51
|
Yamagata K, Uzu E, Yoshigai Y, Kato C, Tagami M. Oleic acid and oleoylethanolamide decrease interferon-γ-induced expression of PD-L1 and induce apoptosis in human lung carcinoma cells. Eur J Pharmacol 2021; 903:174116. [PMID: 33957086 DOI: 10.1016/j.ejphar.2021.174116] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 04/16/2021] [Accepted: 04/20/2021] [Indexed: 01/10/2023]
Abstract
Inhibition of programmed death-ligand 1 (PD-L1) in cancer cells provides a reasonable avenue to prevent cancer progression. Although oleate is known to exert anti-cancer effects, its PD-L1 inhibitory effects have not been proven. This study investigated the effects of oleic acid and an oleic acid metabolite, oleoylethanolamide (OEA), on PD-L1 expression and biomarkers of tumorigenesis in several cancer cell lines, namely A549, HuH-7, MCF-7, DLD-1, and LoVo cells. Specifically, we analyzed the expression of PD-L1 and several apoptosis-related genes using RT-PCR. Interferon-gamma (IFN-γ)-induced modulation of PD-L1 protein expression was investigated using western blotting. Results indicate that IFN-γ stimulation increased the expression of PD-L1 in the chosen cancer cell lines. The IFN-γ-induced expression of PD-L1 was greater in A549 cells, than in other cancerous cell lines. In A549 cells, oleic acid and OEA decreased IFN-γ-induced expression of PD-L1, Bax, Bcl-2, and caspase 3. Oleic acid and OEA decreased IFN-γ-induced phosphorylation of STAT. These results indicate that oleic acid and OEA inhibit PD-1 expression, and induce apoptosis via STAT phosphorylation. Therefore, oleic acid and OEA may prevent cancer formation through STAT phosphorylation with IFN-γ. These findings provide novel insights into the anti-cancer effects of oleic acid-rich oil, such as olive oil.
Collapse
Affiliation(s)
- Kazuo Yamagata
- Department of Food Bioscience and Biotechnology, College of Bioresource Sciences, Nihon University (NUBS), Fujisawa, Japan.
| | - Erika Uzu
- Department of Food Bioscience and Biotechnology, College of Bioresource Sciences, Nihon University (NUBS), Fujisawa, Japan
| | - Yuri Yoshigai
- Department of Food Bioscience and Biotechnology, College of Bioresource Sciences, Nihon University (NUBS), Fujisawa, Japan
| | - Chihiro Kato
- Department of Food Bioscience and Biotechnology, College of Bioresource Sciences, Nihon University (NUBS), Fujisawa, Japan
| | - Motoki Tagami
- Department of Internal Medicine, Sanraku Hospital, Chiyoda-Ku, Tokyo, Japan
| |
Collapse
|
52
|
Giulitti F, Petrungaro S, Mandatori S, Tomaipitinca L, de Franchis V, D'Amore A, Filippini A, Gaudio E, Ziparo E, Giampietri C. Anti-tumor Effect of Oleic Acid in Hepatocellular Carcinoma Cell Lines via Autophagy Reduction. Front Cell Dev Biol 2021; 9:629182. [PMID: 33614661 PMCID: PMC7892977 DOI: 10.3389/fcell.2021.629182] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 01/14/2021] [Indexed: 12/11/2022] Open
Abstract
Oleic acid (OA) is a component of the olive oil. Beneficial health effects of olive oil are well-known, such as protection against liver steatosis and against some cancer types. In the present study, we focused on OA effects in hepatocellular carcinoma (HCC), investigating responses to OA treatment (50–300 μM) in HCC cell lines (Hep3B and Huh7.5) and in a healthy liver-derived human cell line (THLE-2). Upon OA administration higher lipid accumulation, perilipin-2 increase, and autophagy reduction were observed in HCC cells as compared to healthy cells. OA in the presence of 10% FBS significantly reduced viability of HCC cell lines at 300 μM through Alamar Blue staining evaluation, and reduced cyclin D1 expression in a dose-dependent manner while it was ineffective on healthy hepatocytes. Furthermore, OA increased cell death by about 30%, inducing apoptosis and necrosis in HCC cells but not in healthy hepatocytes at 300 μM dosage. Moreover, OA induced senescence in Hep3B, reduced P-ERK in both HCC cell lines and significantly inhibited the antiapoptotic proteins c-Flip and Bcl-2 in HCC cells but not in healthy hepatocytes. All these results led us to conclude that different cell death processes occur in these two HCC cell lines upon OA treatment. Furthermore, 300 μM OA significantly reduced the migration and invasion of both HCC cell lines, while it has no effects on healthy cells. Finally, we investigated autophagy role in OA-dependent effects by using the autophagy inducer torin-1. Combined OA/torin-1 treatment reduced lipid accumulation and cell death as compared to single OA treatment. We therefore concluded that OA effects in HCC cells lines are, at least, in part dependent on OA-induced autophagy reduction. In conclusion, we report for the first time an autophagy dependent relevant anti-cancer effect of OA in human hepatocellular carcinoma cell lines.
Collapse
Affiliation(s)
- Federico Giulitti
- Department of Anatomical, Histological, Forensic Medicine, and Orthopedic Sciences, Sapienza University of Rome, Rome, Italy
| | - Simonetta Petrungaro
- Department of Anatomical, Histological, Forensic Medicine, and Orthopedic Sciences, Sapienza University of Rome, Rome, Italy
| | - Sara Mandatori
- Department of Anatomical, Histological, Forensic Medicine, and Orthopedic Sciences, Sapienza University of Rome, Rome, Italy
| | - Luana Tomaipitinca
- Department of Anatomical, Histological, Forensic Medicine, and Orthopedic Sciences, Sapienza University of Rome, Rome, Italy
| | - Valerio de Franchis
- Department of Anatomical, Histological, Forensic Medicine, and Orthopedic Sciences, Sapienza University of Rome, Rome, Italy
| | - Antonella D'Amore
- Department of Anatomical, Histological, Forensic Medicine, and Orthopedic Sciences, Sapienza University of Rome, Rome, Italy
| | - Antonio Filippini
- Department of Anatomical, Histological, Forensic Medicine, and Orthopedic Sciences, Sapienza University of Rome, Rome, Italy
| | - Eugenio Gaudio
- Department of Anatomical, Histological, Forensic Medicine, and Orthopedic Sciences, Sapienza University of Rome, Rome, Italy
| | - Elio Ziparo
- Department of Anatomical, Histological, Forensic Medicine, and Orthopedic Sciences, Sapienza University of Rome, Rome, Italy
| | - Claudia Giampietri
- Department of Anatomical, Histological, Forensic Medicine, and Orthopedic Sciences, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
53
|
Saeed HK, Sutar Y, Patel P, Bhat R, Mallick S, Hatada AE, Koomoa DLT, Lange I, Date AA. Synthesis and Characterization of Lipophilic Salts of Metformin to Improve Its Repurposing for Cancer Therapy. ACS OMEGA 2021; 6:2626-2637. [PMID: 33553880 PMCID: PMC7859945 DOI: 10.1021/acsomega.0c04779] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 01/14/2021] [Indexed: 06/12/2023]
Abstract
Epidemiological evidence has accentuated the repurposing of metformin hydrochloride for cancer treatment. However, the extreme hydrophilicity and poor permeability of metformin hydrochloride are responsible for its poor anticancer activity in vitro and in vivo. Here, we report the synthesis and characterization of several lipophilic metformin salts containing bulky anionic permeation enhancers such as caprate, laurate, oleate, cholate, and docusate as counterions. Of various counterions tested, only docusate was able to significantly improve the lipophilicity and lipid solubility of metformin. To evaluate the impact of the association of anionic permeation enhancers with metformin, we checked the in vitro anticancer activity of various lipophilic salts of metformin using drug-sensitive (MYCN-2) and drug-resistant (SK-N-Be2c) neuroblastoma cells as model cancer cells. Metformin hydrochloride showed a very low potency (IC50 ≈ >100 mM) against MYCN-2 and SK-N-Be2c cells. Anionic permeation enhancers showed a considerably higher activity (IC50 ≈ 125 μM to 1.6 mM) against MYCN-2 and SK-N-Be2c cells than metformin. The association of metformin with most of the bulky anionic agents negatively impacted the anticancer activity against MYCN-2 and SK-N-Be2c cells. However, metformin docusate showed 700- to 4300-fold improvement in anticancer potency compared to metformin hydrochloride and four- to five-fold higher in vitro anticancer activity compared to sodium docusate, indicating a synergistic association between metformin and docusate. A similar trend was observed when we tested the in vitro activity of metformin docusate, sodium docusate, and metformin hydrochloride against hepatocellular carcinoma (HepG2) and triple-negative breast cancer (MDA-MB-231) cells.
Collapse
Affiliation(s)
- Hiwa K. Saeed
- Department
of Pharmaceutical Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawai’i at Hilo, Hilo Hawaii 96720, United States
| | - Yogesh Sutar
- Department
of Pharmaceutical Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawai’i at Hilo, Hilo Hawaii 96720, United States
| | - Pratikkumar Patel
- Department
of Pharmaceutical Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawai’i at Hilo, Hilo Hawaii 96720, United States
| | - Roopal Bhat
- Department
of Pharmaceutical Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawai’i at Hilo, Hilo Hawaii 96720, United States
- Department
of Pharmaceutics, Shree Chanakya Education
Society’s Indira College of Pharmacy, Tathawade, Pune, Maharashtra 411033, India
| | - Sudipta Mallick
- Department
of Pharmaceutical Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawai’i at Hilo, Hilo Hawaii 96720, United States
| | - Alyssa E. Hatada
- Department
of Pharmaceutical Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawai’i at Hilo, Hilo Hawaii 96720, United States
| | - Dana-Lynn T. Koomoa
- Department
of Pharmaceutical Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawai’i at Hilo, Hilo Hawaii 96720, United States
| | - Ingo Lange
- Department
of Pharmaceutical Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawai’i at Hilo, Hilo Hawaii 96720, United States
| | - Abhijit A. Date
- Department
of Pharmaceutical Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawai’i at Hilo, Hilo Hawaii 96720, United States
| |
Collapse
|
54
|
Pal A, Verma P, Paul S, Majumder I, Kundu R. Two species of Ulva inhibits the progression of cervical cancer cells SiHa by means of autophagic cell death induction. 3 Biotech 2021; 11:52. [PMID: 33489671 PMCID: PMC7801572 DOI: 10.1007/s13205-020-02576-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 12/01/2020] [Indexed: 12/24/2022] Open
Abstract
Edible green algal seaweeds, namely Ulva intestinalis and Ulva lactuca constitute a significant repository of popular herbal medicines in the Traditional Chinese Medicinal system. The present study aimed to assess the anticancer potential of these algal members and its mode of action in cervical cancer cells SiHa. The algal samples primarily extracted in methanol was fractionated into hexane, chloroform, and aqueous algal fractions, which inhibited the proliferation of SiHa cells in a dose-dependent manner, with the algal chloroform fractions harbouring the lowest IC50 dose of 141.38 µg/ml in U. intestinalis and 445.278 µg/ml in U. lactuca. These algal chloroform fractions when studied for their in-depth mode of action, were found to damage and pulverise the nuclei, resulting in a concomitant increase in subG0-phase of SiHa cells, studied by flow cytometry. The algal treatment also caused an increase in the number of acidic vesicles and enhanced the expression of LC3BII, p62 and atg12 proteins, which together pointed out autophagy as the induced mode of cell death. Upregulated Bax and p53 expression along with decreased Bcl2 expression also correlated to autophagic cell death. Decreased expression of E6 viral oncogene was noted as a significant response to algal fractions. Lastly, these potent algal fractions when characterised pharmacologically through GC-MS analysis were found to be rich in unsaturated fatty acids, majorly palmitic acid. Hence, this study concludes that the two species of Ulva successfully decreased the proliferation of SiHa cervical cancer cells through autophagy, hinting at palmitic acid being the major responsible bioactive compound in both.
