1
|
Lopes IP, Bagatela BS, Lopes AP, Silveira EF, Petri G, Santos JFR, Perez MM, da Veiga GL, da Costa Aguiar Alves B, Perazzo FF, Feder D, Fonseca FLA. Preclinical Effects of Melatonin on the Development of Ehrlich's Tumor: A Biochemical, Cognitive, and Molecular Approach. Recent Pat Anticancer Drug Discov 2025; 20:113-120. [PMID: 36825727 DOI: 10.2174/1574892818666230223160858] [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: 07/18/2022] [Revised: 12/22/2022] [Accepted: 12/28/2022] [Indexed: 02/25/2023]
Abstract
BACKGROUND It has already been shown that melatonin is an antitumoral molecule that affects malignant cells via some mechanisms. The benefit played by this hormone on cancer is due to its antioxidant effects. OBJECTIVE This study aimed to evaluate the preclinical effects of melatonin in mice with the Ehrlich ascites tumor. METHODS Twenty Balb/c male mice with Ehrlich tumor were treated with different melatonin doses. Their inflammatory and oxidative stress were accessed by gene expression. Hepatotoxicity and hematological parameters were also evaluated through biochemical analyses. Animal welfare was analysed weekly from the categories guided by the NC3Rs. RESULTS Gene expression analyses have shown that only Tnfα and Sod1 were expressed in all groups studied. Only the M-3 group showed increased Tnfα expression compared to the control. All groups treated with melatonin showed decreased Sod1 expression compared to the control. No signs of hepatotoxicity were caused by any of the melatonin doses used in the treatment. CONCLUSION In animals with Ehrlich´s tumor treated with melatonin, a decrease in oxidative stress, an amelioration in welfare and in cognitive tasks could be observed, even if the treatment has not reduced the size of the tumor itself. In parallel with the already patented use of melatonin in the treatment of sleep disorders or chronic kidney disease, our results propose its use to improve the general well-being of breast cancer patients.
Collapse
Affiliation(s)
- Ivan Pereira Lopes
- Laboratório de Análises Clínicas do Centro, Universitário FMABC, Santo André, SP, Brazil
| | - Bianca Souza Bagatela
- Laboratório de Análises Clínicas do Centro, Universitário FMABC, Santo André, SP, Brazil
| | - Andrey Pereira Lopes
- Laboratório de Análises Clínicas do Centro, Universitário FMABC, Santo André, SP, Brazil
| | | | - Giuliana Petri
- Laboratório de Análises Clínicas do Centro, Universitário FMABC, Santo André, SP, Brazil
| | | | - Matheus Moreira Perez
- Laboratório de Análises Clínicas do Centro, Universitário FMABC, Santo André, SP, Brazil
| | | | | | - Fabio Ferreira Perazzo
- Departamento de Ciências Farmacêuticas, Universidade Federal de São Paulo (UNIFESP), Diadema, SP, Brazil
| | - David Feder
- Laboratório de Análises Clínicas do Centro, Universitário FMABC, Santo André, SP, Brazil
| | - Fernando Luiz Affonso Fonseca
- Laboratório de Análises Clínicas do Centro, Universitário FMABC, Santo André, SP, Brazil
- Departamento de Ciências Farmacêuticas, Universidade Federal de São Paulo (UNIFESP), Diadema, SP, Brazil
| |
Collapse
|
2
|
Hassan HAFM, Sedky NK, Nafie MS, Mahdy NK, Fawzy IM, Fayed TW, Preis E, Bakowsky U, Fahmy SA. Sustainable Nanomedicine: Enhancement of Asplatin's Cytotoxicity In Vitro and In Vivo Using Green-Synthesized Zinc Oxide Nanoparticles Formed via Microwave-Assisted and Gambogic Acid-Mediated Processes. Molecules 2024; 29:5327. [PMID: 39598716 PMCID: PMC11596978 DOI: 10.3390/molecules29225327] [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: 10/04/2024] [Revised: 10/30/2024] [Accepted: 11/06/2024] [Indexed: 11/29/2024] Open
Abstract
Chemoresistance encountered using conventional chemotherapy demands novel treatment approaches. Asplatin (Asp), a novel platinum (IV) prodrug designed to release cisplatin and aspirin in a reductive environment, has demonstrated high cytotoxicity at reduced drug resistance. Herein, we investigated the ability of green-synthesized nanocarriers to enhance Asp's efficacy. Zinc oxide nanoparticles (ZnO-NPs) were synthesized using a green microwave-assisted method with the reducing and capping agent gambogic acid (GA). These nanoparticles were then loaded with Asp, yielding Asp@ZnO-NPs. Transmission electron microscopy was utilized to study the morphological features of ZnO-NPs. Cell viability studies conducted on MDA-MB-231 breast cancer cells demonstrated the ability of the Asp@ZnO-NPs treatment to significantly decrease Asp's half-maximal inhibitory concentration (IC50) (5 ± 1 µg/mL). This was further demonstrated using flow cytometric analysis that revealed the capacity of Asp@ZnO-NPs treatment to significantly increase late apoptotic fractions. Furthermore, in vivo studies carried out using solid Ehrlich carcinoma-bearing mice showed significant tumor volume reduction with the Asp@ZnO-NPs treatment (156.3 ± 7.6 mm3), compared to Asp alone (202.3 ± 8.4 mm3) and untreated controls (342.6 ± 10.3 mm3). The histopathological analysis further demonstrated the increased necrosis in Asp@ZnO-NPs-treated group. This study revealed that Asp@ZnO-NPs, synthesized using an eco-friendly approach, significantly enhanced Asp's anticancer activity, offering a sustainable solution for potent anticancer formulations.
