51
|
Pan Y, Cheng A, Wang M, Yin Z, Jia R. The Dual Regulation of Apoptosis by Flavivirus. Front Microbiol 2021; 12:654494. [PMID: 33841381 PMCID: PMC8024479 DOI: 10.3389/fmicb.2021.654494] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 02/25/2021] [Indexed: 12/11/2022] Open
Abstract
Apoptosis is a form of programmed cell death, which maintains cellular homeostasis by eliminating pathogen-infected cells. It contains three signaling pathways: death receptor pathway, mitochondria-mediated pathway, and endoplasmic reticulum pathway. Its importance in host defenses is highlighted by the observation that many viruses evade, hinder or destroy apoptosis, thereby weakening the host’s immune response. Flaviviruses such as Dengue virus, Japanese encephalitis virus, and West Nile virus utilize various strategies to activate or inhibit cell apoptosis. This article reviews the research progress of apoptosis mechanism during flaviviruses infection, including flaviviruses proteins and subgenomic flaviviral RNA to regulate apoptosis by interacting with host proteins, as well as various signaling pathways involved in flaviviruses-induced apoptosis, which provides a scientific basis for understanding the pathogenesis of flaviviruses and helps in developing an effective antiviral therapy.
Collapse
Affiliation(s)
- Yuhong Pan
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
| | - Anchun Cheng
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
| | - Mingshu Wang
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
| | - Zhongqiong Yin
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
| | - Renyong Jia
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
| |
Collapse
|
52
|
Ahmad B, Gamallat Y, Khan MF, Din SR, Israr M, Ahmad M, Tahir N, Azam N, Rahman KU, Xin W, Zexu W, Linjie P, Su P, Liang W. Natural Polyphyllins (I, II, D, VI, VII) Reverses Cancer Through Apoptosis, Autophagy, Mitophagy, Inflammation, and Necroptosis. Onco Targets Ther 2021; 14:1821-1841. [PMID: 33732000 PMCID: PMC7956893 DOI: 10.2147/ott.s287354] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 02/19/2021] [Indexed: 12/15/2022] Open
Abstract
Cancer is the second leading cause of mortality worldwide. Conventional therapies, including surgery, radiation, and chemotherapy, have limited success because of secondary resistance. Therefore, safe, non-resistant, less toxic, and convenient drugs are urgently required. Natural products (NPs), primarily sourced from medicinal plants, are ideal for cancer treatment because of their low toxicity and high success. NPs cure cancer by regulating different pathways, such as PI3K/AKT/mTOR, ER stress, JNK, Wnt, STAT3, MAPKs, NF-kB, MEK-ERK, inflammation, oxidative stress, apoptosis, autophagy, mitophagy, and necroptosis. Among the NPs, steroid saponins, including polyphyllins (I, II, D, VI, and VII), have potent pharmacological, analgesic, and anticancer activities for the induction of cytotoxicity. Recent research has demonstrated that polyphyllins (PPs) possess potent effects against different cancers through apoptosis, autophagy, inflammation, and necroptosis. This review summarizes the available studies on PPs against cancer to provide a basis for future research.