Collapse
Affiliation(s)
- Asmita Pal
- Department of Botany, Centre of Advanced Studies, University of Calcutta, 35, Ballygunge Circular Road, Kolkata, 700019 West Bengal India
| | - Preeti Verma
- Department of Botany, Centre of Advanced Studies, University of Calcutta, 35, Ballygunge Circular Road, Kolkata, 700019 West Bengal India
| | - Subhabrata Paul
- Department of Botany, Centre of Advanced Studies, University of Calcutta, 35, Ballygunge Circular Road, Kolkata, 700019 West Bengal India
- Present Address: Bioprospecting Laboratory, School of Biotechnology, Presidency University, Kolkata, West Bengal India
| | - Indira Majumder
- Department of Botany, Centre of Advanced Studies, University of Calcutta, 35, Ballygunge Circular Road, Kolkata, 700019 West Bengal India
| | - Rita Kundu
- Department of Botany, Centre of Advanced Studies, University of Calcutta, 35, Ballygunge Circular Road, Kolkata, 700019 West Bengal India
| |
Collapse
|
55
|
Yang B, Yang N, Chen Y, Zhu M, Lian Y, Xiong Z, Wang B, Feng L, Jia X. An Integrated Strategy for Effective-Component Discovery of Astragali Radix in the Treatment of Lung Cancer. Front Pharmacol 2021; 11:580978. [PMID: 33628171 PMCID: PMC7898675 DOI: 10.3389/fphar.2020.580978] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 11/17/2020] [Indexed: 01/07/2023] Open
Abstract
Lung cancer is one of the most devastating diseases worldwide, with high incidence and mortality worldwide, and the anticancer potential of traditional Chinese medicine (TCM) has been gradually recognized by the scientific community. Astragali Radix (AR) is commonly used in traditional Chinese medicine in the treatment of lung cancer and has a certain clinical effect, but effective components and targets are still unclear. In the study, we established an integrated strategy for effective-component discovery of AR in the treatment of lung cancer based on a variety of techniques. First, the effective components and potential targets of AR were deciphered by the "component-target-disease" network using network pharmacology, and potential signal pathways on lung cancer were predicted by Gene Ontology (GO) biological function enrichment analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses. Then, the therapeutic effects of AR in the treatment of lung cancer were evaluated in vivo using A/J mice, and the potential targets related to autophagy and potential signal pathway were verified by Western blot analysis, immunofluorescence staining, and real-time PCR technology at protein and gene expression level. Finally, metabolism in vitro by rat intestinal flora and cell membrane immobilized chromatography technology were used to screen the effective components of AR in the treatment of lung cancer, and remaining components from the cell immobilized chromatography were collected and analyzed by ultra-performance liquid chromatography-electrospray quadrupole time-of-flight mass spectrometry (UPLC-Q-TOF-MS). The screening results of the integrated strategy showed that calycosin-7-O-β-D-glucoside, ononin, calycosin, astragaloside IV, astragaloside II, cycloastragenol, and formononetin may be effective components of AR in the treatment of lung cancer, and they may play a role in the treatment of lung cancer through autophagy and p53/AMPK/mTOR signaling pathway. The integrated strategy for effective-component discovery provided a valuable reference mode for finding the pharmacodynamic material basis of complex TCM systems. In addition, the prediction for targets and signal pathways laid a foundation for further study on the mechanism of AR in the treatment of lung cancer.
Collapse
Affiliation(s)
- Bing Yang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China,Nanjing University of Chinese Medicine, Nanjing, China,State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Nan Yang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yaping Chen
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China,State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Maomao Zhu
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China,State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Yuanpei Lian
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China,State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Zhiwei Xiong
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China,State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Bei Wang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China,State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Liang Feng
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China,State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China,*Correspondence: Liang Feng, ; Xiaobin Jia,
| | - Xiaobin Jia
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China,Nanjing University of Chinese Medicine, Nanjing, China,State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China,*Correspondence: Liang Feng, ; Xiaobin Jia,
| |
Collapse
|
56
|
Mosaddeghi P, Eslami M, Farahmandnejad M, Akhavein M, Ranjbarfarrokhi R, Khorraminejad-Shirazi M, Shahabinezhad F, Taghipour M, Dorvash M, Sakhteman A, Zarshenas MM, Nezafat N, Mobasheri M, Ghasemi Y. A systems pharmacology approach to identify the autophagy-inducing effects of Traditional Persian medicinal plants. Sci Rep 2021; 11:336. [PMID: 33431946 PMCID: PMC7801619 DOI: 10.1038/s41598-020-79472-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 12/09/2020] [Indexed: 01/29/2023] Open
Abstract
Aging is correlated with several complex diseases, including type 2 diabetes, neurodegeneration diseases, and cancer. Identifying the nature of this correlation and treatment of age-related diseases has been a major subject of both modern and traditional medicine. Traditional Persian Medicine (TPM) embodies many prescriptions for the treatment of ARDs. Given that autophagy plays a critical role in antiaging processes, the present study aimed to examine whether the documented effect of plants used in TPM might be relevant to the induction of autophagy? To this end, the TPM-based medicinal herbs used in the treatment of the ARDs were identified from modern and traditional references. The known phytochemicals of these plants were then examined against literature for evidence of having autophagy inducing effects. As a result, several plants were identified to have multiple active ingredients, which indeed regulate the autophagy or its upstream pathways. In addition, gene set enrichment analysis of the identified targets confirmed the collective contribution of the identified targets in autophagy regulating processes. Also, the protein-protein interaction (PPI) network of the targets was reconstructed. Network centrality analysis of the PPI network identified mTOR as the key network hub. Given the well-documented role of mTOR in inhibiting autophagy, our results hence support the hypothesis that the antiaging mechanism of TPM-based medicines might involve autophagy induction. Chemoinformatics study of the phytochemicals using docking and molecular dynamics simulation identified, among other compounds, the cyclo-trijuglone of Juglans regia L. as a potential ATP-competitive inhibitor of mTOR. Our results hence, provide a basis for the study of TPM-based prescriptions using modern tools in the quest for developing synergistic therapies for ARDs.
Collapse
Affiliation(s)
- Pouria Mosaddeghi
- grid.412571.40000 0000 8819 4698Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran ,grid.412571.40000 0000 8819 4698Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, P.O. Box 71345-1583, Shiraz, Iran ,grid.412571.40000 0000 8819 4698Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran ,grid.412571.40000 0000 8819 4698Cellular and Molecular Medicine Student Research Group, School of Medicine, Shiraz University of Medical Science, Shiraz, Iran
| | - Mahboobeh Eslami
- grid.412571.40000 0000 8819 4698Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran ,grid.412571.40000 0000 8819 4698Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, P.O. Box 71345-1583, Shiraz, Iran
| | - Mitra Farahmandnejad
- grid.412571.40000 0000 8819 4698Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran ,grid.412571.40000 0000 8819 4698Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, P.O. Box 71345-1583, Shiraz, Iran ,grid.412571.40000 0000 8819 4698Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran ,grid.412571.40000 0000 8819 4698Cellular and Molecular Medicine Student Research Group, School of Medicine, Shiraz University of Medical Science, Shiraz, Iran
| | - Mahshad Akhavein
- grid.412571.40000 0000 8819 4698Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran ,grid.412571.40000 0000 8819 4698Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, P.O. Box 71345-1583, Shiraz, Iran ,grid.412571.40000 0000 8819 4698Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran ,grid.412571.40000 0000 8819 4698Cellular and Molecular Medicine Student Research Group, School of Medicine, Shiraz University of Medical Science, Shiraz, Iran
| | - Ratin Ranjbarfarrokhi
- grid.412571.40000 0000 8819 4698Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran ,grid.412571.40000 0000 8819 4698Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, P.O. Box 71345-1583, Shiraz, Iran ,grid.412571.40000 0000 8819 4698Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran ,grid.412571.40000 0000 8819 4698Cellular and Molecular Medicine Student Research Group, School of Medicine, Shiraz University of Medical Science, Shiraz, Iran
| | - Mohammadhossein Khorraminejad-Shirazi
- grid.412571.40000 0000 8819 4698Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran ,grid.412571.40000 0000 8819 4698Cellular and Molecular Medicine Student Research Group, School of Medicine, Shiraz University of Medical Science, Shiraz, Iran
| | - Farbod Shahabinezhad
- grid.412571.40000 0000 8819 4698Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran ,grid.412571.40000 0000 8819 4698Cellular and Molecular Medicine Student Research Group, School of Medicine, Shiraz University of Medical Science, Shiraz, Iran
| | - Mohammadjavad Taghipour
- grid.412571.40000 0000 8819 4698Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran ,grid.412571.40000 0000 8819 4698Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, P.O. Box 71345-1583, Shiraz, Iran ,grid.412571.40000 0000 8819 4698Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran ,grid.412571.40000 0000 8819 4698Cellular and Molecular Medicine Student Research Group, School of Medicine, Shiraz University of Medical Science, Shiraz, Iran
| | - Mohammadreza Dorvash
- grid.412571.40000 0000 8819 4698Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran ,grid.412571.40000 0000 8819 4698Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran ,grid.412571.40000 0000 8819 4698Cellular and Molecular Medicine Student Research Group, School of Medicine, Shiraz University of Medical Science, Shiraz, Iran
| | - Amirhossein Sakhteman
- grid.412571.40000 0000 8819 4698Department of Medicinal Chemistry, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran ,grid.9668.10000 0001 0726 2490Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Mohammad M. Zarshenas
- grid.412571.40000 0000 8819 4698Department of Phytopharmaceuticals (Traditional Pharmacy), School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran ,grid.412571.40000 0000 8819 4698Medicinal Plants Processing Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Navid Nezafat
- grid.412571.40000 0000 8819 4698Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran ,grid.412571.40000 0000 8819 4698Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, P.O. Box 71345-1583, Shiraz, Iran
| | - Meysam Mobasheri
- grid.472338.9Department of Biotechnology, Faculty of Advanced Sciences and Technology, Tehran Islamic Azad University of Medical Sciences, Tehran, Iran ,Iranian Institute of New Sciences (IINS), Tehran, Iran
| | - Younes Ghasemi
- grid.412571.40000 0000 8819 4698Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran ,grid.412571.40000 0000 8819 4698Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, P.O. Box 71345-1583, Shiraz, Iran
| |
Collapse
|
57
|
Patra S, Bhol CS, Panigrahi DP, Praharaj PP, Pradhan B, Jena M, Bhutia SK. Gamma irradiation promotes chemo-sensitization potential of gallic acid through attenuation of autophagic flux to trigger apoptosis in an NRF2 inactivation signalling pathway. Free Radic Biol Med 2020; 160:111-124. [PMID: 32755671 DOI: 10.1016/j.freeradbiomed.2020.06.022] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 05/25/2020] [Accepted: 06/08/2020] [Indexed: 12/21/2022]
Abstract
Ionizing radiation has the potential to cause structural modification or change in electrochemical properties in parent lead pharmacophores that exhibit enhanced bioactivity. Gallic acid (GA), a triphenolic compound has displayed potent anticancer drug potency due to its withstanding antioxidant propensity. This study uncovered the comparative efficacy of gamma-irradiated gallic acid (GAIR) in the modulation of an antioxidant system for regulation apoptosis and autophagy. GAIR exhibited remarkable anti-proliferative efficacy as shown by MTT, clonogenic survival, and scratch assay. In addition to this, GAIR promoted intrinsic apoptosis through mitochondrial superoxide generation. GAIR decreased the activity of antioxidant enzymes by downregulating nuclear factor erythroid 2-related factor 2 (NRF2) and its downstream effector molecules NAD(P)H Quinone Dehydrogenase 1 (NQO1) and gamma-glutamylcysteine synthetase (GCLC). Simultaneously, GAIR attenuated autophagosome-lysosome fusion without altering the lysosomal activity. Inhibition of autophagic flux resulted in the accumulation of lipid droplets (LDs) such as hexadecanoic acid and oleic acid that fuelled superoxide generation leading to apoptosis. In the meantime, under oxidative upset, conversion of LDs to free fatty acids reduced leading to inhibition of ATP generation that subsequently provoked apoptosis. The effects of autophagy inhibition by GAIR on the therapeutic efficacy of chemotherapeutic drugs was studied and the co-treatment markedly decreased the cell viability and increased apoptosis. Further, GAIR exhibited potent antitumor activity in Dalton's Lymphoma-tumor bearing mice through modulation of apoptosis and autophagy without toxic activity. In conclusion, change in electrochemical properties by gamma radiation enhances the anticancer efficacy of gallic acid through superoxide mediated apoptosis fuelled by inhibition of lipophagy in an NRF2 dependent signaling pathway.
