1
|
He M, Wu H, Hu L, Liu N, Zhang G, Wang S. Regulatory mechanism of the Glabrene against non-small cell lung cancer by suppressing FGFR3. ENVIRONMENTAL TOXICOLOGY 2025; 40:412-428. [PMID: 38517198 DOI: 10.1002/tox.24235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 02/29/2024] [Accepted: 03/04/2024] [Indexed: 03/23/2024]
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) is a highly malignant tumor with limited effective treatment options. This study aimed to investigate the regulatory mechanism of Glabrene on NSCLC through its interaction with FGFR3. METHODS HCC827 cells were implanted into nude mice and treated with Glabrene. Tumor volume was monitored at 0, 3, 6, and 9 days after medical treatment. Tissue analysis included Hematoxylin and Eosin (HE) and Terminal deoxynucleotidyl transferase (TdT)-mediated dUTP Nick End Labeling (TUNEL) staining, as well as immunohistochemistry for Ki67, ERK1/2, and p-ERK1/2 expression. Cell viability was determined with the CCK8 method. We utilized immunofluorescence techniques to observe apoptosis, as well as the levels of E-cadherin and Vimentin expression. Cellular proliferation was determined via plate cloning assay and cellular mobility was determined via scratch assay. Cellular invasion ability was assessed via a transwell assay. mRNA and protein levels of FGFR3, MMP1, MMP9, vimentin, E-cadherin, ERK1/2, and p-ERK1/2 were detected via qPCR and Western blot. IGF-1, VEGF, and Estradiol (E2) levels were measured through Enzyme linked immunosorbent assay (ELISA). RESULTS This study verified that Glabrene was capable of suppressing tumor growth in NSCLC mice, reversing tumor tissue's pathological morphology, attenuating the capacities of cancerous cells' proliferation, migration, and invasion, and leading to apoptosis. Besides, Glabrene could reduce the FGFR3 expression in HCC827 cells. Over-expression of FGFR3 promotes the proliferation of HCC827 cells, increase both contents of IGF-1, VEGF, and E2, and expressions of MMP1, MMP9, vimentin, and p-ERK1/2, while Glabrene inhibited FGFR3. Glabrene, and inhibition of FGFR3 expression were capable of decreasing FGFR3, MMP1, MMP9, vimentin, and p-ERK1/2 expression, as well as contents of IGF-1, VEGF, and E2 in model mice and HCC827 cells, and promoting the expression of E-cadherin. CONCLUSION Glabrene has the potential as a therapeutic agent for NSCLC by reducing cancer invasion and migration through the inhibition of ERK1/2 phosphorylation and suppression of epithelial-mesenchymal transition (EMT).
Collapse
Affiliation(s)
- Miao He
- College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, China
- Department of Hematology and Oncology, Chongqing Oncology Hematology Department, Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Huiling Wu
- College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, China
- Bone and joint rehabilitation department, The Affiliated Rehabilitation Hospital of Chongqing Medical University, Chongqing, China
| | - Lingjing Hu
- Department of Hematology and Oncology, Chongqing Oncology Hematology Department, Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Nan Liu
- Department of Hematology and Oncology, Chongqing Oncology Hematology Department, Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Guoduo Zhang
- Department of Hematology and Oncology, Chongqing Oncology Hematology Department, Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Shumei Wang
- College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, China
| |
Collapse
|
2
|
He J, Dong C, Song X, Qiu Z, Zhang H, Jiang Y, Liu T, Man X. Methyltransferase-like 7B participates in bladder cancer via ACSL3 m 6A modification in a ferroptosis manner. Biol Direct 2025; 20:9. [PMID: 39833962 PMCID: PMC11744867 DOI: 10.1186/s13062-025-00597-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 01/06/2025] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Bladder cancer (BC) is a malignant tumor. Methyltransferase-like 7B (MEETL7B) is a methyltransferase and its role in BC has not yet been revealed. METHOD Stable METTL7B knockdown or overexpression were achieved by lentiviral transduction in SW780 and TCCSUP cell lines. Xenografts tumors were established via subcutaneous injection of stable transfectants in BALB/c mice. RESULTS A database search indicated that METTL7B was elevated in BC and it was validated in BC cell lines. METTL7B silencing suppressed cell proliferation and tumorigenesis in vitro and in vivo. Besides, METTL7B knockdown induced cell cycle arrest in G1 phase with a reduction in cyclin D1(CCND1), CDK4, and CDK6 levels and an elevation in CDKN2D levels in cells. Considering that ferroptosis is emerging as a therapeutic target for cancer, and the possible relationship between METTL7B and antioxidant enzymes. We, here, examined that ectopic METTL7B expression abolished ferroptosis markers in cells raised by Erastin treatment, including the production of lipid ROS, the increased cellular iron and MDA content, the decreased gene expression of ACSL3, FANCD2, and FADS2, as well as the mitochondrial injury observed by electron microscopy. Mechanically, ectopic METTL7B expression promoted m6A modification on ACSL3 mRNA. Gain of functional experiment exhibited that METTL7B inhibited Erastin-induced ferroptosis via ACSL3. Overexpressed PLAGL2 is identified as a possible independent predictor in BC and bioinformatics predicted the potential binding sites between PLAGL2 and METTL7B promoter region. Dual luciferase and chromatin immunoprecipitation analysis provided evidence that PLAGL2 directly binds to METTL7B promoter region. CONCLUSIONS METTL7B is involved in BC development and progression. METTL7B may mediate m6A modification on ACSL3 mRNA to negatively regulate ferroptosis in BC cells, which provides a potential therapeutic target for BC via ferroptosis.
Collapse
Affiliation(s)
- Jiani He
- Department of Surgical Oncology and Breast Surgery, The First Hospital of China Medical University, 155 North Nanjing Street, Heping District, Shenyang, Liaoning, China
| | - Changming Dong
- Department of Urology, The First Hospital of China Medical University, 155 North Nanjing Street, Heping District, Shenyang, Liaoning, China
- Institute of Urology, China Medical University, Shenyang, Liaoning, China
| | - Xiandong Song
- Department of Urology, The First Hospital of China Medical University, 155 North Nanjing Street, Heping District, Shenyang, Liaoning, China
- Institute of Urology, China Medical University, Shenyang, Liaoning, China
- Department of Urology, Shenyang Fifth People Hospital, Shenyang, Liaoning, China
| | - Zhongkai Qiu
- Institute of Urology, Benxi Central Hospital, 29 Shengli Road, Mingshan District, Benxi, Liaoning, China
| | - Hao Zhang
- Department of Urology, The First Hospital of China Medical University, 155 North Nanjing Street, Heping District, Shenyang, Liaoning, China
- Institute of Urology, China Medical University, Shenyang, Liaoning, China
| | - Yuanjun Jiang
- Department of Urology, The First Hospital of China Medical University, 155 North Nanjing Street, Heping District, Shenyang, Liaoning, China
- Institute of Urology, China Medical University, Shenyang, Liaoning, China
| | - Tao Liu
- Department of Urology, The First Hospital of China Medical University, 155 North Nanjing Street, Heping District, Shenyang, Liaoning, China
- Institute of Urology, China Medical University, Shenyang, Liaoning, China
| | - Xiaojun Man
- Department of Urology, The First Hospital of China Medical University, 155 North Nanjing Street, Heping District, Shenyang, Liaoning, China.
- Institute of Urology, China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
3
|
El-Shoura EAM, Abdelzaher LA, Mahmoud NI, Farghaly OA, Sabry M, Girgis Shahataa M, Salem EA, Saad HM, Elhussieny O, Kozman MR, Atwa AM. Combined sulforaphane and β-sitosterol mitigate olanzapine-induced metabolic disorders in rats: Insights on FOXO, PI3K/AKT, JAK/STAT3, and MAPK signaling pathways. Int Immunopharmacol 2024; 140:112904. [PMID: 39116489 DOI: 10.1016/j.intimp.2024.112904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 07/27/2024] [Accepted: 08/05/2024] [Indexed: 08/10/2024]
Abstract
One of the best antipsychotics for treating schizophrenia and bipolar disorders is olanzapine (OLA). However, its use is restricted owing to unfavorable adverse effects as liver damage, dyslipidemia, and weight gain. The primary objective of the present investigation was to examine the signaling mechanisms that underlie the metabolic disruption generated by OLA. Besides, the potential protective effect of sulforaphane (SFN) and β-sitosterol (βSS) against obesity and metabolic toxicity induced by OLA were inspected as well. A total of five groups of male Wistar rats were established, including the control, OLA, SFN+OLA, βSS+OLA, and the combination + OLA groups. Hepatic histopathology, biochemical analyses, ultimate body weights, liver function, oxidative stress, and pro-inflammatory cytokines were evaluated. In addition to the relative expression of FOXO, the signaling pathways for PI3K/AKT, JAK/STAT3, and MAPK were assessed as well. All biochemical and hepatic histopathological abnormalities caused by OLA were alleviated by SFN and/or βSS. A substantial decrease in systolic blood pressure (SBP), proinflammatory cytokines, serum lipid profile parameters, hepatic MDA, TBIL, AST, and ALT were reduced through SFN or/and βSS. To sum up, the detrimental effects of OLA are mediated by alterations in the Akt/FOXO3a/ATG12, Ras/SOS2/Raf-1/MEK/ERK1/2, and Smad3,4/TGF-β signaling pathways. The administration of SFN and/or βSS has the potential to mitigate the metabolic deficit, biochemical imbalances, hepatic histological abnormalities, and the overall unfavorable consequences induced by OLA by modulating the abovementioned signaling pathways.
Collapse
Affiliation(s)
- Ehab A M El-Shoura
- Department of Clinical Pharmacy, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut 71524, Egypt.
| | - Lobna A Abdelzaher
- Department of Pharmacology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Nesreen I Mahmoud
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Nahda University, Beni-Suef, Egypt
| | - Omar A Farghaly
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Egypt
| | - Mostafa Sabry
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Egypt
| | - Mary Girgis Shahataa
- Department of Pharmacology, Faculty of Medicine, Beni-Suef University, Beni-Suef, Egypt
| | - Esraa A Salem
- Department of Medical Physiology, Faculty of Medicine, Menoufia University, Shebeen ElKom, 32511, Egypt
| | - Hebatallah M Saad
- Department of Pathology, Faculty of Veterinary Medicine, Matrouh University, Marsa Matruh 51744, Egypt
| | - Omnya Elhussieny
- Department of Histology and Cytology, Faculty of Veterinary Medicine, Matrouh University, Marsa Matruh 51744, Egypt
| | - Magy R Kozman
- Clinical Pharmacy Department, Faculty of Pharmacy, Misr University for Science and Technology, Giza 12563, Egypt
| | - Ahmed M Atwa
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Egyptian Russian University, Cairo, Egypt; Pharmacology and Toxicology Department, Faculty of Pharmacy, Al-Ayen Iraqi University, Thi-Qar 64001, Iraq
| |
Collapse
|
4
|
Jiang P, Jiang W, Li X, Zhu Q. Combination of Formononetin and Sulforaphane Natural Drug Repress the Proliferation of Cervical Cancer Cells via Impeding PI3K/AKT/mTOR Pathway. Appl Biochem Biotechnol 2024; 196:6726-6744. [PMID: 38401043 DOI: 10.1007/s12010-024-04873-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/12/2024] [Indexed: 02/26/2024]
Abstract
Natural substances have been demonstrated to be an unrivalled source of anticancer drugs in the present era of pharmacological development. Plant-based substances, together with their derivatives through analogues, play a significant character in the treatment of cancer by altering the tumor microenvironment and several signaling pathways. In this study, it was investigated whether the natural drugs, formononetin (FN) and sulforaphane (SFN), when combined, assess the efficacy of inhibiting cervical cancer cell proliferation by impeding the PI3K/Akt/mTOR signaling pathway in HeLa cells. The cells were treated with the combination of FN and SFN (FN + SFN) in various concentrations (0-50 µM) for 24 h and then analyzed for various experiments. The combination of FN + SFN-mediated cytotoxicity was analyzed by MTT assay. DCFH-DA staining was used to assess the ROS measurement, and apoptotic changes were studied by dual (AO/EtBr) staining assays. Protein expressions of cell survival, cell cycle, proliferation, and apoptosis protein were evaluated by flow cytometry and western blotting. Results showed that the cytotoxicity of FN and SFN was determined to be around 23.7 µM and 26.92 µM, respectively. Combining FN and SFN causes considerable cytotoxicity in HeLa cells, with an IC50 of 21.6 µM after 24-h incubation. Additionally, HeLa cells treated with FN and SFN together showed increased apoptotic signals and considerable ROS generation. Consequently, by preventing the production of PI3K, AKT, and mToR-mediated regulation of proliferation and cell cycle-regulating proteins, the combined use of FN + SFN has been regarded as a chemotherapeutic medication. Further research will need to be done shortly to determine how effectively the co-treatment promotes apoptosis to employ them economically.