Collapse
Affiliation(s)
- Hatem A. F. M. Hassan
- Medway School of Pharmacy, University of Kent, Chatham Maritime, Kent ME4 4TB, UK;
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt;
| | - Nada K. Sedky
- Department of Biochemistry, School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation, R5 New Garden City, New Administrative Capital, Cairo 11835, Egypt;
| | - Mohamed S. Nafie
- Department of Chemistry, College of Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates;
- Chemistry Department, Faculty of Science, Suez Canal University, Ismailia 41522, Egypt
| | - Noha Khalil Mahdy
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt;
- School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation, R5 New Garden City, New Administrative Capital, Cairo 11835, Egypt
| | - Iten M. Fawzy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Future University in Egypt, Cairo 11835, Egypt
| | - Toka Waleed Fayed
- School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation, R5 New Garden City, New Administrative Capital, Cairo 11835, Egypt
| | - Eduard Preis
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, Robert-Koch-Str. 4, 35037 Marburg, Germany
| | - Udo Bakowsky
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, Robert-Koch-Str. 4, 35037 Marburg, Germany
| | - Sherif Ashraf Fahmy
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, Robert-Koch-Str. 4, 35037 Marburg, Germany
| |
Collapse
|
3
|
Alotaibi BS, El-Masry TA, Selim H, El-Bouseary MM, El-Sheekh MM, Makhlof MEM, El-Nagar MMF. New insights into the anticancer effects of Polycladia crinita aqueous extract and its selenium nanoformulation against the solid Ehrlich carcinoma model in mice via VEGF, notch 1, NF-кB, cyclin D1, and caspase 3 signaling pathway. Front Pharmacol 2024; 15:1345516. [PMID: 38469406 PMCID: PMC10926956 DOI: 10.3389/fphar.2024.1345516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 02/05/2024] [Indexed: 03/13/2024] Open
Abstract
Background: Phaeophyceae species are enticing interest among researchers working in the nanotechnology discipline, because of their diverse biological activities such as anti-inflammatory, antioxidant, anti-microbial, and anti-tumor. In the present study, the anti-cancer properties of Polycladia crinita extract and green synthesized Polycladia crinita selenium nanoparticles (PCSeNPs) against breast cancer cell line (MDA-MB-231) and solid Ehrlich carcinoma (SEC) were investigated. Methods: Gas chromatography-mass spectroscopy examinations of Polycladia crinita were determined and various analytical procedures, such as SEM, TEM, EDX, and XRD, were employed to characterize the biosynthesized PCSeNPs. In vitro, the anticancer activity of free Polycladia crinita and PCSeNPs was evaluated using the viability assay against MDA-MB-231, and also cell cycle analysis by flow cytometry was determined. Furthermore, to study the possible mechanisms behind the in vivo anti-tumor action, mice bearing SEC were randomly allocated into six equal groups (n = 6). Group 1: Tumor control group, group 2: free SeNPs, group 3: 25 mg/kg Polycladia crinita, group 4: 50 mg/kg Polycladia crinita, group 5: 25 mg/kg PCSeNPs, group 6: 50 mg/kg PCSeNPs. Results: Gas chromatography-mass spectroscopy examinations of Polycladia crinita extract exposed the presence of many bioactive compounds, such as 4-Octadecenoic acid-methyl ester, Tetradecanoic acid, and n-Hexadecenoic acid. These compounds together with other compounds found, might work in concert to encourage the development of anti-tumor activities. Polycladia crinita extract and PCSeNPs were shown to inhibit cancer cell viability and early cell cycle arrest. Concentrations of 50 mg/kg of PCSeNPs showed suppression of COX-2, NF-кB, VEGF, ki-67, Notch 1, and Bcl-2 protein levels. Otherwise, showed amplification of the caspase 3, BAX, and P53 protein levels. Moreover, gene expression of caspase 3, caspase 9, Notch 1, cyclin D1, NF-кB, IL-6, and VEGF was significantly more effective with PCSeNPs than similar doses of free extract. Conclusion: The PCSeNPs mediated their promising anti-cancerous action by enhancing apoptosis and mitigating inflammation, which manifested in promoting the total survival rate and the tumor volume decrease.
Collapse
Affiliation(s)
- Badriyah S. Alotaibi
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Thanaa A. El-Masry
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Hend Selim
- Department of Biochemistry, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Maisra M. El-Bouseary
- Department of Microbiology and Immunology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | | | - Mofida E. M. Makhlof
- Botany and Microbiology Department, Faculty of Science, Damanhour University, Damanhour, Egypt
| | - Maysa M. F. El-Nagar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| |
Collapse
|
4
|
Abd El-Salam M, El-Tanbouly G, Bastos J, Metwaly H. Suppression of VEGF and inflammatory cytokines, modulation of Annexin A1 and organ functions by galloylquinic acids in breast cancer model. Sci Rep 2023; 13:12268. [PMID: 37507468 PMCID: PMC10382581 DOI: 10.1038/s41598-023-37654-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 06/25/2023] [Indexed: 07/30/2023] Open
Abstract
The ongoing development of novel drugs for breast cancer aims to improve therapeutic outcomes, reduce toxicities, and mitigate resistance to chemotherapeutic agents. Doxorubicin (Dox) is known for its significant side effects caused by non-specific cytotoxicity. In this study, we investigated the antitumor activity of galloylquinic acids (BF) and the beneficial role of their combination with Dox in an Ehrlich ascites carcinoma (EAC)-bearing mouse model, as well as their cytotoxic effect on MCF-7 cells. The EAC-mice were randomized into five experimental groups: normal saline, Dox (2 mg/kg, i.p), BF (150 mg/kg, orally), Dox and BF combined mixture, and a control group. Mice were subjected to a 14-day treatment regimen. Results showed that BF compounds exerted chemopreventive effects in EAC mice group by increasing mean survival time, decreasing tumor volume, inhibiting ascites tumor cell count, modulating body weight changes, and preventing multi-organ histopathological alterations. BF suppressed the increased levels of inflammatory mediators (IL-6 and TNF-α) and the angiogenic marker VEGF in the ascitic fluid. In addition, BF and their combination with Dox exhibited significant cytotoxic activity on MCF-7 cells by inhibiting cell viability and modulating Annexin A1 level. Moreover, BF treatments could revert oxidative stress, restore liver and kidney functions, and normalize blood cell counts.
Collapse
Affiliation(s)
- Mohamed Abd El-Salam
- Department of Pharmacognosy, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, 11152, Egypt.
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, D02 VN51, Ireland.
| | - Ghada El-Tanbouly
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, 11152, Egypt
| | - Jairo Bastos
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, 14040-900, Brazil
| | - Heba Metwaly
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Alexandria University, Alexandria, 21500, Egypt.
| |
Collapse
|
5
|
Kabil MF, Mahmoud MY, Bakr AF, Zaafar D, El-Sherbiny IM. Switching indication of PEGylated lipid nanocapsules-loaded with rolapitant and deferasirox against breast cancer: Enhanced in-vitro and in-vivo cytotoxicity. Life Sci 2022; 305:120731. [PMID: 35753435 DOI: 10.1016/j.lfs.2022.120731] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 06/11/2022] [Accepted: 06/20/2022] [Indexed: 12/12/2022]
Abstract
Breast cancer (BC) is considered the leading cause of mortality and morbidity among adult women worldwide, and it is associated with many genetic or hormonal factors. Despite the advanced therapeutic and theranostic strategies for BC treatment, cancer metastasis and relapse are often observed among patients which lead to therapeutic failure. Accordingly, among the repositioned medication against BC proliferation is neurokinin receptor antagonists and iron chelating agents especially rolapitant HCl (RP) and deferasirox (DFO), respectively. However, RP and DFO are classified as class II with low aqueous solubility. Both drugs were nanoformulated into PEGylated lipid nanocapsules (LNCs) for enhancing their aqueous solubility and augmenting their efficacy. RP-LNCs, DFO-LNCs and their combinations were evaluated according to particle size (PS), zeta potential, polydispersity index (PDI) and surface morphology. Importantly, the antitumor effect of these novel molecules and their nanoforms was evaluated against the suppression of Ehrlich Ascites tumor model using female mice. Results revealed that RP-LNCs, DFO-LNCs and RP/DFO-LNCs exerted PS from 45.23 ± 3.54 to 60.1 ± 3.32 nm with PDI around 0.20 which indicates homogenous particles distribution. Also, RP-LNCs, DFO-LNCs and RP/DFO-LNCs displayed surface charges of +16.6 ± 6.9, -13.3 ± 5.82 and - 20.2 ± 5.40 mV, respectively. The obtained LNCs conferred a high potent cytotoxic effect against MCF7 cancer cells as compared to parent drugs, with IC50 of 10.86 ± 0.89, 3.34 ± 0.99 and 2.24 ± 0.97 μg/mL for RP-LNCs, DFO-LNCs and RP/DFO-LNCs, respectively. The in-vivo pharmacodynamics effect of the developed nano-formulations showed superior antitumor effect for the individual drugs rather than their combinations as compared to the control group. The current study confirmed the potential of RP and DFO nanoforms as promising therapeutic agents for BC treatment.