Collapse
Affiliation(s)
- Srimanta Patra
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, India
| | - Chandra Sekhar Bhol
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, India
| | | | | | - Biswajita Pradhan
- PG Department of Botany, Berhampur University, Berhampur, 760007, India
| | - Mrutyunjay Jena
- PG Department of Botany, Berhampur University, Berhampur, 760007, India
| | - Sujit Kumar Bhutia
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, India.
| |
Collapse
|
58
|
Yoon BK, Lim ZY, Jeon WY, Cho NJ, Kim JH, Jackman JA. Medicinal Activities and Nanomedicine Delivery Strategies for Brucea javanica Oil and Its Molecular Components. Molecules 2020; 25:E5414. [PMID: 33228061 PMCID: PMC7699344 DOI: 10.3390/molecules25225414] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 11/11/2020] [Accepted: 11/16/2020] [Indexed: 12/16/2022] Open
Abstract
Brucea javanica oil (BJO) is widely used in traditional Chinese medicine to treat various types of cancer and inflammatory diseases. There is significant interest in understanding the medicinal activities of BJO and its molecular components, especially quassinoids, and in exploring how they can be incorporated into nanomedicine delivery strategies for improved application prospects. Herein, we cover the latest progress in developing different classes of drug delivery vehicles, including nanoemulsions, liposomes, nanostructured lipid carriers, and spongosomes, to encapsulate BJO and purified quassinoids. An introduction to the composition and medicinal activities of BJO and its molecular components, including quassinoids and fatty acids, is first provided. Application examples involving each type of drug delivery vehicle are then critically presented. Future opportunities for nanomedicine delivery strategies in the field are also discussed and considered within the context of translational medicine needs and drug development processes.
Collapse
Affiliation(s)
- Bo Kyeong Yoon
- School of Chemical Engineering and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Korea; (B.K.Y.); (Z.Y.L.); (W.-Y.J.)
| | - Zheng Yi Lim
- School of Chemical Engineering and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Korea; (B.K.Y.); (Z.Y.L.); (W.-Y.J.)
- School of Materials Science and Engineering, Nanyang Technological University, Singapore 637553, Singapore;
| | - Won-Yong Jeon
- School of Chemical Engineering and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Korea; (B.K.Y.); (Z.Y.L.); (W.-Y.J.)
- Omni Colab Corporation, Suwon 16229, Korea
| | - Nam-Joon Cho
- School of Materials Science and Engineering, Nanyang Technological University, Singapore 637553, Singapore;
| | - Jeong Hoon Kim
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul 06351, Korea;
| | - Joshua A. Jackman
- School of Chemical Engineering and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Korea; (B.K.Y.); (Z.Y.L.); (W.-Y.J.)
| |
Collapse
|
59
|
Chen FX, Du N, Hu J, Ning F, Mei X, Li Q, Peng L. Intramuscular accumulation of pentadecanoic acid activates AKT1 to phosphorylate NCOR1 and triggers FOXM1-mediated apoptosis in the pathogenesis of sarcopenia. Am J Transl Res 2020; 12:5064-5079. [PMID: 33042406 PMCID: PMC7540105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 08/23/2020] [Indexed: 06/11/2023]
Abstract
Sarcopenia is an age-associated disorder that results in skeletal muscle loss. Apoptosis and inflammation are the two major contributors to sarcopenia. Emerging evidence has shown that long-chain fatty acids (LCFAs) are implicated in the muscles of sarcopenic animal models. However, it is unknown whether LCFAs are correlated with apoptosis or inflammation in the pathogenesis of sarcopenia. Herein, we found that pentadecanoic acid (PDA), a C15 LCFA, was significantly accumulated in human sarcopenic muscles. In vitro PDA treatment could dose-dependently induce the expression of the transcription factor FOXM1 (forkhead box M1) and several proapoptotic genes, such as PUMA (p53-upregulated modulator of apoptosis), BAX (B-cell/lymphoma 2-associated X) and APAF1 (apoptotic peptidase activating factor 1), thereby causing apoptosis. Mechanically, PDA activated AKT1 (AKT serine/threonine kinase 1) to phosphorylate NCOR1 (nuclear receptor corepressor 1). The phosphorylated NCOR1 disassociated from the NCOR1-FOXM1 transcriptional complex and could not repress FOXM1-mediated transcription, leading to the induction of PUMA. The activated PUMA further triggered downstream apoptotic signaling, including activation of the BAX, APAF1 and caspase cascades, leading to the occurrence of apoptosis. Alkaline phosphatase or knockdown of AKT1 in vitro reversed the FOXM1-mediated apoptotic signaling. Collectively, our results provide new evidence that LCFAs are involved in the pathogenesis of sarcopenia by activating apoptotic signaling. Attempts to decrease the intake of PDA-containing foods or blocking AKT1 may improve the symptoms of sarcopenia.
Collapse
Affiliation(s)
- Fa-Xiu Chen
- Department of Geriatrics, Jiangxi Provincial People's Hospital Affiliated to Nanchang University Nanchang 330006, Jiangxi, China
| | - Ning Du
- Department of Geriatrics, Jiangxi Provincial People's Hospital Affiliated to Nanchang University Nanchang 330006, Jiangxi, China
| | - Jian Hu
- Department of Geriatrics, Jiangxi Provincial People's Hospital Affiliated to Nanchang University Nanchang 330006, Jiangxi, China
| | - Fang Ning
- Department of Geriatrics, Jiangxi Provincial People's Hospital Affiliated to Nanchang University Nanchang 330006, Jiangxi, China
| | - Xun Mei
- Department of Geriatrics, Jiangxi Provincial People's Hospital Affiliated to Nanchang University Nanchang 330006, Jiangxi, China
| | - Qiang Li
- Department of Geriatrics, Jiangxi Provincial People's Hospital Affiliated to Nanchang University Nanchang 330006, Jiangxi, China
| | - Le Peng
- Department of Geriatrics, Jiangxi Provincial People's Hospital Affiliated to Nanchang University Nanchang 330006, Jiangxi, China
| |
Collapse
|
60
|
Kostrhunova H, Zajac J, Markova L, Brabec V, Kasparkova J. A Multi-action Pt IV Conjugate with Oleate and Cinnamate Ligands Targets Human Epithelial Growth Factor Receptor HER2 in Aggressive Breast Cancer Cells. Angew Chem Int Ed Engl 2020; 59:21157-21162. [PMID: 32750194 DOI: 10.1002/anie.202009491] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Indexed: 01/07/2023]
Abstract
HER2-positive breast cancer is an aggressive subtype that typically responds poorly to standard chemotherapy. To design an anticancer drug selective for HER2-expressing breast cancer, a PtIV prodrug with axial oleate and cinnamate ligands was synthesized. We demonstrate its superior antiproliferative activity in monolayer and 3D spheroid models; the antiproliferative efficiency increases gradually with increasing expression of HER2. The results also suggest that the released PtII compound inhibits the proliferation of cancer cells by a DNA-damage-mediated mechanism. Simultaneously, the released oleic and cinnamic acid can effectively inhibit HER2 expression. To our knowledge, this is the first platinum-based complex inhibiting HER2 expression that does not contain protein or peptide. Moreover, this PtIV prodrug is capable of overcoming the resistance of cancer stem cells (CSCs), inducing death in both CSCs and differentiated cancer cells. Thus, the results substantiate our design strategy and demonstrate the potential of this approach for the development of new, therapeutically relevant compounds.