Collapse
Affiliation(s)
- Ping Jiang
- Gynaecology and Obstetrics, Yantai Mountain Hospital, Yantai, 264005, China
| | - Wei Jiang
- Medical Department, Jinan Maternity and Child Care Hospital, Jinan, 250000, China
| | - Xiujin Li
- Delivery Room, Jinan Maternity and Child Care Hospital, Jinan, 250000, China
| | - Qiuling Zhu
- Delivery Room, Jinan Maternity and Child Care Hospital, Jinan, 250000, China.
| |
Collapse
|
5
|
Anestopoulos I, Paraskevaidis I, Kyriakou S, Giova LE, Trafalis DT, Botaitis S, Franco R, Pappa A, Panayiotidis MI. Isothiocyanates Potentiate Tazemetostat-Induced Apoptosis by Modulating the Expression of Apoptotic Genes, Members of Polycomb Repressive Complex 2, and Levels of Tri-Methylating Lysine 27 at Histone 3 in Human Malignant Melanoma Cells. Int J Mol Sci 2024; 25:2745. [PMID: 38473991 DOI: 10.3390/ijms25052745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 02/22/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
In this study, we utilized an in vitro model consisting of human malignant melanoma as well as non-tumorigenic immortalized keratinocyte cells with the aim of characterizing the therapeutic effectiveness of the clinical epigenetic drug Tazemetostat alone or in combination with various isothiocyanates. In doing so, we assessed markers of cell viability, apoptotic induction, and expression levels of key proteins capable of mediating the therapeutic response. Our data indicated, for the first time, that Tazemetostat caused a significant decrease in viability levels of malignant melanoma cells in a dose- and time-dependent manner via the induction of apoptosis, while non-malignant keratinocytes were more resistant. Moreover, combinatorial treatment protocols caused a further decrease in cell viability, together with higher apoptotic rates. In addition, a significant reduction in the Polycomb Repressive Complex 2 (PRC2) members [e.g., Enhancer of Zeste Homologue 2 (EZH2), Embryonic Ectoderm Development (EED), and suppressor of zeste 12 (SUZ12)] and tri-methylating lysine 27 at Histone 3 (H3K27me3) protein expression levels was observed, at least partially, under specific combinatorial exposure conditions. Reactivation of major apoptotic gene targets was determined at much higher levels in combinatorial treatment protocols than Tazemetostat alone, known to be involved in the induction of intrinsic and extrinsic apoptosis. Overall, we developed an optimized experimental therapeutic platform aiming to ensure the therapeutic effectiveness of Tazemetostat in malignant melanoma while at the same time minimizing toxicity against neighboring non-tumorigenic keratinocyte cells.
Collapse
Affiliation(s)
- Ioannis Anestopoulos
- Department of Cancer Genetics, Therapeutics & Ultrastructural Pathology, The Cyprus Institute of Neurology & Genetics, 6 Iroon Avenue, Ayios Dometios, Nicosia 2371, Cyprus
| | - Ioannis Paraskevaidis
- Department of Cancer Genetics, Therapeutics & Ultrastructural Pathology, The Cyprus Institute of Neurology & Genetics, 6 Iroon Avenue, Ayios Dometios, Nicosia 2371, Cyprus
| | - Sotiris Kyriakou
- Department of Cancer Genetics, Therapeutics & Ultrastructural Pathology, The Cyprus Institute of Neurology & Genetics, 6 Iroon Avenue, Ayios Dometios, Nicosia 2371, Cyprus
| | - Lambrini E Giova
- Department of Cancer Genetics, Therapeutics & Ultrastructural Pathology, The Cyprus Institute of Neurology & Genetics, 6 Iroon Avenue, Ayios Dometios, Nicosia 2371, Cyprus
| | - Dimitrios T Trafalis
- Laboratory of Pharmacology, Medical School, National & Kapodistrian University of Athens, 11527 Athens, Greece
| | - Sotiris Botaitis
- Department of Surgery, School of Medicine, University Hospital, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Rodrigo Franco
- School of Veterinary Medicine & Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
- Redox Biology Centre, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Aglaia Pappa
- Department of Molecular Biology & Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Mihalis I Panayiotidis
- Department of Cancer Genetics, Therapeutics & Ultrastructural Pathology, The Cyprus Institute of Neurology & Genetics, 6 Iroon Avenue, Ayios Dometios, Nicosia 2371, Cyprus
| |
Collapse
|
6
|
Zhang C, Wu Q, Yao K, Jin G, Zhao S, Zhang J, Zheng W, Xu B, Zu Y, Yuan J, Liu K, Guo Y. Sulforaphene suppresses oesophageal cancer growth through mitogen- and stress-activated kinase 2 in a PDX mouse model. Am J Cancer Res 2023; 13:4708-4720. [PMID: 37970356 PMCID: PMC10636680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 09/04/2023] [Indexed: 11/17/2023] Open
Abstract
BACKGROUND Although sulforaphene has potential anticancer effects, little is known about its effect on oesophageal squamous cell carcinoma (ESCC) invasiveness. METHODS To investigate whether sulforaphene inhibits the growth of oesophageal cancer cells, MTT and anchorage-independent cell growth assays were performed. Global changes in the proteome and phosphoproteome of oesophageal cancer cells after sulforaphene treatment were analysed by mass spectrometry (MS), and the underlying molecular mechanism was further verified by in vivo and in vitro experiments. RESULTS Sulforaphene treatment markedly affected proteins that regulate several cellular processes in oesophageal cancer cells, and mitogen- and stress-activated kinase 2 (MSK2) was the main genetic target of sulforaphene in reducing the growth of oesophageal cancer cells. Sulforaphene significantly suppressed ESCC cell proliferation in vitro and reduced the tumour size in an oesophageal patient-derived xenograft (PDX) SCID mouse model. Furthermore, the binding of sulforaphane to MSK2 in vitro was verified using a cellular thermal dhift assay, and the effect of MSK2 knockdown on the ESCC phenotype was observed using a shMSK2 model. CONCLUSION The results showed that sulforaphene suppresses ESCC growth in both human oesophageal squamous cells and PDX mouse model by inhibiting MSK2 expression, implicating sulforaphene as a promising candidate for ESCC treatment.
Collapse
Affiliation(s)
- Chengjuan Zhang
- Center of Bio-Repository, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhou, Henan, P. R. China
- Henan Key Laboratory of Molecular PathologyZhengzhou, Henan, P. R. China
| | - Qiong Wu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou UniversityZhengzhou, Henan, P. R. China
- China-US (Henan) Hormel Cancer InstituteZhengzhou, Henan, P. R. China
| | - Ke Yao
- Department of Cellular and Molecular Biology, University of MinnesotaAustin, MN, USA
| | - Guoguo Jin
- Henan Key Laboratory of Chronic Disease Management, Fuwai Central China Cardiovascular HospitalZhengzhou, Henan, P. R. China
| | - Simin Zhao
- Department of Pathology, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhou, Henan, P. R. China
| | - Junxia Zhang
- Academy of Chinese Medical Sciences, Henan University of Chinese MedicineZhengzhou, Henan, P. R. China
| | - Wenjin Zheng
- Center of Bio-Repository, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhou, Henan, P. R. China
| | - Benling Xu
- Department of Immunotherapy, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhou, Henan, P. R. China
| | - Yingling Zu
- Department of Hematology, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhou, Henan, P. R. China
| | - Jing Yuan
- Center of Bio-Repository, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhou, Henan, P. R. China
| | - Kangdong Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou UniversityZhengzhou, Henan, P. R. China
- China-US (Henan) Hormel Cancer InstituteZhengzhou, Henan, P. R. China
| | - Yongjun Guo
- Department of Molecular Pathology, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhou, Henan, P. R. China
| |
Collapse
|
7
|
Sanati M, Afshari AR, Ahmadi SS, Moallem SA, Sahebkar A. Modulation of the ubiquitin-proteasome system by phytochemicals: Therapeutic implications in malignancies with an emphasis on brain tumors. Biofactors 2023; 49:782-819. [PMID: 37162294 DOI: 10.1002/biof.1958] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 04/20/2023] [Indexed: 05/11/2023]
Abstract
Regarding the multimechanistic nature of cancers, current chemo- or radiotherapies often fail to eradicate disease pathology, and frequent relapses or resistance to therapies occur. Brain malignancies, particularly glioblastomas, are difficult-to-treat cancers due to their highly malignant and multidimensional biology. Unfortunately, patients suffering from malignant tumors often experience poor prognoses and short survival periods. Thus far, significant efforts have been conducted to discover novel and more effective modalities. To that end, modulation of the ubiquitin-proteasome system (UPS) has attracted tremendous interest since it affects the homeostasis of proteins critically engaged in various cell functions, for example, cell metabolism, survival, proliferation, and differentiation. With their safe and multimodal actions, phytochemicals are among the promising therapeutic tools capable of turning the operation of various UPS elements. The present review, along with an updated outline of the role of UPS dysregulation in multiple cancers, provided a detailed discussion on the impact of phytochemicals on the UPS function in malignancies, especially brain tumors.
Collapse
Affiliation(s)
- Mehdi Sanati
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran
- Experimental and Animal Study Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Amir R Afshari
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
- Department of Physiology and Pharmacology, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Seyed Sajad Ahmadi
- Department of Ophthalmology, Khatam-Ol-Anbia Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Adel Moallem
- Department of Pharmacology and Toxicology, College of Pharmacy, Al-Zahraa University for Women, Karbala, Iraq
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
8
|
AZ12756122, a novel fatty acid synthase inhibitor, decreases resistance features in EGFR-TKI resistant EGFR-mutated NSCLC cell models. Biomed Pharmacother 2022; 156:113942. [DOI: 10.1016/j.biopha.2022.113942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/25/2022] [Accepted: 10/26/2022] [Indexed: 11/22/2022] Open
|
9
|
Dana AH, Alejandro SP. Role of sulforaphane in endoplasmic reticulum homeostasis through regulation of the antioxidant response. Life Sci 2022; 299:120554. [PMID: 35452639 DOI: 10.1016/j.lfs.2022.120554] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 04/10/2022] [Accepted: 04/11/2022] [Indexed: 02/09/2023]
Abstract
Nowadays, the nutraceutical agent sulforaphane (SFN) shows great versatility in turning on different cellular responses. Mainly, this isothiocyanate acts as a master regulator of cellular homeostasis due to its antioxidant response and cytoplasmic, mitochondrial, and endoplasmic reticulum (ER) protein modulation. Even more, SFN acts as an effective strategy to counteract oxidative stress, apoptosis, and ER stress, among others as seen in different injury models. Particularly, ER stress is buffered by the unfolded protein response (UPR) activation, which is the first instance in orchestrating the recovery of ER function. Interestingly, different studies highlight a close interrelationship between ER stress and oxidative stress, two events driven by the accumulation of reactive oxygen species (ROS). This response inevitably perpetuates itself and acts as a vicious cycle that triggers the development of different pathologies, such as cardiovascular diseases, neurodegenerative diseases, and others. Accordingly, it is vital to target ER stress and oxidative stress to increase the effectiveness of clinical therapies used to treat these diseases. Therefore, our study is focused on the role of SFN in preserving cellular homeostasis balance by regulating the ER stress response through the Nrf2-modulated antioxidant pathway.