Collapse
Affiliation(s)
- Mohamed Fawzi Kabil
- Nanomedicine Research Labs, Center for Materials Science, Zewail City of Science and Technology, 6th of October City, Giza, Egypt
| | - Mohamed Y Mahmoud
- Department of Toxicology and Forensic Medicine, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Alaa F Bakr
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Dalia Zaafar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Modern University for Technology and Information, Cairo, Egypt
| | - Ibrahim M El-Sherbiny
- Nanomedicine Research Labs, Center for Materials Science, Zewail City of Science and Technology, 6th of October City, Giza, Egypt.
| |
Collapse
|
6
|
Abdraboh ME, El-Missiry MA, Othman AI, Taha AN, Elhamed DSA, Amer ME. Constant light exposure and/or pinealectomy increases susceptibility to trichloroethylene-induced hepatotoxicity and liver cancer in male mice. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:60371-60384. [PMID: 35419691 PMCID: PMC9427929 DOI: 10.1007/s11356-022-19976-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 03/25/2022] [Indexed: 05/09/2023]
Abstract
Exposure to light at night, pineal gland impairment, and the environmental pollutant trichloroethylene (TCE) have serious implications for health and contribute to illness, including liver cancer. The adverse effect of the association of continuous exposure to light with decreased melatonin levels and TCE-induced toxicity is not disclosed in target organs. This work explored the role of light and pineal impairment in increasing susceptibility to liver toxicity and cancer upon exposure to TCE. Male albino mice were divided into groups as follows: control group (12-h light/12-h dark cycle), constant light (24-h light), pinealectomized (Pnx) mice, sham surgically treated group, TCE-treated groups subjected to two doses (500 and 1000 mg/kg) at two different light regimens, and combination of Pnx and TCE-treated mice kept at a 12-h light/12-h dark cycle. Melatonin levels were significantly decreased in both Pnx mice and TCE-treated animals at both light regimens. Aspartate transaminase, alanine aminotransferase, activities, and serum bilirubin levels were significantly elevated, whereas albumin levels were markedly decreased in Pnx mice, TCE-treated mice, and the combination group. Histopathological investigations reflected changes in liver function parameters indicating liver injury and induction of cancer. These effects were accompanied by significant increase of the liver cancer biomarker alpha-fetoprotein and the expression of the metastatic markers CD44, TGFβ-1, and VEGF, along with increased oxidative stress indicators and inflammatory cytokines (IL-6, IL-1β, and TNF-α) in both Pnx and TCE-treated mice and the combination group at both light regimens. Taken together, our findings indicated that low melatonin levels, exposure to constant light, and the combination of both factors increases susceptibility to the toxic and carcinogenic effects of TCE on the liver.
Collapse
Affiliation(s)
- Mohamed E Abdraboh
- Zoology Department, Faculty of Science, Mansoura University, Mansoura, Egypt
- Faculty of Science, New Mansoura University, Mansoura, Egypt
| | | | - Azza I Othman
- Zoology Department, Faculty of Science, Mansoura University, Mansoura, Egypt
| | - Ahmed Nageeb Taha
- Neurosurgery Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Dalia S Abd Elhamed
- Zoology Department, Faculty of Science, Mansoura University, Mansoura, Egypt
| | - Maggie E Amer
- Zoology Department, Faculty of Science, Mansoura University, Mansoura, Egypt
| |
Collapse
|
7
|
Diversifying the skin cancer-fighting worthwhile frontiers: How relevant are the itraconazole/ascorbyl palmitate nanovectors? NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2022; 43:102561. [PMID: 35417773 DOI: 10.1016/j.nano.2022.102561] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 03/20/2022] [Accepted: 03/30/2022] [Indexed: 12/13/2022]
Abstract
Fighting malignant neoplasms via repurposing existing drugs could be a welcome move for prosperous cancer remediations. In the current work, nanovehiculation and optimization of the repositioned itraconazole (ITZ) utilizing ascorbyl palmitate (AP) aspasomes would be an auspicious approach. Further, the optimized aspasomes were incorporated in a cream and tracked for skin deposition. The in vivo efficacy of aspasomal cream on mice subcutaneous Ehrlich carcinoma model was also assessed. The optimized aspasomes revealed nano size (67.83 ± 6.16 nm), negative charge (-79.40 ± 2.23 mV), > 95% ITZ entrapment and high colloidal stability. AP yielded substantial antioxidant capacity and pushed the ITZ cytotoxicity forward against A431 cells (IC50 = 5.3±0.27 μg/mL). An appealing privilege was the aspasomal cream that corroborated spreadability, contemplated skin permeation and potentiated in vivo anticancer competence, reflected in 62.68% reduction in the tumor weight. Such synergistic tumor probes set the foundation for futuristic clinical translation and commercialization.
Collapse
|
8
|
Formulation and Characterization of Doxycycline-Loaded Polymeric Nanoparticles for Testing Antitumor/Antiangiogenic Action in Experimental Colon Cancer in Mice. NANOMATERIALS 2022; 12:nano12050857. [PMID: 35269343 PMCID: PMC8912660 DOI: 10.3390/nano12050857] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/22/2022] [Accepted: 02/26/2022] [Indexed: 02/04/2023]
Abstract
Nanotherapeutics can enhance the characteristics of drugs, such as rapid systemic clearance and systemic toxicities. Polymeric nanoparticles (PRNPs) depend on dispersion of a drug in an amorphous state in a polymer matrix. PRNPs are capable of delivering drugs and improving their safety. The primary goal of this study is to formulate doxycycline-loaded PRNPs by applying the nanoprecipitation method. Eudragit S100 (ES100) (for DOX-PRNP1) and hydroxypropyl methyl cellulose phthalate HP55 (for DOX-PRNP2) were tested as the drug carrying polymers and the DOX-PRNP2 showed better characteristics and drug release % and was hence selected to be tested in the biological study. Six different experimental groups were formed from sixty male albino mice. 1,2,-Dimethylhydrazine was used for 16 weeks to induce experimental colon cancer. We compared the oral administration of DOX-PRNP2 in doses of 5 and 10 mg/kg with the free drug. Results indicated that DOX-PRNP2 had greater antitumor activity, as evidenced by an improved histopathological picture for colon specimens as well as a decrease in the tumor scores. In addition, when compared to free DOX, the DOX-PRNP2 reduced the angiogenic indicators VEGD and CD31 to a greater extent. Collectively, the findings demonstrated that formulating DOX in PRNPs was useful in enhancing antitumor activity and can be used in other models of cancers to verify their efficacy and compatibility with our study.