Collapse
Affiliation(s)
- Bashir Ahmad
- Department of Biology, University of Haripur, KPK, I. R. Pakistan.,College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, People's Republic of China
| | - Yaser Gamallat
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou, People's Republic of China
| | | | - Syed Riaz Din
- College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, People's Republic of China
| | - Muhammad Israr
- Department of Biology, University of Haripur, KPK, I. R. Pakistan.,Biochemistry and Molecular Biology, College of Life Science, Hebei Normal University, Hebei, People's Republic of China
| | - Manzoor Ahmad
- Department of Chemistry, Malakand University, Chakdara, KPK, I. R. Pakistan
| | - Naeem Tahir
- College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, People's Republic of China
| | - Nasir Azam
- College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, People's Republic of China
| | - Khalil Ur Rahman
- College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, People's Republic of China
| | - Wang Xin
- Department of Biotechnology, College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, People's Republic of China
| | - Wang Zexu
- Department of Biotechnology, College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, People's Republic of China
| | - Peng Linjie
- Department of Biotechnology, College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, People's Republic of China
| | - Pengyu Su
- College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, People's Republic of China
| | - Wang Liang
- Stem Cell Clinical Research Center, National Joint Engineering Laboratory, Regenerative Medicine Center, The First Affiliated Hospital of Dalian Medical, Dalian City, Liaoning Province, 116011, People's Republic of China
| |
Collapse
|
53
|
Ahmad B, Rehman SU, Azizullah A, Khan MF, Din SRU, Ahmad M, Ali A, Tahir N, Azam N, Gamallat Y, Rahman KU, Ali M, Safi M, Khan I, Qamer S, Oh DH. Molecular mechanisms of anticancer activities of polyphyllin VII. Chem Biol Drug Des 2021; 97:914-929. [PMID: 33342040 DOI: 10.1111/cbdd.13818] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 12/04/2020] [Accepted: 12/13/2020] [Indexed: 12/20/2022]
Abstract
Cancer is the leading cause of mortality in the world. The major therapies for cancer treatment are chemotherapy, surgery, and radiation therapy. All these therapies expensive, toxic and show resistance. The plant-derived compounds are considered safe, cost-effective and target cancer through different pathways. In these pathways include oxidative stress, mitochondrial dependent and independent, STAT3, NF-kB, MAPKs, cell cycle, and autophagy pathways. One of the new plants derived compounds is Polyphyllin VII (PPVII), which target cancer through different molecular mechanisms. In literature, there is a review gap of studies on PPVII; therefore in the current review, we summarized the available studies on PPVII to provide a base for future research.
Collapse
Affiliation(s)
- Bashir Ahmad
- Department of Biology (Botany, Zoology, Biochemistry), The University of Haripur, Haripur, Pakistan.,College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Shafiq Ur Rehman
- Department of Biology (Botany, Zoology, Biochemistry), The University of Haripur, Haripur, Pakistan
| | - Azizullah Azizullah
- Department of Biology (Botany, Zoology, Biochemistry), The University of Haripur, Haripur, Pakistan
| | | | - Syed Riaz Ud Din
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Manzoor Ahmad
- Department of Chemistry, Malakand University, Chakdara, Pakistan
| | - Ashraf Ali
- Department of Chemistry, The University of Haripur, Haripur, Pakistan
| | - Naeem Tahir
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Nasir Azam
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Yaser Gamallat
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Khalil Ur Rahman
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Muhsin Ali
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Mohammad Safi
- Oncology Department First affiliated Hospital of Dalian Medical University, Dalian, China
| | - Imran Khan
- Department of Food Science and Technology, The University of Haripur, Haripur, Pakistan
| | - Samina Qamer
- Department of Zoology, Government College University Faisalabad, Faisalabad, Pakistan
| | - Deog-Hwan Oh
- Department of Food Science and Biotechnology, Kangwon National University, Chuncheon, South Korea
| |
Collapse
|
54
|
Khatun M, Islam F, Gopalan V, Rahman MM, Zuberi N, Khatun L, Rakib MA, Islam MA, Lam AKY, Khanam JA. 2', 4'-dihydroxy-3, 4-methylenedioxychalcone Activate Mitochondrial Apoptosis of Ehrlich Ascites Carcinoma Cells. CURRENT DRUG THERAPY 2020. [DOI: 10.2174/1574885514666191211122437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:
Development of effective cancer-chemotherapy is the most challenging
field due to the toxicity of chemo-agents.
Objective:
As chalcone has been known to have pharmacological applications, here the aim is to
synthesized three chalcone derivatives, 2',4'-dihydroxy-3,4-methylenedioxychalcone (C1), 2'-hydroxy-
2,4, 6-trimethoxychalcone (C2) and 2'-hydroxy-4-methylchalcone (C3) and investigate their anti-cancer
properties against Ehrlich Ascites Carcinoma (EAC) cell.
Method:
Anticancer properties against EAC cells were studied by examining growth inhibition,
MTT assays, tumour-bearing mice survival, tumour weight measurement and haematological profiles.