Collapse
Affiliation(s)
- Arana-Hidalgo Dana
- Department of Cardiovascular Biomedicine, National Institute of Cardiology Ignacio Chávez, Mexico City, Mexico
| | - Silva-Palacios Alejandro
- Department of Cardiovascular Biomedicine, National Institute of Cardiology Ignacio Chávez, Mexico City, Mexico.
| |
Collapse
|
10
|
Zhang Y, Lu Q, Li N, Xu M, Miyamoto T, Liu J. Sulforaphane suppresses metastasis of triple-negative breast cancer cells by targeting the RAF/MEK/ERK pathway. NPJ Breast Cancer 2022; 8:40. [PMID: 35332167 PMCID: PMC8948359 DOI: 10.1038/s41523-022-00402-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 02/22/2022] [Indexed: 12/15/2022] Open
Abstract
Breast cancer metastasis is the main cause of cancer death in women, so far, no effective treatment has inhibited breast cancer metastasis. Sulforaphane (SFN), a natural compound derived from broccoli, has shown potential health benefits in many cancers. However, research on breast cancer metastasis is still insufficient. Here, we showed that SFN, including its two isomers of R-SFN and S-SFN, significantly inhibited TGF-β1-induced migration and invasion in breast cancer cells. Proteomic and phosphoproteomic analysis showed that SFN affected the formation of the cytoskeleton. Subsequent experiments confirmed that SFN significantly inhibited TGF-β1-induced actin stress fiber formation and the expression of actin stress fiber formation-associated proteins, including paxillin, IQGAP1, FAK, PAK2, and ROCK. Additionally, SFN is directly bound to RAF family proteins (including ARAF, BRAF, and CRAF) and inhibited MEK and ERK phosphorylation. These in vitro results indicate that SFN targets the RAF/MEK/ERK signaling pathway to inhibit the formation of actin stress fibers, thereby inhibiting breast cancer cell metastasis.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Molecular and Cellular Physiology, Yamaguchi University, Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan.
| | - Qian Lu
- Department of Molecular and Cellular Physiology, Yamaguchi University, Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Nan Li
- Department of Molecular and Cellular Physiology, Yamaguchi University, Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Ming Xu
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University, Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Tatsuo Miyamoto
- Department of Molecular and Cellular Physiology, Yamaguchi University, Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Jing Liu
- College of Pharmacy, Dalian Medical University, No.9 West Section Lvshun South Road, Dalian, 116044, China.
| |
Collapse
|
11
|
Wang Y, Chen F, Zhang Y, Zheng X, Liu S, Tang M, Wang Z, Wang P, Bao Y, Li D. Biphasic effect of sulforaphane on angiogenesis in hypoxia via modulation of both Nrf2 and mitochondrial dynamics. Food Funct 2022; 13:2884-2898. [PMID: 35179529 DOI: 10.1039/d1fo04112f] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Sulforaphane (SFN) is an isothiocyanate (ITC) derived from a glucosinolate, glucoraphinin found in cruciferous vegetables. There are few studies that focus on the role of SFN in angiogenesis under hypoxic conditions. The effect of SFN on angiogenesis and the underlying mechanisms including the roles of Nrf2 and mitochondrial dynamics were investigated using cultured human umbilical vein endothelial cells (HUVECs) in hypoxia. SFN at low doses (1.25-5 μM) increased hypoxia-induced HUVEC migration and tube formation, and alleviated hypoxia-induced retarded proliferation, but high doses (≥10 μM) exhibited an opposite effect. Under hypoxia, the expression of Nrf2 and heme oxygenase-1 was up-regulated by SFN treatment. Nrf2 knockdown abrogated SFN (2.5 μM)-induced tube formation and further potentiated the inhibitory effect of SFN (10 μM) on angiogenesis. Meanwhile, the mitochondrial function, morphology and expression of dynamic-related proteins suggested that low-dose SFN protected against hypoxia-induced mitochondrial injury and alleviated hypoxia-induced fission Nrf2-dependently without affecting the expression of key effector proteins (Drp1, Fis1, Mfn1/2 and Opa1), while high concentrations (≥10 μM SFN) aggravated hypoxia-induced mitochondrial injury, fission and Drp1 expression, and inhibited Mfn1/2 expression. These findings suggest that SFN biphasically affected the angiogenic capacity of hypoxia challenged HUVECs potentially via mechanisms involving an integrated modulation of Nrf2 and mitochondrial dynamics.
Collapse
Affiliation(s)
- Yaqian Wang
- Department of Nutrition, School of Public Health, Sun Yat-Sen University (Northern Campus), Guangzhou, Guangdong Province 510080, P. R. China. .,Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, Guangdong Province 510080, P. R. China.,Guangdong Engineering Technology Center of Nutrition Transformation, Guangzhou, Guangdong Province 510080, P. R. China
| | - Fangfang Chen
- Department of Nutrition, School of Public Health, Sun Yat-Sen University (Northern Campus), Guangzhou, Guangdong Province 510080, P. R. China. .,Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, Guangdong Province 510080, P. R. China.,Guangdong Engineering Technology Center of Nutrition Transformation, Guangzhou, Guangdong Province 510080, P. R. China
| | - Yuan Zhang
- Department of Geriatrics, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province 510150, P. R. China
| | - Xiangyu Zheng
- Department of Nutrition, School of Public Health, Sun Yat-Sen University (Northern Campus), Guangzhou, Guangdong Province 510080, P. R. China. .,Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, Guangdong Province 510080, P. R. China.,Guangdong Engineering Technology Center of Nutrition Transformation, Guangzhou, Guangdong Province 510080, P. R. China
| | - Shiyan Liu
- Department of Nutrition, School of Public Health, Sun Yat-Sen University (Northern Campus), Guangzhou, Guangdong Province 510080, P. R. China. .,Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, Guangdong Province 510080, P. R. China.,Guangdong Engineering Technology Center of Nutrition Transformation, Guangzhou, Guangdong Province 510080, P. R. China
| | - Meijuan Tang
- Department of Nutrition, School of Public Health, Sun Yat-Sen University (Northern Campus), Guangzhou, Guangdong Province 510080, P. R. China. .,Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, Guangdong Province 510080, P. R. China.,Guangdong Engineering Technology Center of Nutrition Transformation, Guangzhou, Guangdong Province 510080, P. R. China
| | - Ziling Wang
- Department of Nutrition, School of Public Health, Sun Yat-Sen University (Northern Campus), Guangzhou, Guangdong Province 510080, P. R. China. .,Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, Guangdong Province 510080, P. R. China.,Guangdong Engineering Technology Center of Nutrition Transformation, Guangzhou, Guangdong Province 510080, P. R. China
| | - Pan Wang
- Department of Nutrition, School of Public Health, Sun Yat-Sen University (Northern Campus), Guangzhou, Guangdong Province 510080, P. R. China. .,Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, Guangdong Province 510080, P. R. China.,Guangdong Engineering Technology Center of Nutrition Transformation, Guangzhou, Guangdong Province 510080, P. R. China
| | - Yongping Bao
- Norwich Medical School, University of East Anglia, Norwich, Norfolk NR4 7UQ, UK.
| | - Dan Li
- Department of Nutrition, School of Public Health, Sun Yat-Sen University (Northern Campus), Guangzhou, Guangdong Province 510080, P. R. China. .,Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, Guangdong Province 510080, P. R. China.,Guangdong Engineering Technology Center of Nutrition Transformation, Guangzhou, Guangdong Province 510080, P. R. China
| |
Collapse
|
12
|
Niu X, Wu T, Li G, Gu X, Tian Y, Cui H. Insights into the critical role of the PXR in preventing carcinogenesis and chemotherapeutic drug resistance. Int J Biol Sci 2022; 18:742-759. [PMID: 35002522 PMCID: PMC8741843 DOI: 10.7150/ijbs.68724] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 11/21/2021] [Indexed: 12/12/2022] Open
Abstract
Pregnane x receptor (PXR) as a nuclear receptor is well-established in drug metabolism, however, it has pleiotropic functions in regulating inflammatory responses, glucose metabolism, and protects normal cells against carcinogenesis. Most studies focus on its transcriptional regulation, however, PXR can regulate gene expression at the translational level. Emerging evidences have shown that PXR has a broad protein-protein interaction network, by which is implicated in the cross signaling pathways. Furthermore, the interactions between PXR and some critical proteins (e.g., p53, Tip60, p300/CBP-associated factor) in DNA damage pathway highlight its potential roles in this field. A thorough understanding of how PXR maintains genome stability and prevents carcinogenesis will help clinical diagnosis and finally benefit patients. Meanwhile, due to the regulation of CYP450 enzymes CYP3A4 and multidrug resistance protein 1 (MDR1), PXR contributes to chemotherapeutic drug resistance. It is worthy of note that the co-factor of PXR such as RXRα, also has contributions to this process, which makes the PXR-mediated drug resistance more complicated. Although single nucleotide polymorphisms (SNPs) vary between individuals, the amino acid substitution on exon of PXR finally affects PXR transcriptional activity. In this review, we have summarized the updated mechanisms that PXR protects the human body against carcinogenesis, and major contributions of PXR with its co-factors have made on multidrug resistance. Furthermore, we have also reviewed the current promising antagonist and their clinic applications in reversing chemoresistance. We believe our review will bring insight into PXR-targeted cancer therapy, enlighten the future study direction, and provide substantial evidence for the clinic in future.