Collapse
Affiliation(s)
- Hana Kostrhunova
- Czech Academy of Sciences, Institute of Biophysics, Kralovopolska 135, 61265, Brno, Czech Republic
| | - Juraj Zajac
- Czech Academy of Sciences, Institute of Biophysics, Kralovopolska 135, 61265, Brno, Czech Republic
| | - Lenka Markova
- Czech Academy of Sciences, Institute of Biophysics, Kralovopolska 135, 61265, Brno, Czech Republic
| | - Viktor Brabec
- Czech Academy of Sciences, Institute of Biophysics, Kralovopolska 135, 61265, Brno, Czech Republic
| | - Jana Kasparkova
- Czech Academy of Sciences, Institute of Biophysics, Kralovopolska 135, 61265, Brno, Czech Republic
| |
Collapse
|
61
|
Kostrhunova H, Zajac J, Markova L, Brabec V, Kasparkova J. A Multi‐action Pt
IV
Conjugate with Oleate and Cinnamate Ligands Targets Human Epithelial Growth Factor Receptor HER2 in Aggressive Breast Cancer Cells. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202009491] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Hana Kostrhunova
- Czech Academy of Sciences Institute of Biophysics Kralovopolska 135 61265 Brno Czech Republic
| | - Juraj Zajac
- Czech Academy of Sciences Institute of Biophysics Kralovopolska 135 61265 Brno Czech Republic
| | - Lenka Markova
- Czech Academy of Sciences Institute of Biophysics Kralovopolska 135 61265 Brno Czech Republic
| | - Viktor Brabec
- Czech Academy of Sciences Institute of Biophysics Kralovopolska 135 61265 Brno Czech Republic
| | - Jana Kasparkova
- Czech Academy of Sciences Institute of Biophysics Kralovopolska 135 61265 Brno Czech Republic
| |
Collapse
|
62
|
Nanoemulsion-based systems as a promising approach for enhancing the antitumoral activity of pequi oil (Caryocar brasilense Cambess.) in breast cancer cells. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2020.101819] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
63
|
The novel therapeutic potential of bovine α-lactalbumin made lethal to tumour cells (BALMET) and oleic acid in oral squamous cell carcinoma (OSCC). Eur J Cancer Prev 2020; 30:178-187. [PMID: 32694279 DOI: 10.1097/cej.0000000000000617] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Since the serendipitous discovery of bovine α-lactalbumin made lethal to tumour cells (BAMLET)/human α-lactalbumin made lethal to tumour cells there has been an increased interest in the ability of the two components, oleic acid and α-lactalbumin, to form anti-cancer complexes. Here we have investigated the in-vitro efficacy of the BAMLET complex in killing oral cancer (OC) cells, determined the active component of the complex and investigated possible biological mechanisms. MATERIALS AND METHODS Two OC cell lines (±p53 mutation) and one dysplastic cell line were used as a model of progressive oral carcinogenesis. We performed cell viability assays with increasing BAMLET concentrations to determine the cytotoxic potential of the complex. We further analysed the individual components to determine their respective cytotoxicities. siRNA knockdown of p53 was used to determine its functional role in mediating sensitivity to BAMLET. Cell death mechanisms were investigated by flow cytometry, confocal microscopy and the lactate dehydrogenase assay. RESULTS Our results show that BAMLET is cytotoxic to the OC and dysplastic cell lines in a time and dose-dependent manner. The cytotoxic component was found to be oleic acid, which, can induce cytotoxicity even when not in complex. Our results indicate that the mechanism of cytotoxicity occurs through multiple simultaneous events including cell cycle arrest, autophagy like processes with a minor involvement of necrosis. CONCLUSION Deciphering the mechanism of cytotoxicity will aid treatment modalities for OC. This study highlights the potential of BAMLET as a novel therapeutic strategy in oral dysplastic and cancerous cells.
Collapse
|
64
|
Sawyer BT, Qamar L, Yamamoto TM, McMellen A, Watson ZL, Richer JK, Behbakht K, Schlaepfer IR, Bitler BG. Targeting Fatty Acid Oxidation to Promote Anoikis and Inhibit Ovarian Cancer Progression. Mol Cancer Res 2020; 18:1088-1098. [PMID: 32198139 PMCID: PMC7335321 DOI: 10.1158/1541-7786.mcr-19-1057] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 02/10/2020] [Accepted: 03/16/2020] [Indexed: 12/22/2022]
Abstract
Epithelial-derived high-grade serous ovarian cancer (HGSOC) is the deadliest gynecologic malignancy. Roughly 80% of patients are diagnosed with late-stage disease, which is defined by wide-spread cancer dissemination throughout the pelvic and peritoneal cavities. HGSOC dissemination is dependent on tumor cells acquiring the ability to resist anoikis (apoptosis triggered by cell detachment). Epithelial cell detachment from the underlying basement membrane or extracellular matrix leads to cellular stress, including nutrient deprivation. In this report, we examined the contribution of fatty acid oxidation (FAO) in supporting anoikis resistance. We examined expression Carnitine Palmitoyltransferase 1A (CPT1A) in a panel of HGSOC cell lines cultured in adherent and suspension conditions. With CPT1A knockdown cells, we evaluated anoikis by caspase 3/7 activity, cleaved caspase 3 immunofluorescence, flow cytometry, and colony formation. We assessed CPT1A-dependent mitochondrial activity and tested the effect of exogenous oleic acid on anoikis and mitochondrial activity. In a patient-derived xenograft model, we administered etomoxir, an FAO inhibitor, and/or platinum-based chemotherapy. CPT1A is overexpressed in HGSOC, correlates with poor overall survival, and is upregulated in HGSOC cells cultured in suspension. CPT1A knockdown promoted anoikis and reduced viability of cells cultured in suspension. HGSOC cells in suspension culture are dependent on CPT1A for mitochondrial activity. In a patient-derived xenograft model of HGSOC, etomoxir significantly inhibited tumor progression. IMPLICATIONS: Targeting FAO in HGSOC to promote anoikis and attenuate dissemination is a potential approach to promote a more durable antitumor response and improve patient outcomes.
Collapse
Affiliation(s)
- Brandon T Sawyer
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, Colorado
| | - Lubna Qamar
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, Colorado
| | - Tomomi M Yamamoto
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, Colorado
| | - Alexandra McMellen
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, Colorado
| | - Zachary L Watson
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, Colorado
| | - Jennifer K Richer
- Department of Pathology, University of Colorado School of Medicine, Aurora, Colorado
| | - Kian Behbakht
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, Colorado
| | - Isabel R Schlaepfer
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado.
| | - Benjamin G Bitler
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, Colorado.
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, Colorado
| |
Collapse
|
65
|
Inguglia L, Chiaramonte M, Di Stefano V, Schillaci D, Cammilleri G, Pantano L, Mauro M, Vazzana M, Ferrantelli V, Nicolosi R, Arizza V. Salmo salar fish waste oil: Fatty acids composition and antibacterial activity. PeerJ 2020; 8:e9299. [PMID: 32596043 PMCID: PMC7307567 DOI: 10.7717/peerj.9299] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 05/14/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND AND AIMS Fish by-products are generally used to produce fishmeal or fertilizers, with fish oil as a by-product. Despite their importance, fish wastes are still poorly explored and characterized and more studies are needed to reveal their potentiality. The goal of the present study was to qualitatively characterize and investigate the antimicrobial effects of the fish oil extracted from Salmo salar waste samples and to evaluate the potential use of these compounds for treating pathogen infections. METHODS Salmo salar waste samples were divided in two groups: heads and soft tissues. Fatty acids composition, and in particular the content in saturated (SAFAs), mono-unsaturated (MUFAs) and Polyunsaturated (PUFAs) fatty acids, was characterized through GC/MS Thermo Focus GC-DSQ II equipped with a ZB-5 fused silica capillary tubes column. The antimicrobial activity of the salmon waste oils was evaluated through the Minimum Inhibitory Concentration assay and the antibiotics contamination was determined by Liquid Chromatography with tandem Mass Spectrometry (LC-MS/MS) analysis. All experiments were done at least in triplicate. RESULTS GC/MS analysis has shown the specific fatty acid composition of the salmon waste oils and their enrichment in MUFAs and PUFAs, with special reference to omega-3, -6, -7, -9 fatty acids. Furthermore, our study has highlighted the antimicrobial activity of the fish waste oil samples against two Gram+ and Gram- bacterial strains. CONCLUSIONS These data confirm that the fish waste is still quantitatively and qualitatively an important source of available biological properties that could be extracted and utilized representing an important strategy to counteract infective diseases in the context of the circular economy.
Collapse
Affiliation(s)
| | | | | | | | | | - Licia Pantano
- Istituto Zooprofilattico della Sicilia “A.Mirri”, Palermo, Italy, Italy
| | - Manuela Mauro
- STEBICEF, University of Palermo, Palermo, Italy, Italy
| | | | | | | | | |
Collapse
|
66
|
Bojková B, Winklewski PJ, Wszedybyl-Winklewska M. Dietary Fat and Cancer-Which Is Good, Which Is Bad, and the Body of Evidence. Int J Mol Sci 2020; 21:ijms21114114. [PMID: 32526973 PMCID: PMC7312362 DOI: 10.3390/ijms21114114] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/27/2020] [Accepted: 06/02/2020] [Indexed: 12/13/2022] Open
Abstract
A high-fat diet (HFD) induces changes in gut microbiota leading to activation of pro-inflammatory pathways, and obesity, as a consequence of overnutrition, exacerbates inflammation, a known risk factor not only for cancer. However, experimental data showed that the composition of dietary fat has a greater impact on the pathogenesis of cancer than the total fat content in isocaloric diets. Similarly, human studies did not prove that a decrease in total fat intake is an effective strategy to combat cancer. Saturated fat has long been considered as harmful, but the current consensus is that moderate intake of saturated fatty acids (SFAs), including palmitic acid (PA), does not pose a health risk within a balanced diet. In regard to monounsaturated fat, plant sources are recommended. The consumption of plant monounsaturated fatty acids (MUFAs), particularly from olive oil, has been associated with lower cancer risk. Similarly, the replacement of animal MUFAs with plant MUFAs decreased cancer mortality. The impact of polyunsaturated fatty acids (PUFAs) on cancer risk depends on the ratio between ω-6 and ω-3 PUFAs. In vivo data showed stimulatory effects of ω-6 PUFAs on tumour growth while ω-3 PUFAs were protective, but the results of human studies were not as promising as indicated in preclinical reports. As for trans FAs (TFAs), experimental data mostly showed opposite effects of industrially produced and natural TFAs, with the latter being protective against cancer progression, but human data are mixed, and no clear conclusion can be made. Further studies are warranted to establish the role of FAs in the control of cell growth in order to find an effective strategy for cancer prevention/treatment.
Collapse
Affiliation(s)
- Bianka Bojková
- Department of Animal Physiology, Institute of Biology and Ecology, Faculty of Science, P.J. Šafárik University in Košice, 041 54 Košice, Slovakia;
| | - Pawel J. Winklewski
- Department of Human Physiology, Medical University of Gdansk, 80-210 Gdansk, Poland;
- Department of Anatomy and Physiology, Pomeranian University of Slupsk, 76-200 Slupsk, Poland
- Correspondence: ; Tel./Fax: +48-58-3491515
| | | |
Collapse
|
67
|
Chhabra R, Nanjundan M. Lysophosphatidic acid reverses Temsirolimus-induced changes in lipid droplets and mitochondrial networks in renal cancer cells. PLoS One 2020; 15:e0233887. [PMID: 32492043 PMCID: PMC7269261 DOI: 10.1371/journal.pone.0233887] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 05/14/2020] [Indexed: 01/05/2023] Open
Abstract
Increased cytoplasmic lipid droplets (LDs) and elevated AKT/mTOR signaling are characteristics of clear cell renal cell carcinoma (ccRCC). Lysophosphatidic acid (LPA), a potent lipid mitogen generated via autotaxin (elevated in ccRCC), can modulate tumor progression but its role in altering chemotherapeutic sensitivity to mTOR inhibitors is unclear and thus is the focus of the studies presented herein. Using malignant (A-498, 769-P and 786-O) and normal immortalized kidney (HK-2) cell lines, we investigated their cellular responsiveness to Temsirolimus (TEMS, mTOR inhibitor) in the absence or presence of LPA by monitoring alterations in AKT/mTOR pathway mediators (via western blotting), LDs (using LipidTOX and real-time PCR to assess transcript changes in modulators of LD biogenesis/turnover), mitochondrial networks (via immunofluorescence staining for TOM20 and TOM70), as well as cellular viability. We identified that TEMS reduced cellular viability in all renal cell lines, with increased sensitivity in the presence of an autophagy inhibitor. TEMS also altered activation of AKT/mTOR pathway mediators, abundance of LDs, and fragmentation of mitochondrial networks. We observed that these effects were antagonized by LPA. In HK-2 cells, LPA markedly increased LD size and abundance, coinciding with phospho-MAPK and phospho-S6 activation, increased diacylglycerol O-acetyltransferase 2 (DGAT2) mRNA (which produces triacylglycerides), and survival. Inhibiting MAPK partially antagonized LPA-induced LD changes. Collectively, we have identified that LPA can reverse the effects of TEMS by increasing LDs in a MAPK-dependent manner; these results suggest that LPA may contribute to the pathogenesis and chemotherapeutic resistance of ccRCC.