Moreover, apoptosis of EAC cells was investigated by fluorescence microscopy, flowcytometry
and DNA fragmentation assays. Expression of apoptosis related genes were studied by
reverse transcriptase-PCR (RT-PCR).
Results:
Among the compounds, C1 exhibited highest cell growth inhibition at 200 mg/kg/day
(81.71%; P < 0.01). C1 treatment also increased the life span of EAC-bearing mice (82.60%, P <
0.05) with the reduction of tumour burden (<) compared to untreated EAC-bearing
mice. In vitro study indicated that C1 killed EAC-cells in a dose-dependent manner and induced
mitochondria-mediated apoptotic pathways. In addition, C1 treated cells exhibited increased apoptotic
features such as membrane blebbing, chromatin condensation, and nuclear fragmentation after
Hoechst 33342 staining. Increased fragmentation of DNA in gel electrophoresis followed by C1
treatment further confirmed apoptosis of EAC cells. EAC cells treated with C1 showed reduced
Bcl-2 expression in contrast to notable upregulation of p53 and Bax expression. It implied that C1
could reinstate the expression of pro-apoptotic tumour suppressor and inhibit anti-apoptotic genes.
Conclusions:
Thus, C1 showed significant growth inhibitory properties and induced apoptosis of
EAC cells.
Collapse
Affiliation(s)
- Mahbuba Khatun
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi-6205, Bangladesh
| | - Farhadul Islam
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi-6205, Bangladesh
| | - Vinod Gopalan
- Cancer Molecular Pathology, School of Medicine, Griffith University, Gold Coast, QLD-4222, Australia
| | - Md. Motiar Rahman
- Department of Chemistry, University of Rajshahi, Rajshahi-6205, Bangladesh
| | - Natasha Zuberi
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi-6205, Bangladesh
| | - Laboni Khatun
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi-6205, Bangladesh
| | - Md. Abdur Rakib
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi-6205, Bangladesh
| | - Md. Azizul Islam
- Department of Chemistry, University of Rajshahi, Rajshahi-6205, Bangladesh
| | - Alfred King-Yin Lam
- Cancer Molecular Pathology, School of Medicine, Griffith University, Gold Coast, QLD-4222, Australia
| | - Jahan Ara Khanam
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi-6205, Bangladesh
| |
Collapse
|
55
|
Carneiro ACDM, De Vito FB, Moraes-Souza H, Crema VO. RhoA/ROCKs signaling is increased by treatment with TKI-258 and leads to increased apoptosis in SCC-4 oral squamous cell carcinoma cell line. J Oral Pathol Med 2020; 50:394-402. [PMID: 33222274 DOI: 10.1111/jop.13143] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 11/06/2020] [Accepted: 11/12/2020] [Indexed: 01/01/2023]
Abstract
BACKGROUND This study evaluated the effect of treatment with TKI-258 on apoptosis, involving Rho GTPases and their effectors in SCC-4 cells of oral squamous cell carcinoma. METHODS Markers of cell death and apoptosis were analyzed in control and TKI-258-treated SCC-4 cells by flow cytometry. The involvement of Rho GTPases and effectors in the induction of apoptosis by TKI-258 was evaluated by quantification of cleaved PARP. Also, gene expression analysis of those proteins was performed. RESULTS The treatment with TKI-258 led to a significant increase in cell death (7-AAD) and apoptosis (annexin V and cleaved PARP). When Rho GTPases were stimulated with LPA and inhibited with toxin A Clostridium difficile, the percentage of apoptotic cells increased and decreased, respectively. A similar effect was found when the treatment was with TKI-258 combined with LPA and toxin A. Treatment with TKI-258 significantly increased RhoA gene expression, while RhoB, RhoC, Rac1, and Cdc42 decreased significantly. ROCKs inhibitors (Y-27632 and HA-1077) reduced apoptosis compared with control. TKI-258 combined with Y-27632 or HA-1077 led to an increase in apoptosis compared with inhibitors only. Treatment with TKI-258 led to an increase in ROCK1 and ROCK2 gene expression, and a decrease in PAK1 and PAK2 gene expression. CONCLUSIONS TKI-258 stimulates apoptosis in SCC-4 cells of oral squamous cell carcinoma. Possibly, RhoA GTPase and their effectors ROCKs participate in the signaling pathway inhibited by TKI-258. CLINICAL RELEVANCE Therapies with multi-target inhibitors, such as TKI-258, may be promising alternatives for the clinical treatment of oral squamous cell carcinoma.