Collapse
Affiliation(s)
- Xiaxia Niu
- Institute of Toxicology, School of Public Health, Lanzhou University, 730000, Lanzhou, China
| | - Ting Wu
- Institute of Toxicology, School of Public Health, Lanzhou University, 730000, Lanzhou, China
| | - Gege Li
- Institute of Toxicology, School of Public Health, Lanzhou University, 730000, Lanzhou, China
| | - Xinsheng Gu
- Department of Pharmacology, College of Basic Medical Sciences, Hubei University of Medicine, Shiyan 442000, Hubei, China
| | - Yanan Tian
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, USA
| | - Hongmei Cui
- Institute of Toxicology, School of Public Health, Lanzhou University, 730000, Lanzhou, China
| |
Collapse
|
13
|
Yan Y, Zhou Y, Li J, Zheng Z, Hu Y, Li L, Wu W. Sulforaphane downregulated fatty acid synthase and inhibited microtubule-mediated mitophagy leading to apoptosis. Cell Death Dis 2021; 12:917. [PMID: 34620841 PMCID: PMC8497537 DOI: 10.1038/s41419-021-04198-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 08/29/2021] [Accepted: 09/17/2021] [Indexed: 12/19/2022]
Abstract
We previously demonstrated that sulforaphane (SFN) inhibited autophagy leading to apoptosis in human non-small cell lung cancer (NSCLC) cells, but the underlying subcellular mechanisms were unknown. Hereby, high-performance liquid chromatography-tandem mass spectrometry uncovered that SFN regulated the production of lipoproteins, and microtubule- and autophagy-associated proteins. Further, highly expressed fatty acid synthase (FASN) contributed to cancer malignancy and poor prognosis. Results showed that SFN depolymerized microtubules, downregulated FASN, and decreased its binding to α-tubulin; SFN downregulated FASN, acetyl CoA carboxylase (ACACA), and ATP citrate lyase (ACLY) via activating proteasomes and downregulating transcriptional factor SREBP1; SFN inhibited the interactions among α-tubulin and FASN, ACACA, and ACLY; SFN decreased the amount of intracellular fatty acid (FA) and mitochondrial phospholipids; and knockdown of FASN decreased mitochondrial membrane potential (ΔΨm) and increased reactive oxygen species, mitochondrial abnormality, and apoptosis. Further, SFN downregulated mitophagy-associated proteins Bnip3 and NIX, and upregulated mitochondrial LC3 II/I. Transmission electron microscopy showed mitochondrial abnormality and accumulation of mitophagosomes in response to SFN. Combined with mitophagy inducer CCCP or autophagosome–lysosome fusion inhibitor Bafilomycin A1, we found that SFN inhibited mitophagosome–lysosome fusion leading to mitophagosome accumulation. SFN reduced the interaction between NIX and LC3 II/I, and reversed CCCP-caused FA increase. Furthermore, knockdown of α-tubulin downregulated NIX and BNIP3 production, and upregulated LC3 II/I. Besides, SFN reduced the interaction and colocalization between α-tubulin and NIX. Thus, SFN might cause apoptosis via inhibiting microtubule-mediated mitophagy. These results might give us a new insight into the mechanisms of SFN-caused apoptosis in the subcellular level.
Collapse
Affiliation(s)
- Yuting Yan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Beijing Key Laboratory for Tumor Invasion and Metastasis, Beijing, China.,State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Beijing, 100037, China
| | - Yan Zhou
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Beijing Key Laboratory for Tumor Invasion and Metastasis, Beijing, China
| | - Juntao Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Beijing Key Laboratory for Tumor Invasion and Metastasis, Beijing, China
| | - Zhongnan Zheng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Beijing Key Laboratory for Tumor Invasion and Metastasis, Beijing, China
| | - Yabin Hu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Beijing Key Laboratory for Tumor Invasion and Metastasis, Beijing, China
| | - Lei Li
- Central Laboratory, Capital Medical University, Beijing, China.,Capital Medical University, No. 10, Xitoutiao, Beijing, 100069, China
| | - Wei Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China. .,Beijing Key Laboratory for Tumor Invasion and Metastasis, Beijing, China.
| |
Collapse
|
14
|
Sulforaphane: A Broccoli Bioactive Phytocompound with Cancer Preventive Potential. Cancers (Basel) 2021; 13:cancers13194796. [PMID: 34638282 PMCID: PMC8508555 DOI: 10.3390/cancers13194796] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/18/2021] [Accepted: 09/22/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary As of the past decade, phytochemicals have become a major target of interest in cancer chemopreventive and chemotherapeutic research. Sulforaphane (SFN) is a metabolite of the phytochemical glucoraphanin, which is found in high abundance in cruciferous vegetables, such as broccoli, watercress, Brussels sprouts, and cabbage. In both distant and recent research, SFN has been shown to have a multitude of anticancer effects, increasing the need for a comprehensive review of the literature. In this review, we critically evaluate SFN as an anticancer agent and its mechanisms of action based on an impressive number of in vitro, in vivo, and clinical studies. Abstract There is substantial and promising evidence on the health benefits of consuming broccoli and other cruciferous vegetables. The most important compound in broccoli, glucoraphanin, is metabolized to SFN by the thioglucosidase enzyme myrosinase. SFN is the major mediator of the health benefits that have been recognized for broccoli consumption. SFN represents a phytochemical of high interest as it may be useful in preventing the occurrence and/or mitigating the progression of cancer. Although several prior publications provide an excellent overview of the effect of SFN in cancer, these reports represent narrative reviews that focused mainly on SFN’s source, biosynthesis, and mechanisms of action in modulating specific pathways involved in cancer without a comprehensive review of SFN’s role or value for prevention of various human malignancies. This review evaluates the most recent state of knowledge concerning SFN’s efficacy in preventing or reversing a variety of neoplasms. In this work, we have analyzed published reports based on in vitro, in vivo, and clinical studies to determine SFN’s potential as a chemopreventive agent. Furthermore, we have discussed the current limitations and challenges associated with SFN research and suggested future research directions before broccoli-derived products, especially SFN, can be used for human cancer prevention and intervention.
Collapse
|
15
|
Zimmermann M, Kolmar H, Zimmer A. S-Sulfocysteine - Investigation of cellular uptake in CHO cells. J Biotechnol 2021; 335:27-38. [PMID: 34090949 DOI: 10.1016/j.jbiotec.2021.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 05/26/2021] [Accepted: 06/01/2021] [Indexed: 10/21/2022]
Abstract
For the generation of therapeutic proteins in cell culture, high producing clones are used. These clones have a high demand in amino acids to support cell growth and productivity. l-cysteine (Cys) is critical in highly concentrated feeds due to low stability of Cys and low solubility of the oxidation product cystine at neutral pH. S-sulfocysteine (SSC) was developed to substitute the Cys source and fed-batch experiments using SSC showed good cellular performance regarding viable cell density and titer, indicating uptake and metabolization of SSC by Chinese hamster ovary cells. However, the responsible transporter allowing cellular uptake remains unclear and was studied in this work. Due to the structure similarity of SSC with cystine and glutamate, it was proposed that the cystine/glutamate antiporter (xc-) allows cellular uptake of SSC. The uptake was assessed via transporter inhibition using sulfasalazine and transporter overexpression using either sulforaphane or sulforaphane-N-acetylcysteine during fed-batch experiments. Following daily addition of 50 μM and 100 μM sulfasalazine, the extracellular SSC concentration was increased by 65 % and 177 % respectively, suggesting a reduced uptake due to xc- inhibition. In contrast, enhanced transporter activity through 15 μM sulforaphane and sulforaphane-N-acetylcysteine treatment, induced a 60 % and 52 % reduced extracellular SSC concentration, respectively. These inverse uptake results strongly suggest that xc- is facilitating the transport of SSC.
Collapse
Affiliation(s)
- Martina Zimmermann
- Merck Life Science, Upstream R&D, Frankfurter Strasse 250, 64293 Darmstadt, Germany; Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Alarich‑Weiss‑Strasse 4, 64287 Darmstadt, Germany.
| | - Harald Kolmar
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Alarich‑Weiss‑Strasse 4, 64287 Darmstadt, Germany.
| | - Aline Zimmer
- Merck Life Science, Upstream R&D, Frankfurter Strasse 250, 64293 Darmstadt, Germany.
| |
Collapse
|
16
|
Heng WS, Kruyt FAE, Cheah SC. Understanding Lung Carcinogenesis from a Morphostatic Perspective: Prevention and Therapeutic Potential of Phytochemicals for Targeting Cancer Stem Cells. Int J Mol Sci 2021; 22:ijms22115697. [PMID: 34071790 PMCID: PMC8198077 DOI: 10.3390/ijms22115697] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/06/2021] [Accepted: 05/07/2021] [Indexed: 12/24/2022] Open
Abstract
Lung cancer is still one of the deadliest cancers, with over two million incidences annually. Prevention is regarded as the most efficient way to reduce both the incidence and death figures. Nevertheless, treatment should still be improved, particularly in addressing therapeutic resistance due to cancer stem cells—the assumed drivers of tumor initiation and progression. Phytochemicals in plant-based diets are thought to contribute substantially to lung cancer prevention and may be efficacious for targeting lung cancer stem cells. In this review, we collect recent literature on lung homeostasis, carcinogenesis, and phytochemicals studied in lung cancers. We provide a comprehensive overview of how normal lung tissue operates and relate it with lung carcinogenesis to redefine better targets for lung cancer stem cells. Nine well-studied phytochemical compounds, namely curcumin, resveratrol, quercetin, epigallocatechin-3-gallate, luteolin, sulforaphane, berberine, genistein, and capsaicin, are discussed in terms of their chemopreventive and anticancer mechanisms in lung cancer and potential use in the clinic. How the use of phytochemicals can be improved by structural manipulations, targeted delivery, concentration adjustments, and combinatorial treatments is also highlighted. We propose that lung carcinomas should be treated differently based on their respective cellular origins. Targeting quiescence-inducing, inflammation-dampening, or reactive oxygen species-balancing pathways appears particularly interesting.
Collapse
Affiliation(s)
- Win Sen Heng
- Faculty of Medical Sciences, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands; (W.S.H.); (F.A.E.K.)
- Faculty of Medicine and Health Sciences, UCSI University, Kuala Lumpur 56000, Malaysia
| | - Frank A. E. Kruyt
- Faculty of Medical Sciences, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands; (W.S.H.); (F.A.E.K.)
| | - Shiau-Chuen Cheah
- Faculty of Medicine and Health Sciences, UCSI University, Kuala Lumpur 56000, Malaysia
- Correspondence: ; Tel.: +60-3-91018880
| |
Collapse
|
17
|
Dietary isothiocyanates inhibit cancer progression by modulation of epigenome. Semin Cancer Biol 2021; 83:353-376. [PMID: 33434642 DOI: 10.1016/j.semcancer.2020.12.021] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 12/13/2020] [Accepted: 12/27/2020] [Indexed: 12/15/2022]
Abstract
Cell cycle, growth, survival and metabolism are tightly regulated together and failure in cellular regulation leads to carcinogenesis. Several signaling pathways like the PI3K, WNT, MAPK and NFKb pathway exhibit aberrations in cancer and help achieve hallmark capabilities. Clinical research and in vitro studies have highlighted the role of epigenetic alterations in cancer onset and development. Altered gene expression patterns enabled by changes in DNA methylation, histone modifications and RNA processing have proven roles in cancer hallmark acquisition. The reversible nature of epigenetic processes offers robust therapeutic targets. Dietary bioactive compounds offer a vast compendium of effective therapeutic moieties. Isothiocyanates (ITCs) sourced from cruciferous vegetables demonstrate anti-proliferative, pro-apoptotic, anti-inflammatory, anti-migratory and anti-angiogenic effect against several cancers. ITCs also modulate the redox environment, modulate signaling pathways including PI3K, MAPK, WNT, and NFkB. They also modulate the epigenetic machinery by regulating the expression and activity of DNA methyltransferases, histone modifiers and miRNA. This further enhances their transcriptional modulation of key cellular regulators. In this review, we comprehensively assess the impact of ITCs such as sulforaphane, phenethyl isothiocyanate, benzyl isothiocyanate and allyl isothiocyanate on cancer and document their effect on various molecular targets. Overall, this will facilitate consolidation of the current understanding of the anti-cancer and epigenetic modulatory potential of these compounds and recognize the gaps in literature. Further, we discuss avenues of future research to develop these compounds as potential therapeutic entities.