Collapse
Affiliation(s)
- Ravneet Chhabra
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, Florida, United States of America
| | - Meera Nanjundan
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, Florida, United States of America
- * E-mail:
| |
Collapse
|
68
|
Pentadecanoic Acid, an Odd-Chain Fatty Acid, Suppresses the Stemness of MCF-7/SC Human Breast Cancer Stem-Like Cells through JAK2/STAT3 Signaling. Nutrients 2020; 12:nu12061663. [PMID: 32503225 PMCID: PMC7352840 DOI: 10.3390/nu12061663] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/22/2020] [Accepted: 06/02/2020] [Indexed: 02/07/2023] Open
Abstract
Saturated fatty acids possess few health benefits compared to unsaturated fatty acids. However, increasing experimental evidence demonstrates the nutritionally beneficial role of odd-chain saturated fatty acids in human health. In this study, the anti-cancer effects of pentadecanoic acid were evaluated in human breast carcinoma MCF-7/stem-like cells (SC), a cell line with greater mobility, invasiveness, and cancer stem cell properties compared to the parental MCF-7 cells. Pentadecanoic acid exerted selective cytotoxic effects in MCF-7/SC compared to in the parental cells. Moreover, pentadecanoic acid reduced the stemness of MCF-7/SC and suppressed the migratory and invasive ability of MCF-7/SC as evidenced by the results of flow cytometry, a mammosphere formation assay, an aldehyde dehydrogenase activity assay, and Western blot experiments conducted to analyze the expression of cancer stem cell markers—CD44, β-catenin, MDR1, and MRP1—and epithelial–mesenchymal transition (EMT) markers—snail, slug, MMP9, and MMP2. In addition, pentadecanoic acid suppressed interleukin-6 (IL-6)-induced JAK2/STAT3 signaling, induced cell cycle arrest at the sub-G1 phase, and promoted caspase-dependent apoptosis in MCF-7/SC. These findings indicate that pentadecanoic acid can serve as a novel JAK2/STAT3 signaling inhibitor in breast cancer cells and suggest the beneficial effects of pentadecanoic acid-rich food intake during breast cancer treatments.
Collapse
|
69
|
Harsha C, Banik K, Ang HL, Girisa S, Vikkurthi R, Parama D, Rana V, Shabnam B, Khatoon E, Kumar AP, Kunnumakkara AB. Targeting AKT/mTOR in Oral Cancer: Mechanisms and Advances in Clinical Trials. Int J Mol Sci 2020; 21:ijms21093285. [PMID: 32384682 PMCID: PMC7246494 DOI: 10.3390/ijms21093285] [Citation(s) in RCA: 124] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/02/2020] [Accepted: 05/03/2020] [Indexed: 12/18/2022] Open
Abstract
Oral cancer (OC) is a devastating disease that takes the lives of lots of people globally every year. The current spectrum of treatment modalities does not meet the needs of the patients. The disease heterogeneity demands personalized medicine or targeted therapies. Therefore, there is an urgent need to identify potential targets for the treatment of OC. Abundant evidence has suggested that the components of the protein kinase B (AKT)/ mammalian target of rapamycin (mTOR) pathway are intrinsic factors for carcinogenesis. The AKT protein is central to the proliferation and survival of normal and cancer cells, and its downstream protein, mTOR, also plays an indispensable role in the cellular processes. The wide involvement of the AKT/mTOR pathway has been noted in oral squamous cell carcinoma (OSCC). This axis significantly regulates the various hallmarks of cancer, like proliferation, survival, angiogenesis, invasion, metastasis, autophagy, and epithelial-to-mesenchymal transition (EMT). Activated AKT/mTOR signaling is also associated with circadian signaling, chemoresistance and radio-resistance in OC cells. Several miRNAs, circRNAs and lncRNAs also modulate this pathway. The association of this axis with the process of tumorigenesis has culminated in the identification of its specific inhibitors for the prevention and treatment of OC. In this review, we discussed the significance of AKT/mTOR signaling in OC and its potential as a therapeutic target for the management of OC. This article also provided an update on several AKT/mTOR inhibitors that emerged as promising candidates for therapeutic interventions against OC/head and neck cancer (HNC) in clinical studies.
Collapse
Affiliation(s)
- Choudhary Harsha
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India; (C.H.); (K.B.); (S.G.); (R.V.); (D.P.); (V.R.); (B.S.); (E.K.)
| | - Kishore Banik
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India; (C.H.); (K.B.); (S.G.); (R.V.); (D.P.); (V.R.); (B.S.); (E.K.)
| | - Hui Li Ang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore;
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
| | - Sosmitha Girisa
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India; (C.H.); (K.B.); (S.G.); (R.V.); (D.P.); (V.R.); (B.S.); (E.K.)
| | - Rajesh Vikkurthi
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India; (C.H.); (K.B.); (S.G.); (R.V.); (D.P.); (V.R.); (B.S.); (E.K.)
| | - Dey Parama
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India; (C.H.); (K.B.); (S.G.); (R.V.); (D.P.); (V.R.); (B.S.); (E.K.)
| | - Varsha Rana
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India; (C.H.); (K.B.); (S.G.); (R.V.); (D.P.); (V.R.); (B.S.); (E.K.)
| | - Bano Shabnam
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India; (C.H.); (K.B.); (S.G.); (R.V.); (D.P.); (V.R.); (B.S.); (E.K.)
| | - Elina Khatoon
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India; (C.H.); (K.B.); (S.G.); (R.V.); (D.P.); (V.R.); (B.S.); (E.K.)
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore;
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
- Correspondence: (A.P.K.); (A.B.K.); Tel.: +65-6516-5456 (A.P.K.); +91-361-258-2231 (A.B.K.); Fax: +65-6873-9664 (A.P.K.); +91-361-258-2249 (A.B.K.)
| | - Ajaikumar B. Kunnumakkara
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India; (C.H.); (K.B.); (S.G.); (R.V.); (D.P.); (V.R.); (B.S.); (E.K.)
- Correspondence: (A.P.K.); (A.B.K.); Tel.: +65-6516-5456 (A.P.K.); +91-361-258-2231 (A.B.K.); Fax: +65-6873-9664 (A.P.K.); +91-361-258-2249 (A.B.K.)
| |
Collapse
|
70
|
Protective Mechanisms of Avocado Oil Extract Against Ototoxicity. Nutrients 2020; 12:nu12040947. [PMID: 32235401 PMCID: PMC7230542 DOI: 10.3390/nu12040947] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 03/24/2020] [Accepted: 03/25/2020] [Indexed: 02/07/2023] Open
Abstract
Despite the excellent antimicrobial activity of aminoglycoside antibiotics, permanent inner ear damage associated with the use of these drugs has resulted in the need to develop strategies to address the ototoxic risk given their widespread use. In a previous study, we showed that avocado oil protects ear hair cells from damage caused by neomycin. However, the detailed mechanism by which this protection occurs is still unclear. Here, we investigated the auditory cell-protective mechanism of enhanced functional avocado oil extract (DKB122). RNA sequencing followed by pathway analysis revealed that DKB122 has the potential to enhance the expression of detoxification and antioxidant genes associated with glutathione metabolism (Hmox4, Gsta4, Mgst1, and Abcc3) in HEI-OC1 cells. Additionally, DKB122 effectively decreased ROS levels, resulting in the inhibition of apoptosis in HEI-OC1 cells. The expression of the inflammatory genes that encode chemokines and interleukins was also downregulated by DKB122 treatment. Consistent with these results, DKB122 significantly inhibited p65 nuclear migration induced by TNF-α or LPS in HEI-OC1 cells and THP-1 cells and the expression of inflammatory chemokine and interleukin genes induced by TNF-α was significantly reduced. Moreover, DKB122 treatment increased LC3-II and decreased p62 in HEI-OC1 cells, suggesting that DKB122 increases autophagic flux. These results suggest that DKB122 has otoprotective effects attributable to its antioxidant activity, induction of antioxidant gene expression, anti-inflammatory activity, and autophagy activation.
Collapse
|
71
|
Li W, Ren L, Zheng X, Liu J, Wang J, Ji T, Du G. 3- O-Acetyl-11-keto- β -boswellic acid ameliorated aberrant metabolic landscape and inhibited autophagy in glioblastoma. Acta Pharm Sin B 2020; 10:301-312. [PMID: 32082975 PMCID: PMC7016292 DOI: 10.1016/j.apsb.2019.12.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 11/02/2019] [Accepted: 11/25/2019] [Indexed: 02/08/2023] Open
Abstract
Glioblastoma is the most common and aggressive primary tumor in the central nervous system, accounting for 12%-15% of all brain tumors. 3-O-Acetyl-11-keto-β-boswellic acid (AKBA), one of the most active ingredients of gum resin from Boswellia carteri Birdw., was reported to inhibit the growth of glioblastoma cells and subcutaneous glioblastoma. However, whether AKBA has antitumor effects on orthotopic glioblastoma and the underlying mechanisms are still unclear. An orthotopic mouse model was used to evaluate the anti-glioblastoma effects of AKBA. The effects of AKBA on tumor growth were evaluated using MRI. The effects on the alteration of metabolic landscape were detected by MALDI-MSI. The underlying mechanisms of autophagy reducing by AKBA treatment were determined by immunoblotting and immunofluorescence, respectively. Transmission electron microscope was used to check morphology of cells treated by AKBA. Our results showed that AKBA (100 mg/kg) significantly inhibited the growth of orthotopic U87-MG gliomas. Results from MALDI-MSI showed that AKBA improved the metabolic profile of mice with glioblastoma, while immunoblot assays revealed that AKBA suppressed the expression of ATG5, p62, LC3B, p-ERK/ERK, and P53, and increased the ratio of p-mTOR/mTOR. Taken together, these results suggested that the antitumor effects of AKBA were related to the normalization of aberrant metabolism in the glioblastoma and the inhibition of autophagy. AKBA could be a promising chemotherapy drug for glioblastoma.