Collapse
|
9
|
Ammar OA, El-Missiry MA, Othman AI, Amer ME. Melatonin is a potential oncostatic agent to inhibit HepG2 cell proliferation through multiple pathways. Heliyon 2022; 8:e08837. [PMID: 35141433 PMCID: PMC8814902 DOI: 10.1016/j.heliyon.2022.e08837] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 12/22/2021] [Accepted: 01/24/2022] [Indexed: 11/25/2022] Open
Abstract
CONTEXT Chemotherapy is a cornerstone in the treatment of hepatocellular carcinoma (HCC). Melatonin is a pineal hormone that targets various cancers, however, its antitumor pathways are still not fully elucidated. OBJECTIVE This study investigated melatonin's antitumor molecular mechanisms to inhibit the proliferation of HepG2 cells. MATERIALS AND METHODS HepG2 Cells were classified into cells without treatment as a control group and cells treated with melatonin (5.4 mmol/L) for 48 h. Proliferating cell nuclear antigen (PCNA) and marker of proliferation Ki-67 were estimated using immunohistochemical analysis. Apoptosis and cell cycle were evaluated using flow cytometric analysis. Apoptotic markers were detected using RT-qPCR assay. Antioxidants and oxidative stress biomarkers were performed using a colorimetric assay. RESULTS Melatonin produced a remarkable steady decrease in the viability of HepG2 cells at a concentration range between 5-20 mmol/L. Melatonin suppressed cell proliferation in the G2/M phase of the cell cycle (34.97 ± 0.92%) and induced apoptosis (12.43 ± 0.73%) through up-regulating p21 and p53 that was confirmed by the reduction of PCNA and Ki-67 expressions. Additionally, melatonin repressed angiogenesis evidenced by the down-regulation of angiopoietin-2, vascular endothelial growth factor receptor-2 expressions (0.42-fold change), and the level of CD133. Moreover, melatonin augmented the oxidative stress manifested by a marked increase of 4-hydroxynonenal levels with a reduction of glutathione content and superoxide dismutase activity. DISCUSSION AND CONCLUSION Melatonin inhibits proliferation and angiogenesis and induced apoptosis and oxidative stress in HepG2 cells. These results indicate the oncostatic effectiveness of melatonin on liver cancer.
Collapse
Affiliation(s)
- Omar A. Ammar
- Basic Science Department, Delta University for Science and Technology, Gamasa, Egypt
| | | | - Azza I. Othman
- Zoology Department, Faculty of Science, Mansoura University, Egypt
| | - Maggie E. Amer
- Zoology Department, Faculty of Science, Mansoura University, Egypt
| |
Collapse
|
10
|
The ponatinib/gossypol novel combination provides enhanced anticancer activity against murine solid Ehrlich carcinoma via triggering apoptosis and inhibiting proliferation/angiogenesis. Toxicol Appl Pharmacol 2021; 432:115767. [PMID: 34699866 DOI: 10.1016/j.taap.2021.115767] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 10/17/2021] [Accepted: 10/18/2021] [Indexed: 11/20/2022]
Abstract
The search for new antitumor agents or combinations that are more effective and, hopefully, provide fewer health hazards is ongoing. Therefore, this study investigated the efficacy of a novel combination of ponatinib, a multi-targeted tyrosine kinase inhibitor, and the natural phytochemical gossypol against murine solid Ehrlich carcinoma. Six groups of ten mice each received vehicle (I), ponatinib in doses of 10 and 15 mg/kg (II, III) respectively, gossypol in a dose of 4 mg/kg (IV), and ponatinib (10 or 15 mg/kg) in combination with gossypol (4 mg/kg; V, VI). All treatments started on the 12th post-Ehrlich ascites carcinoma (EAC) implantation day and were administered intraperitoneally in daily doses for 3 weeks. Treatment of EAC-bearing mice with ponatinib/gossypol combination improved anticancer efficacy over either drug alone, as demonstrated by greater decreases in tumor weight and volume, and ponatinib (10 mg/kg)/gossypol combination was more efficient than ponatinib (15 mg/kg). Mechanistically, the ponatinib/gossypol combination significantly increased apoptotic markers p53, Bax, and caspase-9 while decreasing anti-apoptotic marker Bcl-2. Furthermore, it greatly decreased proliferative and angiogenic markers, FGFR4 and VEGF, respectively. Histopathology revealed a significant decline in neoplastic cells, the majority of which have necrotic changes and numerous apoptotic bodies, as well as a decrease in mitotic figures and tumor giant cells, indicating the capacity to suppress cancer proliferation/persistence. Overall, gossypol could be used as an adjuvant medication for ponatinib in cancer treatment, possibly leading to successful dose reductions and fewer side effects; however, further research is needed before a clinical application could be feasible.
Collapse
|
11
|
Samec M, Liskova A, Koklesova L, Zhai K, Varghese E, Samuel SM, Šudomová M, Lucansky V, Kassayova M, Pec M, Biringer K, Brockmueller A, Kajo K, Hassan STS, Shakibaei M, Golubnitschaja O, Büsselberg D, Kubatka P. Metabolic Anti-Cancer Effects of Melatonin: Clinically Relevant Prospects. Cancers (Basel) 2021; 13:3018. [PMID: 34208645 PMCID: PMC8234897 DOI: 10.3390/cancers13123018] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/04/2021] [Accepted: 06/14/2021] [Indexed: 02/06/2023] Open
Abstract
Metabolic reprogramming characterized by alterations in nutrient uptake and critical molecular pathways associated with cancer cell metabolism represents a fundamental process of malignant transformation. Melatonin (N-acetyl-5-methoxytryptamine) is a hormone secreted by the pineal gland. Melatonin primarily regulates circadian rhythms but also exerts anti-inflammatory, anti-depressant, antioxidant and anti-tumor activities. Concerning cancer metabolism, melatonin displays significant anticancer effects via the regulation of key components of aerobic glycolysis, gluconeogenesis, the pentose phosphate pathway (PPP) and lipid metabolism. Melatonin treatment affects glucose transporter (GLUT) expression, glucose-6-phosphate dehydrogenase (G6PDH) activity, lactate production and other metabolic contributors. Moreover, melatonin modulates critical players in cancer development, such as HIF-1 and p53. Taken together, melatonin has notable anti-cancer effects at malignancy initiation, progression and metastasing. Further investigations of melatonin impacts relevant for cancer metabolism are expected to create innovative approaches supportive for the effective prevention and targeted therapy of cancers.
Collapse
Affiliation(s)
- Marek Samec
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia; (M.S.); (A.L.); (L.K.); (K.B.)
| | - Alena Liskova
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia; (M.S.); (A.L.); (L.K.); (K.B.)
| | - Lenka Koklesova
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia; (M.S.); (A.L.); (L.K.); (K.B.)
| | - Kevin Zhai
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha P.O. Box 24144, Qatar; (K.Z.); (E.V.); (S.M.S.)
| | - Elizabeth Varghese
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha P.O. Box 24144, Qatar; (K.Z.); (E.V.); (S.M.S.)
| | - Samson Mathews Samuel
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha P.O. Box 24144, Qatar; (K.Z.); (E.V.); (S.M.S.)
| | - Miroslava Šudomová
- Museum of Literature in Moravia, Klašter 1, 66461 Rajhrad, Czech Republic;
| | - Vincent Lucansky
- Biomedical Centre Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Mala Hora 4D, 036 01 Martin, Slovakia;
| | - Monika Kassayova
- Department of Animal Physiology, Institute of Biology and Ecology, Faculty of Science, P. J. Šafarik University, 04001 Košice, Slovakia;
| | - Martin Pec
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia;
| | - Kamil Biringer
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia; (M.S.); (A.L.); (L.K.); (K.B.)
| | - Aranka Brockmueller
- Musculoskeletal Research Group and Tumour Biology, Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilian-University Munich, D-80336 Munich, Germany; (A.B.); (M.S.)
| | - Karol Kajo
- Department of Pathology, St. Elizabeth Cancer Institute Hospital, 81250 Bratislava, Slovakia;
- Biomedical Research Centre, Slovak Academy of Sciences, 81439 Bratislava, Slovakia
| | - Sherif T. S. Hassan
- Department of Applied Ecology, Faculty of Environmental Sciences, Czech University of Life Sciences Prague, Kamýcká 129, 165 00 Prague, Czech Republic;
| | - Mehdi Shakibaei
- Musculoskeletal Research Group and Tumour Biology, Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilian-University Munich, D-80336 Munich, Germany; (A.B.); (M.S.)
| | - Olga Golubnitschaja
- European Association for Predictive, Preventive and Personalised Medicine, EPMA, 1160 Brussels, Belgium;
- Predictive, Preventive and Personalised (3P) Medicine, Department of Radiation Oncology, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, 53127 Bonn, Germany
| | - Dietrich Büsselberg
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha P.O. Box 24144, Qatar; (K.Z.); (E.V.); (S.M.S.)