Collapse
|
18
|
Gu HF, Mao XY, Du M. Metabolism, absorption, and anti-cancer effects of sulforaphane: an update. Crit Rev Food Sci Nutr 2021; 62:3437-3452. [PMID: 33393366 DOI: 10.1080/10408398.2020.1865871] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Cancer is one of the most devastating diseases, and recently, a variety of natural compounds with preventive effects on cancer developments have been reported. Sulforaphane (SFN) is a potent anti-cancer isothiocyanate originating from Brassica oleracea (broccoli). SFN, mainly metabolized via mercapturic acid pathway, has high bioavailability and absorption. The present reviews mainly discussed the metabolism and absorption of SFN and newly discovered mechanistic understanding recent years for SFN's anti-cancer effects including promoting autophagy, inducing epigenetic modifications, suppressing glycolysis and fat metabolism. Moreover, its inhibitory effects on cancer stem cells and synergetic effects with other anti-cancer agents are also reviewed along with the clinical trials in this realm.
Collapse
Affiliation(s)
- Hao-Feng Gu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education; College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Xue-Ying Mao
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education; College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Min Du
- Department of Animal Sciences, Washington State University, Pullman, Washington, USA
| |
Collapse
|
19
|
Zhou Y, Wang Y, Wu S, Yan Y, Hu Y, Zheng Z, Li J, Wu W. Sulforaphane-cysteine inhibited migration and invasion via enhancing mitophagosome fusion to lysosome in human glioblastoma cells. Cell Death Dis 2020; 11:819. [PMID: 33004792 PMCID: PMC7530759 DOI: 10.1038/s41419-020-03024-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/13/2020] [Accepted: 09/15/2020] [Indexed: 12/24/2022]
Abstract
Here we uncovered the involved subcellular mechanisms that sulforaphane-cysteine (SFN-Cys) inhibited invasion in human glioblastoma (GBM). SFN-Cys significantly upregulated 45 and downregulated 14 microtubule-, mitophagy-, and invasion-associated proteins in GBM cells via HPLC-MS/MS and GEO ontology analysis; SFN-Cys disrupted microtubule by ERK1/2 phosphorylation-mediated downregulation of α-tubulin and Stathmin-1 leading to the inhibition of cell migration and invasion; SFN-Cys downregulated invasion-associated Claudin-5 and S100A4, and decreased the interaction of α-tubulin to Claudin-5. Knockdown of Claudin-5 and S100A4 significantly reduced the migration and invasion. Besides, SFN-Cys lowered the expressions of α-tubulin-mediated mitophagy-associated proteins Bnip3 and Nix. Transmission electron microscopy showed more membrane-deficient mitochondria and accumulated mitophagosomes in GBM cells, and mitochondria fusion might be downregulated because that SFN-Cys downregulated mitochondrial fusion protein OPA1. SFN-Cys increased the colocalization and interplay of LC3 to lysosomal membrane-associated protein LAMP1, aggravating the fusion of mitophagosome to lysosome. Nevertheless, SFN-Cys inhibited the lysosomal proteolytic capacity causing LC3II/LC3I elevation but autophagy substrate SQSTM1/p62 was not changed, mitophagosome accumulation, and the inhibition of migration and invasion in GBM cells. These results will help us develop high-efficiency and low-toxicity anticancer drugs to inhibit migration and invasion in GBM.
Collapse
Affiliation(s)
- Yan Zhou
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Yalin Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Sai Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Yuting Yan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Yabin Hu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Zhongnan Zheng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Juntao Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Wei Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China. .,Beijing Key Laboratory for Invasion and Metastasis, Capital Medical University, No. 10, Xitoutiao, You An Men Wai Ave., Feng Tai District, Beijing, 100069, China.
| |
Collapse
|
20
|
Li J, Zhou Y, Yan Y, Zheng Z, Hu Y, Wu W. Sulforaphane-cysteine downregulates CDK4 /CDK6 and inhibits tubulin polymerization contributing to cell cycle arrest and apoptosis in human glioblastoma cells. Aging (Albany NY) 2020; 12:16837-16851. [PMID: 32860670 PMCID: PMC7521484 DOI: 10.18632/aging.103537] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 04/27/2020] [Indexed: 01/24/2023]
Abstract
Here we demonstrated that sulforaphane-cysteine (SFN-Cys) regulated cell cycle-related protein expressions in G0/G1 and G2/M phases of U87MG cells via High Performance Liquid Chromatography-Mass Spectrometry/Mass Spectrometry (HPLC-MS/MS) and proteomics analysis. Further, mRNA products of CDK4, CDK6 and α-tubulin were significantly higher in glioblastoma than those in normal tissues, and these results were significantly correlated to pathological grades and clinical prognosis via analyzing TCGA and CGGA databases. Furthermore, Western blot showed that SFN-Cys downregulated CDK4, CDK6 and p-Rb in a dose-dependent manner and these results were reversed by p-ERK1/2 blocker PD98059 in U87MG and U373MG cells. The reductions of CDK4, CDK6 and p-Rb were reversed by proteasome inhibitor MG132; similarly, the upregulation of 26S proteasome by SFN-Cys was reversed by PD98059. Interestingly, SFN-Cys decreased CDK4 and CDK6 by phosphorylated ERK1/2-caused proteasomal degradation resulting in decreased Rb phosphorylation contributing to cell cycle arrest in G0/G1 phase. Besides, Western blot showed that SFN-Cys downregulated α-tubulin resulting in microtubule disruption and aggregation, and cell cycle arrest in G2/M phase and apoptosis. These results might help us understand the molecular etiology of glioblastoma progression to establish brand-new anti-cancer therapies.
Collapse
Affiliation(s)
- Juntao Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China,Beijing Key Laboratory for Tumor Invasion and Metastasis, Capital Medical University, Beijing, China
| | - Yan Zhou
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China,Beijing Key Laboratory for Tumor Invasion and Metastasis, Capital Medical University, Beijing, China
| | - Yuting Yan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China,Beijing Key Laboratory for Tumor Invasion and Metastasis, Capital Medical University, Beijing, China
| | - Zhongnan Zheng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China,Beijing Key Laboratory for Tumor Invasion and Metastasis, Capital Medical University, Beijing, China
| | - Yabin Hu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China,Beijing Key Laboratory for Tumor Invasion and Metastasis, Capital Medical University, Beijing, China
| | - Wei Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China,Beijing Key Laboratory for Tumor Invasion and Metastasis, Capital Medical University, Beijing, China
| |
Collapse
|
21
|
Wei J, Liu J, Zhang L, Zhu Y, Li X, Zhou G, Zhao Y, Sun Z, Zhou X. Endosulfan induces cardiotoxicity through apoptosis via unbalance of pro-survival and mitochondrial-mediated apoptotic pathways. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 727:138790. [PMID: 32344260 DOI: 10.1016/j.scitotenv.2020.138790] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 03/31/2020] [Accepted: 04/16/2020] [Indexed: 06/11/2023]
Abstract
Although the associations between endosulfan and adverse cardiovascular health have been reported, the toxic effects and underlying mechanism of endosulfan on the heart are not well understood. In this study, we examined the cardiotoxicity induced by endosulfan using Wistar rats and human cardiomyocytes (AC16) cells. Wistar rats were divided into control group (received corn oil alone) and three concentrations of endosulfan groups (1, 5 and 10 mg/kg·bw) by gavage. The AC16 cells were treated with three various concentrations (0, 1.25, 5, and 20 μg/mL) of endosulfan. The results showed that endosulfan induced cytotoxicity through damaging myocardial structure, decreasing the viability of cardiomyocytes, and elevating the serum levels of cardiac troponin I, heart fatty acid binding protein, aspartate aminotransferase, and reactive oxygen species (p < 0.05). Moreover, measurement of mitochondrial function showed that endosulfan could significantly decrease adenosine triphosphate levels and cytochrome c oxidase IV expression in AC16 cells (p < 0.05). In addition, endosulfan obviously inhibited Bcl-2 expression, activated the expressions of cytochrome c/Caspase-9/Caspase-3 signaling pathway, and induced the apoptosis of AC16 cells (p < 0.05). Furthermore, endosulfan significantly increased the expression of Bim, and inhibited the expressions of PI3K/Akt/FoxO3a signaling pathways in cardiomyocytes (p < 0.05). These results suggest that endosulfan may induce cardiotoxicity by inducing myocardial apoptosis resulting from activation of mitochondria-mediated apoptosis pathway and inhibition of pro-survival signaling pathways, which might be helpful in elucidating the mechanism of cardiac dysfunction induced by endosulfan.
Collapse
Affiliation(s)
- Jialiu Wei
- Key Laboratory of Cardiovascular Epidemiology & Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, China
| | - Jianhui Liu
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, China
| | - Lianshuang Zhang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, China
| | - Yupeng Zhu
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, China
| | - Xiangyang Li
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, China
| | - Guiqing Zhou
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, China
| | - Yanzhi Zhao
- Yanjing Medical College, Capital Medical University, Beijing, China.
| | - Zhiwei Sun
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, China
| | - Xianqing Zhou
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, China.
| |
Collapse
|
22
|
Santos PW, Machado ART, De Grandis R, Ribeiro DL, Tuttis K, Morselli M, Aissa AF, Pellegrini M, Antunes LMG. Effects of sulforaphane on the oxidative response, apoptosis, and the transcriptional profile of human stomach mucosa cells in vitro. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2020; 854-855:503201. [PMID: 32660825 DOI: 10.1016/j.mrgentox.2020.503201] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 05/14/2020] [Accepted: 05/15/2020] [Indexed: 11/25/2022]
Abstract
Oxidative stress is a critical factor in the pathogenesis of several gastrointestinal diseases. Sulforaphane (SFN), a bioactive compound found in cruciferous vegetables, activates the redox-sensitive nuclear erythroid 2-related factor 2 (NRF2). In addition to its protective role, SFN exerts cytotoxic effects on cancer cells. However, there is a lack of information concerning the toxicity of SFN in normal cells. We investigated the effects of SFN on cell viability, antioxidant defenses, and gene expression in human stomach mucosa cells (MNP01). SFN reduced ROS formation and protected the cells against induced oxidative stress but high concentrations increased apoptosis. An intermediate SFN concentration (8 μM) was chosen for RNA sequencing studies. We observed upregulation of genes of the NRF2 (antioxidant) pathway, the DNA damage response, and apoptosis signaling; whereas SFN downregulated cell cycle and DNA repair pathway genes. SFN may be cytoprotective at low concentrations and cytotoxic at high concentrations.