Collapse
Key Words
- AKBA
- AKBA, 3-O-acetyl-11-keto-β-boswellic acid
- Autophagy
- DAPI, 4′,-6-diamidino-2-phenylindole
- G3P, glycerol-3-phosphate
- G6P, glucose-6-phosphate
- GBM, glioblastomas
- GL/FFA, glycerolipid/free fatty acid
- Glioblastoma
- IDH1/2, isocitrate dehydrogenases 1/2
- ITO, indium tin oxide
- LA, linoleic acid
- MALDI-MSI
- MALDI-MSI, matrix-assisted laser desorption ionization-mass spectrometry imaging
- NAA, N-acetyl-l-aspartic acid
- NEDC, N-(1-naphthyl) ethylenediamine dihydrochloride
- OA, oleic acid
- PA, phosphatidic acid
- PE, phosphatidylethanolamine
- PG, phosphatidylglycerols
- PI, phosphatidylinositol
- PS, phosphatidylserine
- Phospholipids
- TIC, total ion current
- TMZ, temozolomide
Collapse
Affiliation(s)
- Wan Li
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Liwen Ren
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Xiangjin Zheng
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Jinyi Liu
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Jinhua Wang
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Tengfei Ji
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Guanhua Du
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
72
|
Anticancer and antimicrobial effects of novel ciprofloxacin fatty acids conjugates. Eur J Med Chem 2020; 185:111810. [DOI: 10.1016/j.ejmech.2019.111810] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 10/18/2019] [Accepted: 10/21/2019] [Indexed: 12/21/2022]
|
73
|
Huerta-Ángeles G, Brandejsová M, Kopecká K, Ondreáš F, Medek T, Židek O, Kulhánek J, Vagnerová H, Velebný V. Synthesis and Physicochemical Characterization of Undecylenic Acid Grafted to Hyaluronan for Encapsulation of Antioxidants and Chemical Crosslinking. Polymers (Basel) 2019; 12:E35. [PMID: 31878337 PMCID: PMC7023664 DOI: 10.3390/polym12010035] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 12/05/2019] [Accepted: 12/21/2019] [Indexed: 12/31/2022] Open
Abstract
In this work, a new amphiphilic derivative made of 10-undecylenic acid grafted to hyaluronan was prepared by mixed anhydrides. The reaction conditions were optimized, and the effect of the molecular weight (Mw), reaction time, and the molar ratio of reagents was explored. Using this methodology, a degree of substitution up to 50% can be obtained. The viscosity of the conjugate can be controlled by varying the substitution degree. The physicochemical characterization of the modified hyaluronan was performed by infrared spectroscopy, Nuclear Magnetic Resonance, Size-Exclusion Chromatography combined with Multiangle Laser Light Scattering (SEC-MALLS), and rheology. The low proton motility and self-aggregation of the amphiphilic conjugate produced overestimation of the degree of substitution. Thus, a novel method using proton NMR was developed. Encapsulation of model hydrophobic guest molecules, coenzyme Q10, curcumin, and α-tocopherol into the micellar core was also investigated by solvent evaporation. HA-UDA amphiphiles were also shown to self-assemble into spherical nanostructures (about 300 nm) in water as established by dynamic light scattering. Furthermore, HA-UDA was crosslinked via radical polymerization mediated by ammonium persulphate (APS/TEMED). The cross-linking was also tested by photo-polymerization catalyzed by Irgacure 2959. The presence of the hydrophobic moiety decreases the swelling degree of the prepared hydrogels compared to methacrylated-HA. Here, we report a novel hybrid hyaluronan (HA) hydrogel system of physically encapsulated active compounds and chemical crosslinking for potential applications in drug delivery.
Collapse
|
74
|
Khan T, Relitti N, Brindisi M, Magnano S, Zisterer D, Gemma S, Butini S, Campiani G. Autophagy modulators for the treatment of oral and esophageal squamous cell carcinomas. Med Res Rev 2019; 40:1002-1060. [PMID: 31742748 DOI: 10.1002/med.21646] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 10/16/2019] [Accepted: 11/08/2019] [Indexed: 02/06/2023]
Abstract
Oral squamous cell carcinomas (OSCC) and esophageal squamous cell carcinomas (ESCC) exhibit a survival rate of less than 60% and 40%, respectively. Late-stage diagnosis and lack of effective treatment strategies make both OSCC and ESCC a significant health burden. Autophagy, a lysosome-dependent catabolic process, involves the degradation of intracellular components to maintain cell homeostasis. Targeting autophagy has been highlighted as a feasible therapeutic strategy with clinical utility in cancer treatment, although its associated regulatory mechanisms remain elusive. The detection of relevant biomarkers in biological fluids has been anticipated to facilitate early diagnosis and/or prognosis for these tumors. In this context, recent studies have indicated the presence of specific proteins and small RNAs, detectable in circulating plasma and serum, as biomarkers. Interestingly, the interplay between biomarkers (eg, exosomal microRNAs) and autophagic processes could be exploited in the quest for targeted and more effective therapies for OSCC and ESCC. In this review, we give an overview of the available biomarkers and innovative targeted therapeutic strategies, including the application of autophagy modulators in OSCC and ESCC. Additionally, we provide a viewpoint on the state of the art and on future therapeutic perspectives combining the early detection of relevant biomarkers with drug discovery for the treatment of OSCC and ESCC.
Collapse
Affiliation(s)
- Tuhina Khan
- Department of Biotechnology, Chemistry, and Pharmacy, Department of Excellence 2018-2022, University of Siena, Siena, Italy
| | - Nicola Relitti
- Department of Biotechnology, Chemistry, and Pharmacy, Department of Excellence 2018-2022, University of Siena, Siena, Italy
| | - Margherita Brindisi
- Department of Pharmacy, Department of Excellence 2018-2022, University of Napoli Federico IL, Napoli, Italy
| | - Stefania Magnano
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152-160, Pearse Street, Dublin, Dublin 2, Ireland
| | - Daniela Zisterer
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152-160, Pearse Street, Dublin, Dublin 2, Ireland
| | - Sandra Gemma
- Department of Biotechnology, Chemistry, and Pharmacy, Department of Excellence 2018-2022, University of Siena, Siena, Italy
| | - Stefania Butini
- Department of Biotechnology, Chemistry, and Pharmacy, Department of Excellence 2018-2022, University of Siena, Siena, Italy
| | - Giuseppe Campiani
- Department of Biotechnology, Chemistry, and Pharmacy, Department of Excellence 2018-2022, University of Siena, Siena, Italy
| |
Collapse
|
75
|
Induction of sub-G0 arrest and apoptosis by seed extract of Moringa peregrina (Forssk.) Fiori in cervical and prostate cancer cell lines. JOURNAL OF INTEGRATIVE MEDICINE-JIM 2019; 17:410-422. [DOI: 10.1016/j.joim.2019.09.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 05/20/2019] [Indexed: 12/24/2022]
|
76
|
Ogino N, Miyagawa K, Kusanaga M, Hayashi T, Minami S, Oe S, Honma Y, Harada M. Involvement of sarco/endoplasmic reticulum calcium ATPase-mediated calcium flux in the protective effect of oleic acid against lipotoxicity in hepatocytes. Exp Cell Res 2019; 385:111651. [PMID: 31568762 DOI: 10.1016/j.yexcr.2019.111651] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 09/22/2019] [Accepted: 09/26/2019] [Indexed: 12/14/2022]
Abstract
Elevated free fatty acids, particularly saturated ones such as palmitic acid, may play an important role in the lipotoxic mechanism of nonalcoholic fatty liver disease (NAFLD). Saturated fatty acids induce autophagy dysfunction and endoplasmic reticulum (ER) stress leading to apoptosis in hepatocytes. However, unsaturated fatty acids, such as oleic acid, are nontoxic and can even prevent saturated fatty acid-induced toxicity in vitro. Although emerging evidence has suggested that ER calcium flux disruption in hepatocytes is involved in NAFLD pathogenesis, the roles of fatty acids in autophagy and ER calcium flux still remain unclear. We demonstrated that oleic acid ameliorated palmitic acid-induced autophagy arrest and ER stress in parallel with ER calcium depletion in hepatocytes. Moreover, we found that the effect of oleic acid against autophagy arrest was reversed by the pharmacological inhibition of sarcoplasmic reticulum Ca2+-ATPase (SERCA), which influxes calcium to ER. These data suggest that SERCA-mediated ER calcium flux is greatly involved in fatty acid-induced lipotoxicity in hepatocytes, and the prevention of ER calcium depletion may restore saturated fatty acid-induced autophagy arrest in hepatocytes.
Collapse
Affiliation(s)
- Noriyoshi Ogino
- Third Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan.
| | - Koichiro Miyagawa
- Third Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Masashi Kusanaga
- Third Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Tsuguru Hayashi
- Third Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Sota Minami
- Third Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Shinji Oe
- Third Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Yuichi Honma
- Third Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Masaru Harada
- Third Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan.
| |
Collapse
|
77
|
Ikumawoyi VO, Awodele O, Agbaje EO, Alimba CG, Bakare AA, Akinloye O. Bioactivity and modulatory functions of Napoleona vogelii on oxidative stress-induced micronuclei and apoptotic biomarkers in mice. Toxicol Rep 2019; 6:963-974. [PMID: 31673498 PMCID: PMC6816133 DOI: 10.1016/j.toxrep.2019.09.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 09/17/2019] [Accepted: 09/20/2019] [Indexed: 02/08/2023] Open
Abstract
Napoleona vogelii is used in traditional medicine for the management of pain, inflammatory conditions and cancer. This study was conducted to investigate the modulatory mechanisms of methanol stem bark extract of N. vogelii on induction of micronuclei, apoptotic biomarkers and in vivo antioxidant enzymes in mice. Forty male albino mice were randomly divided into eight groups (n = 5) and were administered distilled water (DW, 5 mL/kg) as negative control, 100, 200 or 400 mg/kg of the extract respectively for 28 days before the injection of cyclophosphamide (CP, 40 mg/kg) i.p. on the 28th day. The remaining groups were administered 100, 200 or 400 mg/kg of the extract only for 28 days. Twenty four hours after injection of CP or administration of the last dose of extract, animals were euthanized by cervical dislocation and blood samples collected for determination of in vivo antioxidants, the spleen harvested for immunohistochemical expression of NFκB, Bcl-2, Bax and p53. Bone marrow smears were also made for the micronucleus assay. Treatment with the extract resulted in a significant (p < 0.0001) reduction in frequency of micronucleated polychromatic erythrocytes (MNPCEs) compared to CP exposed control conferring protection of 75.09, 94.74 and 96.84% at 100, 200 or 400 mg/kg respectively. In extract and CP exposed animals, there were significant (p < 0.05) increases in GSH, GST and SOD with a corresponding significant (p < 0.05) reduction in MDA. In addition, the extract significantly downregulated cytoplasmic levels of NFκB and Bcl-2 and upregulated Bax and p53. These findings demonstrate that N. vogelli may serve as an interesting lead for chemo-preventive drug development.