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia;
- European Association for Predictive, Preventive and Personalised Medicine, EPMA, 1160 Brussels, Belgium;
| |
Collapse
|
12
|
El-Missiry MA, Shabana S, Ghazala SJ, Othman AI, Amer ME. Melatonin exerts a neuroprotective effect against γ-radiation-induced brain injury in the rat through the modulation of neurotransmitters, inflammatory cytokines, oxidative stress, and apoptosis. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:31108-31121. [PMID: 33598836 DOI: 10.1007/s11356-021-12951-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 02/09/2021] [Indexed: 05/11/2023]
Abstract
The current study aimed to investigate the ameliorative effect of melatonin (MLT) against brain injury in rats undergoing whole-body exposure to γ-radiation. Male Wistar rats were whole-body exposed to 4-Gy γ-radiation from a cesium-137 source. MLT (10 mg/kg) was orally administrated 30 minutes before irradiation and continued once daily for 1 and 7 days after exposure. In the irradiated rats, the plasma levels of glutamate were increased, while the gamma-aminobutyric acid (GABA) levels were decreased, and MLT improved the disturbed glutamate and GABA levels. These effects paralleled an increase in pro-inflammatory cytokines (IL-1b, IL-6, and TNF-a) and C-reactive protein as well as a decrease in IL-10 in the plasma of the irradiated rats. MLT treatment markedly reduced these effects, indicating its anti-inflammatory impact. Immunohistochemical studies demonstrated a remarkable upregulation of caspase-3 and P53 expression, indicating the increased apoptosis in the brain of irradiated rats. MLT significantly downregulated the expression of these parameters compared with that in the irradiated rats, indicating its anti-apoptotic effect. Oxidative stress is developed in the brain as evidenced by increased levels of malondialdehyde; decreased activities of superoxide dismutase, catalase, and glutathione peroxidase; and decreased content of glutathione in the brain. MLT remarkably ameliorated the development of oxidative stress in the brain of the irradiated rats indicating its antioxidant impact. The histopathological results were consistent with the biochemical and immunohistochemical results and showed that MLT remarkably protected the histological structure of brain tissue compared with that in the irradiated rats. In conclusion, MLT showed potential neuroprotective properties by increasing the release of neurotransmitters, antioxidants, and anti-inflammatory factors and reducing pro-inflammatory cytokines and apoptosis in the brain of irradiated rats. MLT can be beneficial in clinical and occupational settings requiring radiation exposure; however, additional studies are required to elucidate its neuroprotective effect in humans.
Collapse
Affiliation(s)
| | - Sameh Shabana
- Faculty of Science, Mansoura University, Mansoura, Egypt
| | - Sara J Ghazala
- Faculty of Science, Mansoura University, Mansoura, Egypt
| | - Azza I Othman
- Faculty of Science, Mansoura University, Mansoura, Egypt
| | - Maggie E Amer
- Faculty of Science, Mansoura University, Mansoura, Egypt
| |
Collapse
|
13
|
Liu K, Xue B, Bai G, Zhang W. Downregulation of Diacylglycerol kinase zeta (DGKZ) suppresses tumorigenesis and progression of cervical cancer by facilitating cell apoptosis and cell cycle arrest. Bioengineered 2021; 12:1517-1529. [PMID: 33926342 PMCID: PMC8806244 DOI: 10.1080/21655979.2021.1918505] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Diacylglycerol kinase zeta (DGKZ) participates in cancer progression. Here, the current work aims to identify the functional role of DGKZ in cervical cancer (CC). DGKZ expression in cervical cancer tissues and paired adjacent normal cervical tissues was assessed using Immunohistochemistry assay. SiHa and HeLa cells were transfected with lentivirus plasmids (sh-DGKZ or sh-NC) to evaluate the effects of DGKZ knockdown on cell proliferation, apoptosis and cell cycle distribution in vitro. Furthermore, BALB/c nude mice were injected subcutaneously with Lentivirus-sh-DGKZ-SiHa cells or Lentivirus-sh-NC-SiHa cells to analyze the influence of DGKZ silencing on tumor growth of CC in vivo. Moreover, the potential molecular mechanisms were predicted by GO and KEGG analysis and preliminarily explored through PathScan Analysis. Elevated DGKZ expression in cervical tumor was observed. Downregulation of DGKZ repressed proliferation and boosted apoptosis of SiHa and HeLa cells and induced cell cycle arrest at G0/G1 phase. In addition, Knockdown of DGKZ restrained tumor growth in tumor xenograft mice. Importantly, GO and KEGG analysis displayed that differentially expressed proteins induced by silence of DGKZ were mostly enriched in autophagy or mitophagy, indicating that the functions of DGKZ on cell proliferation and tumor growth may be associated with autophagy or mitophagy. PathScan analysis presented that PI3K-AKT and TAK1-NF-κB signaling pathways were prominently inhibited in SiHa cells transfected with sh-DGKZ. In summary, downregulation of DGKZ impeded cell proliferation, boosted cell apoptosis and induced cell cycle arrest to suppress tumorigenesis and progression of cervical cancer.
Collapse
Affiliation(s)
- Keying Liu
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China.,Department of Gynecology and Obstetrics, Xi'an North Hospital, Xi'an, Shaanxi Province, China
| | - Biyun Xue
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi Province, China
| | - Guiqin Bai
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Wentao Zhang
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| |
Collapse
|
14
|
Mehrzadi S, Pourhanifeh MH, Mirzaei A, Moradian F, Hosseinzadeh A. An updated review of mechanistic potentials of melatonin against cancer: pivotal roles in angiogenesis, apoptosis, autophagy, endoplasmic reticulum stress and oxidative stress. Cancer Cell Int 2021; 21:188. [PMID: 33789681 PMCID: PMC8011077 DOI: 10.1186/s12935-021-01892-1] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 03/23/2021] [Indexed: 12/19/2022] Open
Abstract
Cancers are serious life-threatening diseases which annually are responsible for millions of deaths across the world. Despite many developments in therapeutic approaches for affected individuals, the rate of morbidity and mortality is high. The survival rate and life quality of cancer patients is still low. In addition, the poor prognosis of patients and side effects of the present treatments underscores that finding novel and effective complementary and alternative therapies is a critical issue. Melatonin is a powerful anticancer agent and its efficiency has been widely documented up to now. Melatonin applies its anticancer abilities through affecting various mechanisms including angiogenesis, apoptosis, autophagy, endoplasmic reticulum stress and oxidative stress. Regarding the implication of mentioned cellular processes in cancer pathogenesis, we aimed to further evaluate the anticancer effects of melatonin via these mechanisms.