Collapse
Affiliation(s)
- Patrick Wellington Santos
- Department of Clinical Analysis, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, SP, Brazil
| | - Ana Rita Thomazela Machado
- Department of Clinical Analysis, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, SP, Brazil
| | - Rone De Grandis
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, SP, Brazil
| | - Diego Luis Ribeiro
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, SP, Brazil
| | - Katiuska Tuttis
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, SP, Brazil
| | - Marco Morselli
- Department of Molecular, Cell, and Developmental Biology, University of California at Los Angeles, CA, USA
| | - Alexandre Ferro Aissa
- Department of Clinical Analysis, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, SP, Brazil
| | - Matteo Pellegrini
- Department of Molecular, Cell, and Developmental Biology, University of California at Los Angeles, CA, USA
| | - Lusânia Maria Greggi Antunes
- Department of Clinical Analysis, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, SP, Brazil.
| |
Collapse
|
23
|
Rai R, Gong Essel K, Mangiaracina Benbrook D, Garland J, Daniel Zhao Y, Chandra V. Preclinical Efficacy and Involvement of AKT, mTOR, and ERK Kinases in the Mechanism of Sulforaphane against Endometrial Cancer. Cancers (Basel) 2020; 12:E1273. [PMID: 32443471 PMCID: PMC7281543 DOI: 10.3390/cancers12051273] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 05/14/2020] [Accepted: 05/14/2020] [Indexed: 12/12/2022] Open
Abstract
Sulforaphane exerts anti-cancer activity against multiple cancer types. Our objective was to evaluate utility of sulforaphane for endometrial cancer therapy. Sulforaphane reduced viability of endometrial cancer cell lines in association with the G2/M cell cycle arrest and cell division cycle protein 2 (Cdc2) phosphorylation, and intrinsic apoptosis. Inhibition of anchorage-independent growth, invasion, and migration of the cell lines was associated with sulforaphane-induced alterations in epithelial-to-mesenchymal transition (EMT) markers of increased E-cadherin and decreased N-cadherin and vimentin expression. Proteomic analysis identified alterations in AKT, mTOR, and ERK kinases in the networks of sulforaphane effects in the Ishikawa endometrial cancer cell line. Western blots confirmed sulforaphane inhibition of AKT, mTOR, and induction of ERK with alterations in downstream signaling. AKT and mTOR inhibitors reduced endometrial cancer cell line viability and prevented further reduction by sulforaphane. Accumulation of nuclear phosphorylated ERK was associated with reduced sensitivity to the ERK inhibitor and its interference with sulforaphane activity. Sulforaphane induced apoptosis-associated growth inhibition of Ishikawa xenograft tumors to a greater extent than paclitaxel, with no evidence of toxicity. These results verify sulforaphane's potential as a non-toxic treatment candidate for endometrial cancer and identify AKT, mTOR, and ERK kinases in the mechanism of action with interference in the mechanism by nuclear phosphorylated ERK.
Collapse
Affiliation(s)
- Rajani Rai
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (R.R.); (D.M.B.); (J.G.)
| | - Kathleen Gong Essel
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
| | - Doris Mangiaracina Benbrook
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (R.R.); (D.M.B.); (J.G.)
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
| | - Justin Garland
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (R.R.); (D.M.B.); (J.G.)
| | - Yan Daniel Zhao
- Biostatistics & Epidemiology, College of Public Health University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
| | - Vishal Chandra
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (R.R.); (D.M.B.); (J.G.)
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
| |
Collapse
|
24
|
Vekaria M, Tirgar P. Promising Anticancer Potential of Herbal Compounds against Breast Cancer: A Systemic Review. ASIAN JOURNAL OF PHARMACEUTICAL RESEARCH AND HEALTH CARE 2020. [DOI: 10.18311/ajprhc/2021/26698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
25
|
Sulforaphane-Loaded Ultradeformable Vesicles as A Potential Natural Nanomedicine for the Treatment of Skin Cancer Diseases. Pharmaceutics 2019; 12:pharmaceutics12010006. [PMID: 31861672 PMCID: PMC7023209 DOI: 10.3390/pharmaceutics12010006] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 12/09/2019] [Accepted: 12/17/2019] [Indexed: 12/31/2022] Open
Abstract
Sulforaphane is a multi-action drug and its anticancer activity is the reason for the continuous growth of attention being paid to this drug. Sulforaphane shows an in vitro antiproliferative activity against melanoma and other skin cancer diseases. Unfortunately, this natural compound cannot be applied in free form on the skin due to its poor percutaneous permeation determined by its physico-chemical characteristics. The aim of this investigation was to evaluate ethosomes® and transfersomes® as ultradeformable vesicular carriers for the percutaneous delivery of sulforaphane to be used for the treatment of skin cancer diseases. The physico-chemical features of the ultradeformable vesicles were evaluated. Namely, ethosomes® and transfersomes® had mean sizes <400 nm and a polydispersity index close to 0. The stability studies demonstrated that the most suitable ultradeformable vesicles to be used as topical carriers of sulforaphane were ethosomes® made up of ethanol 40% (w/v) and phospholipon 90G 2% (w/v). In particular, in vitro studies of percutaneous permeation through human stratum corneum and epidermis membranes showed an increase of the percutaneous permeation of sulforaphane. The antiproliferative activity of sulforaphane-loaded ethosomes® was tested on SK-MEL 28 and improved anticancer activity was observed in comparison with the free drug.
Collapse
|
26
|
Bayat Mokhtari R, Baluch N, Morgatskaya E, Kumar S, Sparaneo A, Muscarella LA, Zhao S, Cheng HL, Das B, Yeger H. Human bronchial carcinoid tumor initiating cells are targeted by the combination of acetazolamide and sulforaphane. BMC Cancer 2019; 19:864. [PMID: 31470802 PMCID: PMC6716820 DOI: 10.1186/s12885-019-6018-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 08/06/2019] [Indexed: 12/19/2022] Open
Abstract
Background Bronchial carcinoids are neuroendocrine tumors that present as typical (TC) and atypical (AC) variants, the latter being more aggressive, invasive and metastatic. Studies of tumor initiating cell (TIC) biology in bronchial carcinoids has been hindered by the lack of appropriate in-vitro and xenograft models representing the bronchial carcinoid phenotype and behavior. Methods Bronchial carcinoid cell lines (H727, TC and H720, AC) were cultured in serum-free growth factor supplemented medium to form 3D spheroids and serially passaged up to the 3rd generation permitting expansion of the TIC population as verified by expression of stemness markers, clonogenicity in-vitro and tumorigenicity in both subcutaneous and orthotopic (lung) models. Acetazolamide (AZ), sulforaphane (SFN) and the AZ + SFN combination were evaluated for targeting TIC in bronchial carcinoids. Results Data demonstrate that bronchial carcinoid cell line 3rd generation spheroid cells show increased drug resistance, clonogenicity, and tumorigenic potential compared with the parental cells, suggesting selection and expansion of a TIC fraction. Gene expression and immunolabeling studies demonstrated that the TIC expressed stemness factors Oct-4, Sox-2 and Nanog. In a lung orthotopic model bronchial carcinoid, cell line derived spheroids, and patient tumor derived 3rd generation spheroids when supported by a stroma, showed robust tumor formation. SFN and especially the AZ + SFN combination were effective in inhibiting tumor cell growth, spheroid formation and in reducing tumor formation in immunocompromised mice. Conclusions Human bronchial carcinoid tumor cells serially passaged as spheroids contain a higher fraction of TIC exhibiting a stemness phenotype. This TIC population can be effectively targeted by the combination of AZ + SFN. Our work portends clinical relevance and supports the therapeutic use of the novel AZ+ SFN combination that may target the TIC population of bronchial carcinoids.
Collapse
Affiliation(s)
- Reza Bayat Mokhtari
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON, Canada. .,Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, ON, Canada. .,Institute of Medical Science, University of Toronto, Toronto, ON, Canada. .,The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, 686 Bay St., Rm 15.9714, Toronto, Ontario, M5G 0A4, Canada.
| | - Narges Baluch
- Department of Pediatrics, Queen's University, 76 Stuart St, Kingston, ON, K7L 2V7, Canada
| | - Evgeniya Morgatskaya
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Sushil Kumar
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Williams Science Hall 3035, Department of Pharmaceutical Sciences 601 S. Saddle Creek Rd, Omaha, NE, 68106, USA
| | - Angelo Sparaneo
- Laboratory of Oncology, Fondazione IRCCS Casa Sollievo della Sofferenza, viale Cappuccini, 71013, San Giovanni Rotondo, FG, Italy
| | - Lucia Anna Muscarella
- Laboratory of Oncology, Fondazione IRCCS Casa Sollievo della Sofferenza, viale Cappuccini, 71013, San Giovanni Rotondo, FG, Italy
| | - Sheyun Zhao
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Hai-Ling Cheng
- Institute of Biomaterials & Biomedical Engineering, University of Toronto, 164 College Street, Rosebrugh Building, Room 407, Toronto, ON, M5S 3G9, Canada
| | - Bikul Das
- Thoreau Laboratory for Global Health, M2D2, University of Massachusetts-Lowell, Innovation Hub, 110 Canal St, Lowell, MA, 01852, USA.,KaviKrishna Laboratory, Indian Institute of Technology Complex, Guwahati, India
| | - Herman Yeger
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON, Canada.,Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, ON, Canada.,Institute of Medical Science, University of Toronto, Toronto, ON, Canada.,The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, 686 Bay St., Rm 15.9714, Toronto, Ontario, M5G 0A4, Canada
| |
Collapse
|
27
|
Sulforaphane metabolites inhibit migration and invasion via microtubule-mediated Claudins dysfunction or inhibition of autolysosome formation in human non-small cell lung cancer cells. Cell Death Dis 2019; 10:259. [PMID: 30874545 PMCID: PMC6420664 DOI: 10.1038/s41419-019-1489-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 02/26/2019] [Accepted: 02/27/2019] [Indexed: 12/11/2022]
Abstract
Both sulforaphane-cysteine (SFN-Cys) and sulforaphane-N-acetyl-l-cysteine (SFN-NAC) inhibited cancer migration and invasion, but the underlying mechanisms were not clear. Here we uncovered via tissue microarray assay that high expression of invasion-associated Claudin-5 was correlated to malignant grades in human non-small cell lung cancer (NSCLC) cells. Further, SFN-Cys (10 µM) induced the accumulated phosphorylation of ERK1/2, leading to downregulation of Claudin-5 and upregulation of Claudin-7, and the decrease of Claudin-1 in SK-1 cells and increase of Claudin-1 in A549 cells; knockdown of Claudin-5 significantly reduced invasion, whereas knockdown of Claudin-7 increased invasion; knockdown of Claudin-1 reduced invasion in SK-1 cells, whereas it increased invasion in A549 cells, indicating that SFN-Cys regulated Claudins and inhibited invasion depending on Claudin isotypes and cell types. Furthermore, immunofluorescence staining showed that SFN-Cys triggered microtubule disruption and knockdown of α-tubulin downregulated Claudin-1, 5, and 7, and inhibited migration and invasion, indicating that microtubule disruption contributed to invasive inhibition. Co-immunoprecipitation and confocal microscopy observation showed that SFN-Cys lowered the interaction between α-tubulin and Claudin-1 or 5, or 7. Meanwhile, Western blotting and immunofluorescence staining showed that SFN-NAC (15 µM) downregulated α-tubulin resulting in microtubule disruption; knockdown of α-tubulin increased SFN-NAC-induced LC3 II accumulation in SK-1 cells. Combined with the inhibitor of autolysosome formation, Bafilomycin A1 (100 nM), SFN-NAC inhibited invasion via accumulating LC3 II and blocking formation of autolysosome. Further, SFN-NAC upregulated microtubule-stabilizing protein Tau; knockdown of Tau reduced LC3 II/LC3 I inhibiting migration and invasion. These results indicated that SFN-Cys inhibited invasion via microtubule-mediated Claudins dysfunction, but SFN-NAC inhibited invasion via microtubule-mediated inhibition of autolysosome formation in human NSCLC cells.
