways and induced cell cycle arrest, marked by a significant downregulation of cyclin D1 in SMN-treated tumors. Significant decreases in RNA content and protein expression levels of Ki-67 and proliferating cell nuclear antigen were observed in ET cells. Additionally, SMN downregulated vascular endothelial growth factor and nuclear factor-kappa B levels indicating anti-angiogenesis activity of this agent. SMN upregulated the expression of E-cadherin in tumor tissue suggesting, that SMN has potential ability to inhibit metastasis. Tumor tissue from SMN-treated animals showed a remarkable degeneration and reduction in the neoplastic cell density. CONCLUSIONS The anticancer effect was associated with apparent apoptosis in neoplastic cells with abundance of multifocal necrotic areas. SMN was found to inhibit ET growth via enhancing apoptosis, inhibition of cell division and reduction in angiogenesis in vivo. Hypothetical scheme of SMN antitumor effects (mechanism of signaling) in solid ET in vivo. SMN anticancer effect may be mediated by molecular mediators that affect proliferation, cell cycle activity, apoptotic pathways, angiogenesis, and metastasis.
Collapse
Affiliation(s)
- Maggie E Amer
- Zoology Department, Faculty of Sciences, Mansoura University, Mansoura, Egypt.
| | - Maher A Amer
- Zoology Department, Faculty of Sciences, Mansoura University, Mansoura, Egypt
| | - Azza I Othman
- Zoology Department, Faculty of Sciences, Mansoura University, Mansoura, Egypt
| | - Doaa A Elsayed
- Zoology Department, Faculty of Sciences, Mansoura University, Mansoura, Egypt
| | | | - Omar A Ammar
- Basic Science Department, Delta University for Science and Technology, Gamasa, Egypt
| |
Collapse
|
14
|
Mahmoud MA, Okda TM, Omran GA, Abd-Alhaseeb MM. Rosmarinic acid suppresses inflammation, angiogenesis, and improves paclitaxel induced apoptosis in a breast cancer model via NF3 κB-p53-caspase-3 pathways modulation. J Appl Biomed 2021; 19:202-209. [PMID: 34907739 DOI: 10.32725/jab.2021.024] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 10/25/2021] [Indexed: 12/20/2022] Open
Abstract
Rosmarinic acid is a natural polyphenolic compound that is found in different plant species and used for different medicinal purposes. This study aimed to investigate the chemo-preventive effect of rosmarinic acid and evaluate its antitumor efficacy alone or in combination with Paclitaxel in breast cancer mice model. Ehrlich induced mice mammary solid tumor model was used in the study. Mice were treated with oral rosmarinic acid and intraperitoneal Paclitaxel. Inflammation, angiogenesis, and apoptosis were checked. Enzyme linked immunosorbent assay (ELISA), quantitative real time PCR, and immunohistochemical methods were performed. Rosmarinic acid used prior to tumor induction suppressed NF-κB, TNF-α, vascular endothelial growth factor (VEGF) serum levels, and VEGF receptors. It also triggered apoptosis by restoring the levels of P53, Bcl-2, Bax, and caspase-3. Furthermore, in Ehrlich solid tumor mice, rosmarinic acid, and/or Paclitaxel significantly suppressed tumor growth with an increase in apoptotic markers P53 and Caspase-3 levels, and suppressed the Bcl2/Bax ratio. Rosmarinic acid exerted chemo-preventive and therapeutic potential alone or in combination with Paclitaxel. Moreover, rosmarinic acid targets numerous signaling pathways associated with breast cancer.
Collapse
Affiliation(s)
- Marwa A Mahmoud
- Damanhour University, Faculty of Pharmacy, Department of Biochemistry, Behira, Egypt
| | - Tark M Okda
- Damanhour University, Faculty of Pharmacy, Department of Biochemistry, Behira, Egypt
| | - Gamal A Omran
- Damanhour University, Faculty of Pharmacy, Department of Biochemistry, Behira, Egypt
| | - Mohammad M Abd-Alhaseeb
- Damanhour University, Faculty of Pharmacy, Department of Pharmacology and Toxicology, Behira, Egypt
| |
Collapse
|