Collapse
Affiliation(s)
- Victor Olabowale Ikumawoyi
- Department of Pharmacology Therapeutics and Toxicology, Faculty of Basic Medical Sciences, College of Medicine, University of Lagos, PMB 12003, Idi-Araba, Lagos, Nigeria
| | - Olufunsho Awodele
- Department of Pharmacology Therapeutics and Toxicology, Faculty of Basic Medical Sciences, College of Medicine, University of Lagos, PMB 12003, Idi-Araba, Lagos, Nigeria
| | - Esther Oluwatoyin Agbaje
- Department of Pharmacology Therapeutics and Toxicology, Faculty of Basic Medical Sciences, College of Medicine, University of Lagos, PMB 12003, Idi-Araba, Lagos, Nigeria
| | - Chibuisi Gideon Alimba
- Leibniz Research Centre for Working Environment and Human Factors (IfADo), Technical University of Dortmund, 44139 Dortmund, Germany
- Cell Biology and Genetics Unit, Department of Zoology, Faculty of Science, University of Ibadan, Nigeria
| | - Adekunle Akeem Bakare
- Cell Biology and Genetics Unit, Department of Zoology, Faculty of Science, University of Ibadan, Nigeria
| | - Oluyemi Akinloye
- Department of Medical Laboratory Science, Faculty of Basic Medical Sciences, College of Medicine University of Lagos, Idi-Araba, Lagos, Nigeria
| |
Collapse
|
78
|
Halczy-Kowalik L, Drozd A, Stachowska E, Drozd R, Żabski T, Domagała W. Fatty acids distribution and content in oral squamous cell carcinoma tissue and its adjacent microenvironment. PLoS One 2019; 14:e0218246. [PMID: 31242216 PMCID: PMC6594603 DOI: 10.1371/journal.pone.0218246] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 05/29/2019] [Indexed: 12/25/2022] Open
Abstract
Squamous cell carcinoma of the oral cavity mucosa grows under conditions of poor oxygenation and nutrient scarcity. Reprogramming of lipid biosynthesis accompanies tumor growth, but the conditions under which it occurs are not fully understood. The fatty acid content of the serum, tumor tissue and adjacent tumor microenvironment was measured by gas chromatography in 30 patients with squamous cell carcinoma grade 1-3. Twenty-five fatty acids were identified; their frequencies and percentages in each of the environments were assessed. Nineteen of the twenty-five fatty acids were found in tumor tissue, tumor adjacent tissue and blood serum. Of them, 8 were found in all thirty patients. Percentages of C16:0 and C18:1n9 were highest in the tumor, C18:1n9 and C16:0 were highest in tumor adjacent tissue, and C16:0 and C18:0 were highest in blood serum. The frequencies and amounts of C22:1n13, C22:4n6, C22:5n3 and C24:1 in tumor adjacent tissues were higher than those in blood serum, independent of the tumor grade. The correlations between the amount of fatty acid and tumor grade were the strongest in tumor adjacent tissues. The correlations between particular fatty acids were most prevalent for grade 1+2 tumors and were strongest for grade 3 tumors. In the adjacent tumor microenvironment, lipogenesis was controlled by C22:6w3. In blood serum, C18:1trans11 limited the synthesis of long-chain fatty acids. Our research reveals intensive lipid changes in oral cavity SCC adjacent to the tumor microenvironment and blood serum of the patients. Increase in percentage of some of the FAs in the path: blood serum-tumor adjacent microenvironment-tumor, and it is dependent on tumor grade. This dependency is the most visible in the tumor adjacent environment.
Collapse
Affiliation(s)
- Ludmiła Halczy-Kowalik
- Clinic of Maxillofacial Surgery, Pomeranian Medical University, Szczecin, Poland
- * E-mail:
| | - Arleta Drozd
- Department of Biochemistry and Human Nutrition, Pomeranian Medical University, Szczecin, Poland
| | - Ewa Stachowska
- Department of Biochemistry and Human Nutrition, Pomeranian Medical University, Szczecin, Poland
| | - Radosław Drozd
- Department of Immunology, Microbiology and Physiological Chemistry, West Pomeranian University of Technology, Szczecin, Poland
| | - Tomasz Żabski
- Clinic of Maxillofacial Surgery, Pomeranian Medical University, Szczecin, Poland
| | - Wenancjusz Domagała
- Department of Pathomorphology, Pomeranian Medical University, Szczecin, Poland
| |
Collapse
|
79
|
Li J, Xiang X, Xu Z. Cilostazol protects against myocardial ischemia and reperfusion injury by activating transcription factor EB (TFEB). Biotechnol Appl Biochem 2019; 66:555-563. [PMID: 30994947 DOI: 10.1002/bab.1754] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 04/15/2019] [Indexed: 12/13/2022]
Abstract
Although cilostazol was proved to have antitumor biological effects, its function in myocardial ischemia and reperfusion (I/R) injury and the underlying mechanisms were not fully illustrated yet. In this study, a rat model of I/R injury was constructed and quantitative real-time PCR, Western blot, and immunofluorescence (IF) assay were performed. Our results showed that cilostazol increased LC3 II/LC3 I ratio, reduced p62 abundance, and promoted the expressions of LAMP1, LAMP2, cathepsin B, and cathepsin D, indicating that cilostazol could activate autophagy and elevated lysosome activation. Following analysis showed that cilostazol enhanced nuclear protein expression of transcription factor EB (TFEB), an important regulator of autophagy-lysosome pathway. Furthermore, CCI-779, an inhibitor of TFEB, could reverse the effects of cilostazol on autophagic activity and lysosome activation. Importantly, cilostazol suppressed I/R injury-induced apoptosis by decreasing the cleavage of caspase 3 and PARP. Enzyme-linked immunosorbent assay showed that cilostazol reduced the serum levels of CTn1 and CK-MB and decreased infract size caused by I/R injuries. Altogether this study suggested that cilostazol protects against I/R injury by regulating autophagy, lysosome, and apoptosis in a rat model of I/R injury. The protective mechanism of cilostazol was partially through increasing the transcriptional activity of TFEB.
Collapse
Affiliation(s)
- Jiangjin Li
- Department of Cardiology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, People's Republic of China
| | - Xiaoli Xiang
- Department of Cardiology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, People's Republic of China
| | - Zuo Xu
- Department of Cardiology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, People's Republic of China
| |
Collapse
|
80
|
The serum biomarker chemerin promotes tumorigenesis and metastasis in oral squamous cell carcinoma. Clin Sci (Lond) 2019; 133:681-695. [PMID: 30804218 DOI: 10.1042/cs20181023] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 02/20/2019] [Accepted: 02/21/2019] [Indexed: 02/08/2023]
Abstract
Chemerin, which is encoded by retinoic acid receptor responder 2 (RARRES2), has been found to be related to malignant tumours, but its role in the development of oral squamous cell carcinoma (OSCC) is largely unexplored. In the present study, a higher serum level of chemerin was evident in patients with OSCC than in healthy individuals, and this high level of chemerin significantly decreased after tumour resection. In addition, high chemerin levels were positively associated with advanced tumour stage and lymph node metastasis. The expression levels of chemerin and Chemerin Receptor 23 (ChemR23) were positively correlated with the migration and invasion of OSCC cell lines. Recombinant chemerin (R-chemerin) enhanced the in vitro migration, invasion and proliferation of OSCC cells in a concentration-dependent manner, and short hairpin RNAs (shRNAs) targeting RARRES2 decreased chemerin expression and inhibited OSCC cell metastasis and proliferation both in vitro and in vivo Additionally, R-chemerin activated manganese superoxide dismutase (SOD2) and increased the amount of intracellular hydrogen peroxide (H2O2), leading to a significant decrease in E-cadherin expression and dramatic increase in the expression of phosphorylated ERK1/2 (p-ERK1/2), Slug, Vimentin and N-cadherin, but shRNAs targeting RARRES2 reversed these effects. Moreover, knockdown of ChemR23 with small interfering RNAs (siRNA) significantly inhibited chemerin-induced OSCC cell migration/invasion and SOD2 activity. Our results revealed that chemerin is a novel biomarker for OSCC. Chemerin/ChemR23 promotes tumorigenesis and metastasis in OSCC and may be a new therapeutic target for OSCC.
Collapse
|
81
|
Paine MRL, Liu J, Huang D, Ellis SR, Trede D, Kobarg JH, Heeren RMA, Fernández FM, MacDonald TJ. Three-Dimensional Mass Spectrometry Imaging Identifies Lipid Markers of Medulloblastoma Metastasis. Sci Rep 2019; 9:2205. [PMID: 30778099 PMCID: PMC6379434 DOI: 10.1038/s41598-018-38257-0] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 12/18/2018] [Indexed: 12/11/2022] Open
Abstract
Treatment for medulloblastoma (MB) — the most common malignant pediatric brain tumor — includes prophylactic radiation administered to the entire brain and spine due to the high incidence of metastasis to the central nervous system. However, the majority of long-term survivors are left with permanent and debilitating neurocognitive impairments as a result of this therapy, while the remaining 30–40% of patients relapse with terminal metastatic disease. Development of more effective targeted therapies has been hindered by our lack of understanding of the underlying mechanisms regulating the metastatic process in this disease. To understand the mechanism by which MB metastasis occurs, three-dimensional matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI) experiments were performed on whole brains from a mouse model of human medulloblastoma. Analyzing the tumor and surrounding normal brain in its entirety enabled the detection of low abundance, spatially-heterogeneous lipids associated with tumor development. Boundaries of metastasizing and non-metastasizing primary tumors were readily defined, leading to the identification of lipids associated with medulloblastoma metastasis, including phosphatidic acids, phosphatidylethanolamines, phosphatidylserines, and phosphoinositides. These lipids provide a greater insight into the metastatic process and may ultimately lead to the discovery of biomarkers and novel targets for the diagnosis and treatment of metastasizing MB in humans.