Collapse
|
28
|
Wang Y, Zhou Y, Zheng Z, Li J, Yan Y, Wu W. Sulforaphane metabolites reduce resistance to paclitaxel via microtubule disruption. Cell Death Dis 2018; 9:1134. [PMID: 30429459 PMCID: PMC6235886 DOI: 10.1038/s41419-018-1174-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 10/23/2018] [Accepted: 10/24/2018] [Indexed: 12/16/2022]
Abstract
Long treatment with paclitaxel (PTX) might increase resistance and side-effects causing a failure in cancer chemotherapy. Here we uncovered that either sulforaphane-cysteine (SFN-Cys) or sulforaphane-N-acetyl-cysteine (SFN-NAC) induced apoptosis via phosphorylated ERK1/2-mediated upregulation of 26 S proteasome and Hsp70, and downregulation of βIII-tubulin, XIAP, Tau, Stathmin1 and α-tubulin causing microtubule disruption in human PTX-resistant non-small cell lung cancer (NSCLC) cells. Knockdown of either βIII-tubulin or α-tubulin via siRNA increased cell sensitivity to PTX, indicating that these two proteins help cells increase the resistance. Tissue microarray analysis showed that overexpression of βIII-tubulin correlated to NSCLC malignant grading. Immunofluorescence staining also showed that SFN metabolites induced a nest-like microtubule protein distribution with aggregation and disruption. Co-immunoprecipitation showed that SFN metabolites reduced the interaction between βIII-tubulin and Tau, and that between α-tubulin and XIAP. The combination of PTX with SFN metabolites decreased the resistance to PTX, and doses of both PTX and SFN metabolites, and enhanced apoptosis resulting from activated Caspase-3-caused microtubule degradation. Importantly, the effective dose of SFN metabolites combined with 20 nM PTX will be low to 4 μM. Thus, we might combine SFN metabolites with PTX for preclinical trial. Normally, more than 20 μM SFN metabolites only leading to apoptosis for SFN metabolites hindered their applications. These findings will help us develop a low-resistance and high-efficiency chemotherapy via PTX/SFN metabolites combination.
Collapse
Affiliation(s)
- Yalin Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, P.R. China
- Beijing Key Laboratory of Tumor Invasion and Metastasis Research, Capital Medical University, Beijing, P.R. China
| | - Yan Zhou
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, P.R. China
- Beijing Key Laboratory of Tumor Invasion and Metastasis Research, Capital Medical University, Beijing, P.R. China
| | - Zhongnan Zheng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, P.R. China
- Beijing Key Laboratory of Tumor Invasion and Metastasis Research, Capital Medical University, Beijing, P.R. China
| | - Juntao Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, P.R. China
- Beijing Key Laboratory of Tumor Invasion and Metastasis Research, Capital Medical University, Beijing, P.R. China
| | - Yuting Yan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, P.R. China
- Beijing Key Laboratory of Tumor Invasion and Metastasis Research, Capital Medical University, Beijing, P.R. China
| | - Wei Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, P.R. China.
- Beijing Key Laboratory of Tumor Invasion and Metastasis Research, Capital Medical University, Beijing, P.R. China.
- Institute of Brain Tumor, Beijing Institute for Brain Disorders, Capital Medical University, No. 10, Xitoutiao, You An Men Wai Ave., Feng Tai District, Beijing, 100069, P.R. China.
| |
Collapse
|
29
|
Briones-Herrera A, Eugenio-Pérez D, Reyes-Ocampo JG, Rivera-Mancía S, Pedraza-Chaverri J. New highlights on the health-improving effects of sulforaphane. Food Funct 2018; 9:2589-2606. [PMID: 29701207 DOI: 10.1039/c8fo00018b] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In this paper, we review recent evidence about the beneficial effects of sulforaphane (SFN), which is the most studied member of isothiocyanates, on both in vivo and in vitro models of different diseases, mainly diabetes and cancer. The role of SFN on oxidative stress, inflammation, and metabolism is discussed, with emphasis on those nuclear factor E2-related factor 2 (Nrf2) pathway-mediated mechanisms. In the case of the anti-inflammatory effects of SFN, the point of convergence seems to be the downregulation of the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), with the consequent amelioration of other pathogenic processes such as hypertrophy and fibrosis. We emphasized that SFN shows opposite effects in normal and cancer cells at many levels; for instance, while in normal cells it has protective actions, in cancer cells it blocks the induction of factors related to the malignity of tumors, diminishes their development, and induces cell death. SFN is able to promote apoptosis in cancer cells by many mechanisms, the production of reactive oxygen species being one of the most relevant ones. Given its properties, SFN could be considered as a phytochemical at the forefront of natural medicine.
Collapse
Affiliation(s)
- Alfredo Briones-Herrera
- Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | | | | | | | | |
Collapse
|
30
|
Bernkopf DB, Daum G, Brückner M, Behrens J. Sulforaphane inhibits growth and blocks Wnt/β-catenin signaling of colorectal cancer cells. Oncotarget 2018; 9:33982-33994. [PMID: 30338040 PMCID: PMC6188060 DOI: 10.18632/oncotarget.26125] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 09/06/2018] [Indexed: 12/18/2022] Open
Abstract
The naturally occurring isothiocyanate sulforaphane (SFN) from cruciferous vegetables is associated with growth inhibition of various cancer types, including colorectal cancer. Colorectal cancer is most frequently driven by hyperactive Wnt/β-catenin signaling. Here, we show that SFN treatment reduced growth of three unrelated colorectal cancer cell lines (SW480, DLD1 and HCT116) via induction of cell death and inhibition of proliferation. Importantly, SFN inhibits Wnt/β-catenin signaling in colorectal cancer cells as shown by inhibition of β-catenin-dependent luciferase reporters and repression of β-catenin target genes (AXIN2, LGR5). SFN inhibits Wnt signaling downstream of β-catenin degradation and induces the formation of nuclear β-catenin structures associated with closed chromatin. Co-expression of the transcription factors LEF1 or TCF4 prevented formation of these structures and rescued inhibition of Wnt/β-catenin signaling by SFN. Our findings provide a molecular basis explaining SFN effects in colorectal cancer cells and underline its potential for prevention and therapy of colorectal cancer.
Collapse
Affiliation(s)
- Dominic B Bernkopf
- Experimental Medicine II, Nikolaus-Fiebiger-Center, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Gabriele Daum
- Experimental Medicine II, Nikolaus-Fiebiger-Center, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Martina Brückner
- Experimental Medicine II, Nikolaus-Fiebiger-Center, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Jürgen Behrens
- Experimental Medicine II, Nikolaus-Fiebiger-Center, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| |
Collapse
|
31
|
Kwon Y. Food-derived polyphenols inhibit the growth of ovarian cancer cells irrespective of their ability to induce antioxidant responses. Heliyon 2018; 4:e00753. [PMID: 30186979 PMCID: PMC6121158 DOI: 10.1016/j.heliyon.2018.e00753] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 07/16/2018] [Accepted: 08/22/2018] [Indexed: 11/12/2022] Open
Abstract
The use of plant polyphenols to prevent cancer has been studied extensively. However, recent findings regarding the cancer-promoting effects of some antioxidants have led to reservations regarding the therapeutic use of food-derived antioxidants including polyphenols. The aim of this study was to evaluate the therapeutic potential of food-derived polyphenols and their use and safety in cancer patients. The free-radical scavenging ability of sulforaphane and various food-derived polyphenols including curcumin, epigallocatechin gallate, epicatechin, pelargonidin, and resveratrol was compared with their growth inhibitory effect on ovarian cancer cells. Oxidative stress and/or antioxidant responses and anti-proliferative pathways were evaluated after administering sulforaphane and polyphenols at doses at which they have been shown to inhibit the growth of ovarian cancer cells. No correlation was observed between their ability to scavenge free radicals and their ability to inhibit the growth of ovarian cancer cells. With the exception of epigallocatechin gallate, all of the antioxidants that were tested at doses that inhibited cell growth significantly increased NAD(P)H quinone dehydrogenase I (NQO1) expression but induced cell cycle arrest and/or apoptotic signaling. Epigallocatechin gallate exhibited a higher free radical scavenging activity but did not induce NQO1 expression at either the mRNA or at the protein level. Treatment with polyphenols at physiological doses did not significantly alter the growth of ovarian cancer cells or NQO1 expression. Therefore, individual food-derived polyphenols appear to have different anti-cancer mechanisms. Their modes of action in relation to their chemical properties should be established, rather than collectively avoiding the use of these agents as antioxidants.
Collapse
Affiliation(s)
- Youngjoo Kwon
- Department of Food Science and Engineering, Ewha Womans University, Seoul, South Korea
| |
Collapse
|
32
|
Xie Y, Wang X. Lycium barbarum polysaccharides attenuates the apoptosis of hippocampal neurons induced by sevoflurane. Exp Ther Med 2018; 16:1834-1840. [PMID: 30186408 PMCID: PMC6122330 DOI: 10.3892/etm.2018.6426] [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: 10/24/2017] [Accepted: 03/21/2018] [Indexed: 01/29/2023] Open
Abstract
Following the application of inhalational anesthetics, including sevoflurane, patients may suffer from neural injury. The present study was conducted to explore the mechanism involved in Lycium barbarum polysaccharides (LBP) treatment of sevoflurane injured hippocampal neurons. Primary hippocampal neurons were isolated from Sprague Dawley embryonic rats. The Cell Counting Kit-8 (CCK-8) assay was used to detect cell viability. Furthermore, flow cytometry (FCM) was used to determine cell proliferation and apoptosis rates. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and western blot analysis were applied to detect the expression levels of apoptosis-related factors, including activated-Caspase-3, B-cell lymphoma/leukemia-2 (Bcl-2) and Bcl-2 associated X (Bax), phosphorylated extracellular signal-regulated kinase 1/2 (p-ERK1/2) and total ERK1/2. The results showed that LBP promoted cell viability and cell proliferation but inhibited cell apoptosis in neurons injured with 3% sevoflurane, in dose-dependent manners (100, 200 and 400 µg/ml). LBP increased the expression levels of Bcl-2 and p-ERK1/2, and decreased levels of activated-Caspase-3 and Bax in a dose-dependent manner in hippocampal neurons that were injured with sevoflurane. In addition, ERK1/2 inhibitor reversed the above phenomenon in 400 µg/ml LBP and 3% sevoflurane-treated hippocampal neurons. Therefore, the present study indicated that LBP protected hippocampal neurons from sevoflurane injury, including aberrant cell apoptosis, via the ERK1/2 pathway.
Collapse
Affiliation(s)
- Yuhai Xie
- Department of Anesthesiology, Qinghai Red Cross Hospital, Xining, Qinghai 810000, P.R. China
| | - Xuejun Wang
- Department of Anesthesiology, Qinghai Red Cross Hospital, Xining, Qinghai 810000, P.R. China
| |
Collapse
|
33
|
Poly-L-Arginine Induces Apoptosis of NCI-H292 Cells via ERK1/2 Signaling Pathway. J Immunol Res 2018; 2018:3651743. [PMID: 30013990 PMCID: PMC6022307 DOI: 10.1155/2018/3651743] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 04/22/2018] [Accepted: 05/06/2018] [Indexed: 12/31/2022] Open
Abstract
Cationic protein is a cytotoxic protein secreted by eosinophils and takes part in the damage of airway epithelium in asthma. Poly-L-arginine (PLA), a synthetic cationic protein, is widely used to mimic the biological function of the natural cationic protein in vitro. Previous studies demonstrated the damage of the airway epithelial cells by cationic protein, but the molecular mechanism is unclear. The purpose of this study aimed at exploring whether PLA could induce apoptosis of human airway epithelial cells (NCI-H292) and the underlying mechanism. Methods. The morphology of apoptotic cells was observed by transmission electron microscopy. The rate of apoptosis was analyzed by flow cytometry (FCM). The expressions of the phosphorylation of extracellular signal-regulated kinase 1/2 (ERK1/2), Bcl-2/Bax, and cleaved caspase-3 were assessed by western blot. Results. PLA can induce apoptosis in NCI-H292 cells in a concentration-dependent manner. Moreover, the phosphorylation of the ERK1/2 and the unbalance of Bcl2/Bax, as well as the activation of caspase-3, were involved in the PLA-induced apoptosis. Conclusions. PLA can induce the apoptosis in NCI-H292 cells, and this process at least involved the ERK1/2 and mitochondrial pathway. The results could have some indications in revealing the apoptotic damage of the airway epithelial cells. Besides, inhibition of cationic protein-induced apoptotic death in airway epithelial cells could be considered as a potential target of anti-injury or antiremodeling in asthmatics.