Collapse
Affiliation(s)
- Saeed Mehrzadi
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hossein Pourhanifeh
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Alireza Mirzaei
- Bone and Joint Reconstruction Research Center, Shafa Orthopedic Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Farid Moradian
- Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Azam Hosseinzadeh
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
15
|
Gulbahce-Mutlu E, Baltaci SB, Menevse E, Mogulkoc R, Baltaci AK. The Effect of Zinc and Melatonin Administration on Lipid Peroxidation, IL-6 Levels, and Element Metabolism in DMBA-Induced Breast Cancer in Rats. Biol Trace Elem Res 2021; 199:1044-1051. [PMID: 32572799 DOI: 10.1007/s12011-020-02238-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 06/08/2020] [Indexed: 02/07/2023]
Abstract
The purpose of this study was to investigate the effects of zinc and melatonin administration on interleukin-6, lipid peroxidation parameters, and element metabolism in DMBA-induced breast cancer in female rats. A total of 42 recently weaned Wistar rats were divided into 5 groups as follows: control (group 1), DMBA control (group 2), DMBA + zinc (group 3), DMBA + melatonin (group 4), and DMBA + melatonin and zinc (group 5). Malondialdehyde (MDA) and glutathione (GSH) levels in breast tissue and blood samples were determined via spectrophotometric methods. In addition, iron, magnesium, zinc, and copper levels in serum samples were determined by atomic emission, and plasma interleukin-6 levels were determined by ELISA method. The highest tissue and plasma MDA and the lowest tissue and erythrocyte GSH levels found in the study were in group 2; the highest tissue and erythrocyte GSH levels and the lowest tissue and plasma MDA levels are in group 5 (P < 0.05). Iron, magnesium, and zinc levels of groups 3, 4, and 5 were higher than the DMBA group without administration (group 2), but the copper values were significantly lower (P < 0.05). The highest IL-6 levels were determined in group 2 while IL-6 levels in the DMBA group (G5) treated with combined melatonin and zinc were lower than all other breast cancer groups (P < 0.05). According to the findings obtained in this presented study, combined zinc and melatonin therapy can contribute to the prevention of tumor growth by improving the disruption in element metabolism and suppressing IL-6 levels and reducing tissue damage that causes the cancer.
Collapse
Affiliation(s)
- Elif Gulbahce-Mutlu
- Medical Facultuy, Department of Medical Biology, KTO Karatay University, Konya, Turkey
| | | | - Esma Menevse
- Department of Biochemistry, Faculty of Medicine, Selçuk University, Konya, Turkey
| | - Rasim Mogulkoc
- Department of Physiology, Faculty of Medicine, Selçuk University, Konya, Turkey
| | | |
Collapse
|
16
|
Castelló CM, Miguel MP, Silveira-Lacerda EDP, Bakuzis AF, Borges NC. B-Mode and Doppler Ultrasonography in a Murine Model of Ehrlich Solid Carcinoma With Different Growth Patterns. Front Oncol 2020; 10:560413. [PMID: 33251133 PMCID: PMC7673667 DOI: 10.3389/fonc.2020.560413] [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: 05/08/2020] [Accepted: 10/13/2020] [Indexed: 11/18/2022] Open
Abstract
Ehrlich solid carcinoma (ESC) is one of the tumor models used in cancer research. Although it is widely used, it has no ultrasonographic descriptions. In this study, serial B-mode and Doppler ultrasonographic examinations were performed for 23 days for ESCs inoculated into 18 Swiss albino mice. The growth patterns were analyzed, and on the basis of their growth curve, the tumors were classified into two groups: fast growth (FG) and slow growth (SG). Ultrasonographic characteristics of the tumor’s capsule, margins, echogenicity, echotexture, vascular index (VI), distribution of vascular flow, and Doppler indices such as the resistive index, pulsatility index, and peak systolic velocity (SV) were analyzed and compared between the two groups. A high VI and earlier blood flow were noted in the FG group (p<0.05). Additionally, SV was higher in the FG group than in the SG group (13.28 ± 0.38 cm/s vs. 8.43 ± 0.26 cm/s). In contrast, a change in echogenicity and flow distribution patterns were observed, especially in FG tumors. Therefore, ESC presented with few ultrasonographic differences between FG and SG tumors, especially vascularization during the initial stages of tumor growth.
Collapse
Affiliation(s)
- Carla Martí Castelló
- Programa de Pós-Graduação em Ciência Animal, Escola de Veterinária e Zootecnia, Universidade Federal de Goiás, Goiânia, Brazil
| | - Marina Pacheco Miguel
- Programa de Pós-Graduação em Ciência Animal, Escola de Veterinária e Zootecnia, Universidade Federal de Goiás, Goiânia, Brazil.,Setor de Patologia, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Brazil
| | | | | | - Naida Cristina Borges
- Programa de Pós-Graduação em Ciência Animal, Escola de Veterinária e Zootecnia, Universidade Federal de Goiás, Goiânia, Brazil
| |
Collapse
|
17
|
Xin T, Lu C. SirT3 activates AMPK-related mitochondrial biogenesis and ameliorates sepsis-induced myocardial injury. Aging (Albany NY) 2020; 12:16224-16237. [PMID: 32721927 PMCID: PMC7485737 DOI: 10.18632/aging.103644] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 06/22/2020] [Indexed: 12/15/2022]
Abstract
Sirtuin-3 (SirT3) and AMPK stimulate mitochondrial biogenesis, which increases mitochondrial turnover and cardiomyocyte regeneration. We studied the effects of SirT3, AMPK, and mitochondrial biogenesis on sepsis-induced myocardial injury. Our data showed that after treating cardiomyocytes with lipopolysaccharide, SirT3 and AMPK levels decreased, and this was followed by mitochondrial dysfunction and cardiomyocyte death. Overexpression of SirT3 activated the AMPK pathway and improved mitochondrial biogenesis, which is required to sustain mitochondrial redox balance, maintain mitochondrial respiration, and suppress mitochondrial apoptosis. Inhibition of mitochondrial biogenesis abolished SirT3/AMPK-induced cardioprotection by causing mitochondrial damage. These findings indicate that SirT3 reduces sepsis-induced myocardial injury by activating AMPK-related mitochondrial biogenesis.
Collapse
Affiliation(s)
- Ting Xin
- Department of Cardiology, Tianjin First Central Hospital, Tianjing 300192, P.R. China
| | - Chengzhi Lu
- Department of Cardiology, Tianjin First Central Hospital, Tianjing 300192, P.R. China
| |
Collapse
|
18
|
Yang M, Li L, Chen S, Li S, Wang B, Zhang C, Chen Y, Yang L, Xin H, Chen C, Xu X, Zhang Q, He Y, Ye J. Melatonin protects against apoptosis of megakaryocytic cells via its receptors and the AKT/mitochondrial/caspase pathway. Aging (Albany NY) 2020; 12:13633-13646. [PMID: 32651992 PMCID: PMC7377846 DOI: 10.18632/aging.103483] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 05/27/2020] [Indexed: 04/19/2023]
Abstract
Clinical studies have shown that melatonin lowers the frequency of thrombocytopenia in patients with cancer undergoing radiotherapy or chemotherapy. Here, we investigated the mechanisms by which melatonin promotes platelet formation and survival. Our results show that melatonin exerted protective effects on serum-free induced apoptosis of CHRF megakaryocytes (MKs). Melatonin promoted the formation of MK colony forming units (CFUs) in a dose-dependent manner. Using doxorubicin-treated CHRF cells, we found that melatonin rescued G2/M cell cycle arrest and cell apoptosis induced by doxorubicin. The expression of p-AKT was increased by melatonin treatment, an effect that was abolished by melatonin receptor blocker. In addition, we demonstrated that melatonin enhanced the recovery of platelets in an irradiated mouse model. Megakaryopoiesis was largely preserved in melatonin-treated mice. We obtained the same results in vivo from bone marrow histology and CFU-MK formation assays. Melatonin may exert these protective effects by directly stimulating megakaryopoiesis and inhibiting megakaryocyte apoptosis through activation of its receptors and AKT signaling.