Collapse
Affiliation(s)
- Martin R L Paine
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, 30332, USA.,Maastricht Multimodal Molecular Imaging Institute, Division of Imaging Mass Spectrometry, Maastricht University, Maastricht, 6229ER, The Netherlands
| | - Jingbo Liu
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Danning Huang
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Shane R Ellis
- Maastricht Multimodal Molecular Imaging Institute, Division of Imaging Mass Spectrometry, Maastricht University, Maastricht, 6229ER, The Netherlands
| | | | | | - Ron M A Heeren
- Maastricht Multimodal Molecular Imaging Institute, Division of Imaging Mass Spectrometry, Maastricht University, Maastricht, 6229ER, The Netherlands.
| | - Facundo M Fernández
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, 30332, USA. .,Integrated Cancer Research Center, Georgia Institute of Technology, Atlanta, GA, 30332, USA. .,Institute of Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA.
| | - Tobey J MacDonald
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| |
Collapse
|
82
|
Kuriakose GC, Lakshmanan M D, Bp A, Rs HK, Th AK, Ananthaswamy K, C J. Extract of Penicillium sclerotiorum an endophytic fungus isolated from Cassia fistula L. induces cell cycle arrest leading to apoptosis through mitochondrial membrane depolarization in human cervical cancer cells. Biomed Pharmacother 2018; 105:1062-1071. [PMID: 30021342 DOI: 10.1016/j.biopha.2018.06.094] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 06/02/2018] [Accepted: 06/15/2018] [Indexed: 01/10/2023] Open
Abstract
Seventeen endophytic fungi were isolated from various tissues of Cassia fistula and the ethyl acetate extracts obtained from 21-day cultures of all the endophytic fungal isolates were initially screened for their cytotoxicity against HeLa (human cervical carcinoma) cells using MTT assay. Of these, Penicillium sclerotiorum extract (PSE), significantly affected the viability of HeLa cells in a dose-dependent manner. The extract of P. Sclerotiorum was further analyzed by GC-MS, which showed three compounds, hexadecanoic acid, oleic acid and benzoic acid to be the major active principles in the extracts.The extract was further tested for invitro cytotoxicity against five cancer cell lines. Of the cell lines tested, HeLa cells showed maximum sensitivity followed by A549, while A431 and U251 were moderately sensitive and MCF-7 was insensitive to the treatment. In addition, normal human embryonic kidney cells, HEK293 remained insensitive to the treatment. Furthermore, the mechanism of cytotoxic activity exhibited by PSE was investigated by evaluating cell cycle progression and apoptotic induction in HeLa cells. Cell cycle analysis revealed that the PSE arrested cells at S and G2/M phase of the cell cycle in a dose-dependent manner. Annexin V- Propidium iodide double staining showed that, the extract potentiates apoptosis rather than necrosis in cells. This was supported by the down regulation in the proapoptotic protein BCL2 and up regulation of BAX (BCL2 Associated X), tumor suppressor protein, p53 and Apaf-1 [Apoptotic Peptidase Activating Factor 1]. Loss of mitochondrial membrane potential and a distinct DNA fragmentation pattern observed following the treatment, suggest that the PSE treatment leads to activation of mitochondrial pathway of apoptosis. Further, the extract also exhibited both antioxidant and anti-angiogenic properties. These results indicate that endophytic fungi isolated from medicinal plants may serve as potential sources of the anti-cancerous compounds.
Collapse
Affiliation(s)
- Gini C Kuriakose
- Department of Biochemistry, Indian Institute of Science, Bangalore 560012, India
| | - Divya Lakshmanan M
- Department of Biochemistry, Indian Institute of Science, Bangalore 560012, India; Yenepoya Research Centre, Yenepoya University, University Road, Mangalore 575018, India
| | - Arathi Bp
- Department of Biochemistry, Indian Institute of Science, Bangalore 560012, India
| | - Hari Kumar Rs
- Department of Biochemistry, Indian Institute of Science, Bangalore 560012, India
| | - Anantha Krishna Th
- Department of Biochemistry, Indian Institute of Science, Bangalore 560012, India
| | - Kavya Ananthaswamy
- Department of Biochemistry, Indian Institute of Science, Bangalore 560012, India
| | - Jayabhaskaran C
- Department of Biochemistry, Indian Institute of Science, Bangalore 560012, India.
| |
Collapse
|
83
|
Garcia EJ, Vevea JD, Pon LA. Lipid droplet autophagy during energy mobilization, lipid homeostasis and protein quality control. Front Biosci (Landmark Ed) 2018; 23:1552-1563. [PMID: 29293450 DOI: 10.2741/4660] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Lipid droplets (LDs) have well-established functions as sites for lipid storage and energy mobilization to meet the metabolic demands of cells. However, recent studies have expanded the roles of LDs to protein quality control. Lipophagy, or LD degradation by autophagy, plays a vital role not only in the mobilization of free fatty acids (FFAs) and lipid homeostasis at LDs, but also in the adaptation of cells to certain forms of stress including lipid imbalance. Recent studies have provided new mechanistic insights about the diverse types of lipophagy, in particular microlipophagy. This review summarizes key findings about the mechanisms and functions of lipophagy and highlights a novel function of LD microlipophagy as a mechanism to maintain endoplasmic reticulum (ER) proteostasis under conditions of lipid imbalance.
Collapse
Affiliation(s)
- Enrique J Garcia
- Department of Pathology and Cell Biology, Columbia University, New York, NY, 10032 USA
| | - Jason D Vevea
- HHMI and Dept. of Neuroscience, University of Wisconsin, Madison, WI, 53705 USA
| | - Liza A Pon
- Department of Pathology and Cell Biology, Columbia University, New York, NY, 10032 USA,
| |
Collapse
|
84
|
Pang L, Liu K, Liu D, Lv F, Zang Y, Xie F, Yin J, Shi Y, Wang Y, Chen D. Differential effects of reticulophagy and mitophagy on nonalcoholic fatty liver disease. Cell Death Dis 2018; 9:90. [PMID: 29367738 PMCID: PMC5833629 DOI: 10.1038/s41419-017-0136-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 11/09/2017] [Accepted: 11/09/2017] [Indexed: 12/14/2022]
Abstract
Autophagy affects the pathological progression of non-alcoholic fatty liver disease (NAFLD); however, the precise role of autophagy in NAFLD remains unclear. In this study, we want to identify the role of autophagy including reticulophagy and mitophagy in NAFLD pathogenesis. When HepG2 cells were treated with 400 μM oleic acid (OA), increased reticulophagy was induced 8 h after treatment, which correlated with an anti-apoptotic response as shown by the activation of the PI3K/AKT pathway, an increase in BCL-2 expression, and the downregulation of OA-induced lipotoxicity. When treated with OA for 24 h, DRAM expression-dependent mitophagy resulted in increased apoptosis in HepG2 cells. Inhibition of reticulophagy aggravated and increased lipotoxicity-induced apoptosis 8 h after treatment; however, the inhibition of mitophagy decreased hepatocyte apoptosis after 24 h of OA treatment. Results from the analysis of patient liver samples showed that autophagic flux increased in patients with mild or severe NAFL. PI3K/AKT phosphorylation was observed only in samples from patients with low-grade steatosis, whereas DRAM expression was increased in samples from patients with high-grade steatosis. Together, our results demonstrate that reticulophagy and mitophagy are independent, sequential events that influence NAFLD progression, which opens new avenues for investigating new therapeutics in NAFLD.
Collapse
Affiliation(s)
- Lijun Pang
- Beijing Institute of Hepatology, Capital Medical University, 100069, Beijing, China.,Beijing You'an Hospital, Capital Medical University, 100069, Beijing, China
| | - Kai Liu
- Beijing Institute of Hepatology, Capital Medical University, 100069, Beijing, China.,Beijing You'an Hospital, Capital Medical University, 100069, Beijing, China
| | - Daojie Liu
- Department of Clinical Laboratory, Haidian Maternal & Child Health Hospital, 100080, Beijing, China
| | - Fudong Lv
- Beijing You'an Hospital, Capital Medical University, 100069, Beijing, China
| | - Yunjin Zang
- Organ Transplantation Center, The Affiliated Hospital of Qingdao University, 26603, Qingdao, Shandong, China
| | - Fang Xie
- Beijing Institute of Hepatology, Capital Medical University, 100069, Beijing, China.,Beijing You'an Hospital, Capital Medical University, 100069, Beijing, China
| | - Jiming Yin
- Beijing Institute of Hepatology, Capital Medical University, 100069, Beijing, China.,Beijing You'an Hospital, Capital Medical University, 100069, Beijing, China
| | - Ying Shi
- Beijing Institute of Hepatology, Capital Medical University, 100069, Beijing, China.,Beijing You'an Hospital, Capital Medical University, 100069, Beijing, China
| | - Yanjun Wang
- Beijing Institute of Hepatology, Capital Medical University, 100069, Beijing, China.,Beijing You'an Hospital, Capital Medical University, 100069, Beijing, China
| | - Dexi Chen
- Beijing Institute of Hepatology, Capital Medical University, 100069, Beijing, China. .,Beijing You'an Hospital, Capital Medical University, 100069, Beijing, China. .,Organ Transplantation Center, The Affiliated Hospital of Qingdao University, 26603, Qingdao, Shandong, China.
| |
Collapse
|
85
|
Wang W, Zhang L, Chen T, Guo W, Bao X, Wang D, Ren B, Wang H, Li Y, Wang Y, Chen S, Tang B, Yang Q, Chen C. Anticancer Effects of Resveratrol-Loaded Solid Lipid Nanoparticles on Human Breast Cancer Cells. Molecules 2017; 22:molecules22111814. [PMID: 29068422 PMCID: PMC6150230 DOI: 10.3390/molecules22111814] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Accepted: 10/21/2017] [Indexed: 12/13/2022] Open
Abstract
In this study, resveratrol-loaded solid lipid nanoparticles (Res-SLNs) were successfully designed to treat MDA-MB-231 cells. The Res-SLNs were prepared using emulsification and low-temperature solidification method. The Res-SLNs were spherical, with small size, negative charge, and narrow size distribution. Compared with free resveratrol, the Res-SLNs displayed a superior ability in inhibiting the proliferation of MDA-MB-231 cells. In addition, Res-SLNs exhibited much stronger inhibitory effects on the invasion and migration of MDA-MB-231 cells. Western blot analysis revealed that Res-SLNs could promote the ratio of Bax/Bcl-2 but decreased the expression of cyclinD1 and c-Myc. These results indicate that the Res-SLN may have great potential for breast cancer treatment.
Collapse
Affiliation(s)
- Wenrui Wang
- Department of Biotechnology, Bengbu Medical College, Anhui, Bengbu 233030, China.
| | - Lingyu Zhang
- Clinical Testing and Diagnose Experimental Center, Bengbu Medical College, Anhui, Bengbu 233030, China.
| | - Tiantian Chen
- Clinical Testing and Diagnose Experimental Center, Bengbu Medical College, Anhui, Bengbu 233030, China.
| | - Wen Guo
- Department of Biotechnology, Bengbu Medical College, Anhui, Bengbu 233030, China.
| | - Xunxia Bao
- Department of Biotechnology, Bengbu Medical College, Anhui, Bengbu 233030, China.
| | - Dandan Wang
- Department of Biotechnology, Bengbu Medical College, Anhui, Bengbu 233030, China.
| | - Baihui Ren
- Department of Biotechnology, Bengbu Medical College, Anhui, Bengbu 233030, China.
| | - Haifeng Wang
- Clinical Testing and Diagnose Experimental Center, Bengbu Medical College, Anhui, Bengbu 233030, China.
| | - Yu Li
- Clinical Testing and Diagnose Experimental Center, Bengbu Medical College, Anhui, Bengbu 233030, China.
| | - Yueyue Wang
- Clinical Testing and Diagnose Experimental Center, Bengbu Medical College, Anhui, Bengbu 233030, China.
| | - Sulian Chen
- Department of Biochemistry and Molecular Biology, Bengbu Medical College, Anhui, Bengbu 233030, China.
| | - Baoding Tang
- Department of Oncology, Bengbu Medical College, Anhui, Bengbu 233030, China.
| | - Qingling Yang
- Department of Biochemistry and Molecular Biology, Bengbu Medical College, Anhui, Bengbu 233030, China.
| | - Changjie Chen
- Department of Biochemistry and Molecular Biology, Bengbu Medical College, Anhui, Bengbu 233030, China.
| |
Collapse
|