Collapse
|
34
|
Zhang Z, Bergan R, Shannon J, Slatore CG, Bobe G, Takata Y. The Role of Cruciferous Vegetables and Isothiocyanates for Lung Cancer Prevention: Current Status, Challenges, and Future Research Directions. Mol Nutr Food Res 2018; 62:e1700936. [PMID: 29663679 DOI: 10.1002/mnfr.201700936] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Revised: 04/06/2018] [Indexed: 01/07/2023]
Abstract
Lung cancer remains a leading cause of cancer-related deaths in the United States. Although smoking and air pollution exposure are primary risk factors of lung cancer, diet has also been reported to contribute to lung cancer risk. Cruciferous vegetables contain many bioactive compounds that alter the detoxification process of air-borne carcinogenic compounds and, thereby, may decrease lung cancer risk. In the meta-analysis of 31 observational studies, cruciferous vegetable intake is inversely associated with lung cancer risk (summary odds ratio/relative risk = 0.81 and 95% confidence interval = 0.74-0.89 for comparing the highest with lowest intake categories). More observational studies need to measure urinary isothiocyanate (ITC) concentrations and investigate their association with lung cancer risk in populations with relatively high intake of cruciferous vegetables. Current evidence is limited to two phase 2 clinical trials with incomplete reporting. Hence, more short-term clinical phase 2 trials need to examine effects of various amounts and types of cruciferous vegetables on biomarkers of risk and efficacy before a large phase 3 trial can be conducted to assess effects upon lung cancer risk. This would help further elucidate whether the inverse association observed with self-reported cruciferous vegetable intake is indeed due to ITC content or other bioactive compounds.
Collapse
Affiliation(s)
- Zhenzhen Zhang
- OHSU-PSU School of Public Health, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Raymond Bergan
- Division of Hematology/Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Jackilen Shannon
- OHSU-PSU School of Public Health, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Christopher G Slatore
- Health Services Research & Development and Section of Pulmonary & Critical Care Medicine, VA Portland Health Care System, Portland, OR, 97239, USA.,Department of Medicine and Department of Radiation Medicine, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Gerd Bobe
- Linus Pauling Institute, Department of Animal Sciences, Oregon State University, Corvallis, OR, 97331, USA
| | - Yumie Takata
- College of Public Health and Human Sciences, School of Biological and Population Sciences, Oregon State University, Corvallis, OR, 97331, USA
| |
Collapse
|
35
|
Protective Effects of Sulforaphane on Cognitive Impairments and AD-like Lesions in Diabetic Mice are Associated with the Upregulation of Nrf2 Transcription Activity. Neuroscience 2018; 381:35-45. [DOI: 10.1016/j.neuroscience.2018.04.017] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Revised: 04/06/2018] [Accepted: 04/11/2018] [Indexed: 11/19/2022]
|
36
|
Jin CY, Molagoda IMN, Karunarathne WAHM, Kang SH, Park C, Kim GY, Choi YH. TRAIL attenuates sulforaphane-mediated Nrf2 and sustains ROS generation, leading to apoptosis of TRAIL-resistant human bladder cancer cells. Toxicol Appl Pharmacol 2018; 352:132-141. [PMID: 29792947 DOI: 10.1016/j.taap.2018.05.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 05/08/2018] [Accepted: 05/20/2018] [Indexed: 12/12/2022]
Abstract
Tumor necrosis factor-related apoptosis inducing ligand (TRAIL) can preferentially initiate apoptosis in malignant cells with minimal toxicity to normal cells. Unfortunately, many human cancer cells are refractory to TRAIL-induced apoptosis through many unknown mechanisms. Here, we report that TRAIL resistance can be reversed in human bladder cancer cell lines by treatment with sulforaphane (SFN), a well-known chemopreventive isothiocyanate in various cruciferous vegetables. Combined treatment with SFN and TRAIL (SFN/TRAIL) significantly induced apoptosis concomitant with activation of caspases, loss of mitochondrial membrane potential (MMP), Bid truncation, and induction of death receptor 5. Transient knockdown of Bid prevented collapse of MMP induced by SFN/TRAIL, consequently reducing apoptotic effects. Furthermore, SFN increased both the generation of reactive oxygen species (ROS) and the activation of nuclear factor erythroid 2-related factor 2 (Nrf2), which is an anti-oxidant enzyme. Interestingly, TRAIL effectively suppressed SFN-mediated nuclear translocation of Nrf2, and the period of ROS generation was more extended compared to that of treatment with SFN alone. In addition, silencing of Nrf2 increased apoptosis in cells treated with SFN/TRAIL; however, blockade of ROS generation inhibited apoptotic activity. These data suggest that SFN-induced ROS generation promotes TRAIL sensitivity and SFN can be used for the management of TRAIL-resistant cancer.
Collapse
Affiliation(s)
- Cheng-Yun Jin
- School of Pharmaceutical Science, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | | | | | - Sang-Hyuck Kang
- Department of Marine Life Sciences, Jeju National University, Jeju 63243, Republic of Korea
| | - Cheol Park
- Department of Molecular Biology, College of Natural Sciences and Human Ecology, Dongeui University, Busan 67340, Republic of Korea
| | - Gi-Young Kim
- Department of Marine Life Sciences, Jeju National University, Jeju 63243, Republic of Korea.
| | - Yung Hyun Choi
- Department of Biochemistry, College of Oriental Medicine, Dong-Eui University, Busan 47227, Republic of Korea.
| |
Collapse
|
37
|
Wei D, Wang L, Chen Y, Yin G, Jiang M, Liu R, Chen H, Sun X. Yangyin Fuzheng Decoction enhances anti-tumor efficacy of cisplatin on lung cancer. J Cancer 2018; 9:1568-1574. [PMID: 29760794 PMCID: PMC5950585 DOI: 10.7150/jca.24525] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 02/16/2018] [Indexed: 12/31/2022] Open
Abstract
Traditional Chinese medicine has been widely used in cancer treatment in China. Yangyin Fuzheng Decoction is a traditional Chinese compound medicine, composed of 12 traditional Chinese herbs. This study aimed to investigate anti-tumor activity and the underlying mechanisms of Yangyin Fuzheng Decoction combined with cisplatin in the treatment of lung cancer. We established lung cancer model in C57BL/6 mice injected with mouse Lewis lung cancer cells. Our results demonstrated that Yangyin Fuzheng Decoction treatment increased necrotic area in tumor tissue, and significantly enhanced the recruitment of inflammatory cells into the tumor. In addition, Yangyin Fuzheng Decoction treatment enhanced the anti-tumor efficacy of cisplatin and partially recovered mouse body weight loss caused by cisplatin treatment. Mechanistically, we found that Yangyin Fuzheng Decoction upregulated the expression of pro-apoptotic proteins p53 and Bax and suppressed the expression of anti-apoptotic protein Bcl-2. Combined treatment of Yangyin Fuzheng Decoction and cisplatin further increased p53 and Bax levels and suppressed Bcl-2 level. Taken together, these data suggest that Yangyin Fuzheng Decoction could synergistically enhance the apoptotic signaling in cancer cells during chemotherapy. In addition, it has health improving and immune response enhancing effects. Yangyin Fuzheng Decoction could be a promising adjunct agent for lung cancer chemotherapy.
Collapse
Affiliation(s)
- Dongmei Wei
- Department of Traditional Chinese Geriatric Medicine, The First Hospital of Qiqihar, Qiqihaer, 30 Gongyuan Road, Longsha District, Qiqihaer, Heilongjiang, 161005, PR China
| | - Lin Wang
- Department of Nursing, The First Hospital of Qiqihar, Qiqihaer, 30 Gongyuan Road, Longsha District, Qiqihaer, Heilongjiang, 161005, PR China
| | - Yuhan Chen
- Department of Chinese and Western medicine combined with oncology, Academy of Traditional Chinese Medicine Guang'anmen Hospital, No.5, North Line, Xicheng District, Beijing 100053 PR China
| | - Gang Yin
- Department of Traditional Chinese Geriatric Medicine, The First Hospital of Qiqihar, Qiqihaer, 30 Gongyuan Road, Longsha District, Qiqihaer, Heilongjiang, 161005, PR China
| | - Mei Jiang
- Department of Chinese and Western medicine combined with oncology, The First Hospital of Qiqihar, Qiqihaer, 30 Gongyuan Road, Longsha District, Qiqihaer, Heilongjiang, 161005, PR China
| | - Rui Liu
- Biobank of The First Hospital of Qiqihar, The First Hospital of Qiqihar, Qiqihaer, 30 Gongyuan Road, Longsha District, Qiqihaer, Heilongjiang, 161005, PR China
| | - Hong Chen
- Department of Traditional Chinese Geriatric Medicine, The First Hospital of Qiqihar, Qiqihaer, 30 Gongyuan Road, Longsha District, Qiqihaer, Heilongjiang, 161005, PR China
| | - Xiyuan Sun
- Department of Chinese and Western medicine combined with oncology, The First Hospital of Qiqihar, Qiqihaer, 30 Gongyuan Road, Longsha District, Qiqihaer, Heilongjiang, 161005, PR China
| |
Collapse
|
38
|
He D, Zhang S. UNBS5162 inhibits the proliferation of esophageal cancer squamous cells via the PI3K/AKT signaling pathway. Mol Med Rep 2017; 17:549-555. [PMID: 29115622 DOI: 10.3892/mmr.2017.7893] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 09/14/2017] [Indexed: 11/06/2022] Open
Abstract
C‑X‑C motif chemokine ligand (CXCL) signaling has been demonstrated to be involved in cancer invasion and migration; therefore, CXCL antagonists may serve as anticancer drugs by preventing tumor proliferation. The present study aimed to investigate whether a pan antagonist of CXCLs, UNBS5162, may inhibit esophageal cancer proliferation and to identify the underlying mechanisms. Cell proliferation and cell colony formation results, which were determined by a Cell Counting Kit‑8 assay and crystal violet staining, respectively, demonstrated that UNBS5162 inhibited esophageal cancer cell proliferation. Following treatment with UNBS5162, Transwell migration and Matrigel invasion assays, and flow cytometry with Annexin V‑fluorescein isothiocyanate and propidium iodide staining, were performed to investigate cell migration, invasion and apoptosis in human esophageal cancer cells. The results indicated that invasion and migration was reduced in UNBS5162‑treated cells, while apoptosis was increased. Western blotting experiments confirmed that UNBS5162 downregulated the protein expression of proteins associated with the phosphatidylinositol 3‑kinase (PI3K)/AKT signaling pathway, including the levels of phosphorylated (p)‑AKT, p‑mechanistic target of rapamycin kinase, ribosomal protein S6 kinase β1 and cyclin D1. In addition, upregulated expression of programed cell death 4 was observed following UNBS5162 treatment. The present study demonstrated that UNBS5162 is a novel naphthalimide that may have potential therapeutic use for the prevention of esophageal cancer proliferation and metastasis via the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Dan He
- Department of Thoracic Surgery, Affiliated Tumor Hospital, Xinjiang Medical University, Urumqi, Xinjiang 830011, P.R. China
| | - Suolin Zhang
- Department of Thoracic Surgery, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| |
Collapse
|