Collapse
Affiliation(s)
- Mo Yang
- The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, China
- Nanfang Hospital, Southern Medical University, Guangzhou, China
- Lianjiang People’s Hospital, Lianjiang, Guangdong, China
| | - Liang Li
- The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Shichao Chen
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Suyi Li
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Bo Wang
- The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Changhua Zhang
- The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, China
- The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Youpeng Chen
- The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Liuming Yang
- Lianjiang People’s Hospital, Lianjiang, Guangdong, China
| | - Hongwu Xin
- Lianjiang People’s Hospital, Lianjiang, Guangdong, China
| | - Chun Chen
- The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Xiaojun Xu
- The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Qing Zhang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Yulong He
- The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Jieyu Ye
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
19
|
Kayahara GM, Valente VB, Pereira RB, Lopes FYK, Crivelini MM, Miyahara GI, Biasoli ÉR, Oliveira SHP, Bernabé DG. Pineal gland protects against chemically induced oral carcinogenesis and inhibits tumor progression in rats. Oncotarget 2020; 11:1816-1831. [PMID: 32499868 PMCID: PMC7244010 DOI: 10.18632/oncotarget.27551] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 03/14/2020] [Indexed: 12/24/2022] Open
Abstract
Clinical investigations suggest that melatonin suppression and circadian dysfunction may be related to cancer development in shift workers. Studies also show that melatonin suppression after pinealectomy increases cancer incidence in preclinical models. However, no study evaluated the influence of pinealectomy on oral cancer development. In the current study, we investigated the effects of pinealectomy on oral squamous cell carcinoma (OSCC) occurrence and progression in rats. Rats submitted to sham surgery were used as control. Pinealectomy promoted an increase of 140% in OSCC occurrence when compared to sham animals. Tumors from pinealectomized rats displayed a higher volume and thickness than the tumors from sham-operated animals. Pinealectomy induced atrophy of the epithelium adjacent to the oral lesions. Pinealectomized rats showed higher mean number of tumor-associated macrophages and eosinophils in the invasive front of OSCC. In addition, nuclear overexpression of ERK1/2 and p53 was also observed in the front of carcinomas from pinealectomized rats. These results reveal that pineal gland plays a protective role against oral carcinogenesis. The melatonin suppression caused by the pinealectomy might contribute to oral cancer development by acting on ERK1/2 and p53 pathways and regulating tumor inflammation.
Collapse
Affiliation(s)
- Giseli Mitsuy Kayahara
- Psychoneuroimmunology Laboratory, Psychosomatic Research Center, Oral Oncology Center, São Paulo State University (Unesp), School of Dentistry, SP 15050-015, Araçatuba, São Paulo, Brazil.,Department of Diagnosis and Surgery, São Paulo State University (Unesp), School of Dentistry, SP 15050-015, Araçatuba, São Paulo, Brazil
| | - Vitor Bonetti Valente
- Psychoneuroimmunology Laboratory, Psychosomatic Research Center, Oral Oncology Center, São Paulo State University (Unesp), School of Dentistry, SP 15050-015, Araçatuba, São Paulo, Brazil
| | - Rosani Belzunces Pereira
- Psychoneuroimmunology Laboratory, Psychosomatic Research Center, Oral Oncology Center, São Paulo State University (Unesp), School of Dentistry, SP 15050-015, Araçatuba, São Paulo, Brazil
| | - Felipe Yudi Kabeya Lopes
- Psychoneuroimmunology Laboratory, Psychosomatic Research Center, Oral Oncology Center, São Paulo State University (Unesp), School of Dentistry, SP 15050-015, Araçatuba, São Paulo, Brazil
| | - Marcelo Macedo Crivelini
- Department of Diagnosis and Surgery, São Paulo State University (Unesp), School of Dentistry, SP 15050-015, Araçatuba, São Paulo, Brazil
| | - Glauco Issamu Miyahara
- Psychoneuroimmunology Laboratory, Psychosomatic Research Center, Oral Oncology Center, São Paulo State University (Unesp), School of Dentistry, SP 15050-015, Araçatuba, São Paulo, Brazil.,Department of Diagnosis and Surgery, São Paulo State University (Unesp), School of Dentistry, SP 15050-015, Araçatuba, São Paulo, Brazil
| | - Éder Ricardo Biasoli
- Psychoneuroimmunology Laboratory, Psychosomatic Research Center, Oral Oncology Center, São Paulo State University (Unesp), School of Dentistry, SP 15050-015, Araçatuba, São Paulo, Brazil.,Department of Diagnosis and Surgery, São Paulo State University (Unesp), School of Dentistry, SP 15050-015, Araçatuba, São Paulo, Brazil
| | - Sandra Helena Penha Oliveira
- Psychoneuroimmunology Laboratory, Psychosomatic Research Center, Oral Oncology Center, São Paulo State University (Unesp), School of Dentistry, SP 15050-015, Araçatuba, São Paulo, Brazil.,Laboratory of Immunopharmacology, Department of Basic Sciences, São Paulo State University (Unesp), School of Dentistry, SP 15050-015, Araçatuba, São Paulo, Brazil
| | - Daniel Galera Bernabé
- Psychoneuroimmunology Laboratory, Psychosomatic Research Center, Oral Oncology Center, São Paulo State University (Unesp), School of Dentistry, SP 15050-015, Araçatuba, São Paulo, Brazil.,Department of Diagnosis and Surgery, São Paulo State University (Unesp), School of Dentistry, SP 15050-015, Araçatuba, São Paulo, Brazil
| |
Collapse
|
20
|
Liu X, Zhang Y, Wu S, Xu M, Shen Y, Yu M, Fan J, Wei S, Xu C, Huang L, Zhao H, Li X, Ye X. Palmatine induces G2/M phase arrest and mitochondrial-associated pathway apoptosis in colon cancer cells by targeting AURKA. Biochem Pharmacol 2020; 175:113933. [PMID: 32224138 DOI: 10.1016/j.bcp.2020.113933] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 03/20/2020] [Indexed: 12/11/2022]
Abstract
Studies have shown that palmatine (PAL) has anti-cancer effects. However, the activity and potential mechanisms of PAL against colorectal cancer remain elusive. The results showed that PAL significantly inhibited the proliferation of colon cancer cells in vitro and in vivo without significant effect on non-tumorigenic colon cells. Target prediction and clinical sample database analysis suggested that PAL may contribute to colon cancer cells phase arrest and apoptosis by targeting aurora kinase A (AURKA). Inhibition and overexpression of AURKA proved that PAL induces G2/M phase arrest and apoptosis in colon cancer cells by targeting AURKA. Moreover, PAL promoted intracellular Reactive oxygen species (ROS) production and decreased mitochondrial membrane potential (ΔΨm). PAL reduced the levels of AURKA, Bcl-xl and Bcl2 proteins, and promoted the expression of pro-apoptotic proteins P53, P73, Caspase3 and Caspase9, as well as the increase of cytochrome c (cyt. c) in cell lysates in vitro and in vivo. Together, our study confirmed that PAL induced G2/M phase arrest and mitochondrial-associated pathway apoptosis in colon cancer cells by targeting AURKA. PAL may provide a novel solution for the treatment of colon cancer by serving as a new AURKA inhibitor.
Collapse
Affiliation(s)
- Xiaojiang Liu
- Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, Innovation, School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Yaru Zhang
- Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, Innovation, School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Siqi Wu
- Chongqing Productivity Promotion Center of Chinese Traditional Medicine, Modernization, School of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China
| | - Minmin Xu
- Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, Innovation, School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Youfeng Shen
- Chongqing Productivity Promotion Center of Chinese Traditional Medicine, Modernization, School of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China
| | - Min Yu
- Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, Innovation, School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Jinhua Fan
- Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, Innovation, School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Sijia Wei
- Chongqing Productivity Promotion Center of Chinese Traditional Medicine, Modernization, School of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China
| | - Chaohang Xu
- Chongqing Productivity Promotion Center of Chinese Traditional Medicine, Modernization, School of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China
| | - Lu Huang
- Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, Innovation, School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Han Zhao
- Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, Innovation, School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Xuegang Li
- Chongqing Productivity Promotion Center of Chinese Traditional Medicine, Modernization, School of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China.
| | - Xiaoli Ye
- Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, Innovation, School of Life Sciences, Southwest University, Chongqing 400715, China.
| |
Collapse
|
21
|
Tian Y, Song H, Jin D, Hu N, Sun L. MST1-Hippo pathway regulates inflammation response following myocardial infarction through inhibiting HO-1 signaling pathway. J Recept Signal Transduct Res 2020; 40:231-236. [PMID: 32054389 DOI: 10.1080/10799893.2020.1726954] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Context: Mammalian STE20-like protein kinases 1 (MST1) has been found to be associated with cardiomyocyte damage following acute myocardial infarction.Aim: The aim of our study is to explore the influence of MST1 in inflammation response following myocardial infarction.Methods: Cardiomyocyte cell line was used in vitro with hypoxia treatment to establish myocardial infarction model. ELISA, qPCR, Western blots, and siRNA technology were used to analyze the role of MST1 in inflammation response following myocardial infarction.Results: The transcription and expression of MST1 was significantly elevated following myocardial infarction. Loss of MST1 attenuated the levels of inflammation response and thus contributed to the survival of cardiomyocyte in vitro. Mechanistically, MST1 deletion reversed the activity of heme oxygenase-1 (HO-1) and thus reduced hypoxia-mediated cardiomyocyte death.Conclusions: Altogether, in this study, we found that MST1-Hippo pathway is activated in myocardial infarction and contributes to the inflammation response in cardiomyocytes through inhibiting the HO-1 signaling pathway. This finding would provide a potential target to reverse cardiomyocyte viability and reduce inflammation response in myocardial infarction.
Collapse
Affiliation(s)
- Yanan Tian
- Department of Cardiology, The Affiliated Hospital of Chengde Medical College, Chengde, P. R. China
| | - Haijiu Song
- The First Department of Medicine, Chengde City Hospital of traditional Chinese Medicine, Chengde, P. R. China
| | - Dapeng Jin
- Department of Cardiology, The Affiliated Hospital of Chengde Medical College, Chengde, P. R. China
| | - Na Hu
- Department of Cardiology, The Affiliated Hospital of Chengde Medical College, Chengde, P. R. China
| | - Lixian Sun
- Department of Cardiology, The Affiliated Hospital of Chengde Medical College, Chengde, P. R. China
| |
Collapse
|
22
|
Li P, Hu F, Cao X, Luo L, Tu Q. Melatonin receptor protects cardiomyocyte against oxidative stress-induced apoptosis through the MAPK-ERK signaling pathway. J Recept Signal Transduct Res 2020; 40:117-125. [PMID: 31986953 DOI: 10.1080/10799893.2020.1719151] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Peng Li
- Department of Gerontology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, People’s Republic of China
- Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, People’s Republic of China
| | - Fang Hu
- Department of Endocrinology and Metabolism, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, People’s Republic of China
| | - Xin Cao
- Department of Gerontology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, People’s Republic of China
| | - Liyun Luo
- Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, People’s Republic of China
- Department of Cardiology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, People’s Republic of China
| | - Qiuyun Tu
- Department of Gerontology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, People’s Republic of China
- Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, People’s Republic of China
| |
Collapse
|
23
|
Amin AH, Bughdadi FA, Abo-Zaid MA, Ismail AH, El-Agamy SA, Alqahtani A, El-Sayyad HIH, Rezk BM, Ramadan MF. Immunomodulatory effect of papaya (Carica papaya) pulp and seed extracts as a potential natural treatment for bacterial stress. J Food Biochem 2019; 43:e13050. [PMID: 31571245 DOI: 10.1111/jfbc.13050] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 08/06/2019] [Accepted: 09/04/2019] [Indexed: 12/29/2022]
Abstract
The current study evaluated the immunomodulatory effects of Carica papaya pulp and seeds methanol (MeOH) extracts on mice infected with Listeria monocytogenes. Gas chromatography-mass spectrometry analysis identified 10 active constituents in C. papaya seed MeOH extract and 10 compounds in C. papaya pulp MeOH extract. The experimental animals were divided into negative control (G1) group, positive control (G2) group, pulp extract treated (G3) group, and seed extract treated (G4) group. After infection of animals (G2, G3, and G4), treatments were started for 3 weeks. Estimation of the immunological parameters showed a marked decrease in IgM levels and an increase in IgG levels in the treated groups (G3 and G4) compared with those in G2. The proinflammatory cytokines (IL-10, IL-12, IL-1β, IL-6, and TGF-β1) were decreased in the treated groups (G3 and G4) compared with those in G2. Nitric oxide levels were also decreased, and the percentages of phagocytosis increased compared with those of G2. The results demonstrated the immunomodulatory and anti-inflammatory effects of C. papaya pulp and seeds MeOH extracts. PRACTICAL APPLICATIONS: Based on the antioxidant and antibacterial activities exhibited by the pulp and seed MeOH extracts investigated in this study, Carica papaya might be considered as a natural source of phytochemicals that could be utilized in novel foods and pharmaceuticals. Further investigation are needed to identify and purify compounds that might be responsible for the observed effects.
Collapse
Affiliation(s)
- Ali H Amin
- Deanship of Scientific Research, Umm Al-Qura University, Makkah, Saudi Arabia.,Zoology Department, Faculty of Science, Mansoura University, Mansoura, Egypt.,Department of Natural Sciences, Southern University at New Orleans, New Orleans, Louisiana
| | - Faisal A Bughdadi
- Biology Department, University College, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Mabrouk A Abo-Zaid
- Biology Department, Faculty of Science, Jazan university, Jazan, Saudi Arabia
| | - Ahmed H Ismail
- Biology Department, Faculty of Science, Jazan university, Jazan, Saudi Arabia
| | - Sherif A El-Agamy
- Biology Department, Faculty of Science, Jazan university, Jazan, Saudi Arabia
| | - Alaa Alqahtani
- Deanship of Scientific Research, Umm Al-Qura University, Makkah, Saudi Arabia.,College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | | | - Bashir Mahmoud Rezk
- Department of Natural Sciences, Southern University at New Orleans, New Orleans, Louisiana
| | - Mohamed Fawzy Ramadan
- Deanship of Scientific Research, Umm Al-Qura University, Makkah, Saudi Arabia.,Agricultural Biochemistry Department, Faculty of Agriculture, Zagazig University, Zagazig, Egypt
| |
Collapse
|