Collapse
Affiliation(s)
- Anna Cecília Dias Maciel Carneiro
- Structural Biology Department, Institute of Natural and Biological Sciences, Federal University of Triângulo Mineiro, Uberaba, Brazil
| | - Fernanda Bernadelli De Vito
- Medical Clinical Department, Institute of Health Sciences, Federal University of Triângulo Mineiro, Uberaba, Brazil
| | - Helio Moraes-Souza
- Medical Clinical Department, Institute of Health Sciences, Federal University of Triângulo Mineiro, Uberaba, Brazil
| | - Virgínia Oliveira Crema
- Structural Biology Department, Institute of Natural and Biological Sciences, Federal University of Triângulo Mineiro, Uberaba, Brazil
| |
Collapse
|
56
|
Changizi V, Azariasl S, Motevaseli E, Jafari Nodooshan S. Assessment Synergistic Effects of Integrated Therapy with Epigallocatechin-3-Gallate (EGCG) & Arsenic Trioxide and Irradiation on Breast Cancer Cell Line. IRANIAN JOURNAL OF PUBLIC HEALTH 2020; 49:1555-1563. [PMID: 33083333 PMCID: PMC7554386 DOI: 10.18502/ijph.v49i8.3901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Background: Breast cancer is the most common invasive malignancy among women in the world. The current breast cancer therapies pose significant clinical challenges. Low-dose chemotherapy represents a new strategy to treat solid tumors in combination with natural products such as green tea catechins. Epigallocatechin-3-gallate (EGCG) is the major polyphenolic extract from green tea with potent anticancer and antioxidant effects. The purpose of this study was to investigate the effects of EGCG, Arsenic trioxide (ATO) and gamma radiation on MCF-7 cell line. Methods: The anti-proliferative effects of EGCG and ATO individually, moreover in combination with radiation on MCF-7 cells were evaluated with MTT assay. The expression of apoptotic gens (Bax, Bcl-2, Caspase-3 and Fas) was assessed by real-time PCR. Results: Based on the results of MTT assay, EGCG and ATO exhibited dose and time-dependent anti-proliferative effects on MCF-7 cells. The combined therapy of EGCG and ATO in presence and absence radiation could rise cell death up to 80%. Moreover, integrated therapy made Bax up-regulated and Bcl-2 down- regulated. Conclusion: In assessment synergistic effects of integrated therapy with EGCG and ATO and irradiation had been significant impact on low dose chemotherapy for breast cancer treatment.
Collapse
Affiliation(s)
- Vahid Changizi
- Department of Technology of Radiology and Radiotherapy, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Samayeh Azariasl
- Department of Technology of Radiology and Radiotherapy, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Elahe Motevaseli
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Saeedeh Jafari Nodooshan
- School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
57
|
Zhao L, Fu Q, Pan L, Piai A, Chou JJ. The Diversity and Similarity of Transmembrane Trimerization of TNF Receptors. Front Cell Dev Biol 2020; 8:569684. [PMID: 33163490 PMCID: PMC7591462 DOI: 10.3389/fcell.2020.569684] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 09/17/2020] [Indexed: 12/26/2022] Open
Abstract
Receptors in the tumor necrosis factor receptor superfamily (TNFRSF) regulate proliferation of immune cells or induce programmed cell death, and many of them are candidates for antibody-based immunotherapy. Previous studies on several death receptors in the TNFRSF including Fas, p75NTR, and DR5 showed that the transmembrane helix (TMH) of these receptors can specifically oligomerize and their oligomeric states have direct consequences on receptor activation, suggesting a much more active role of TMH in receptor signaling than previously appreciated. Here, we report the structure of the TMH of TNFR1, another well studied member of the TNFRSF, in neutral bicelles that mimic a lipid bilayer. We find that TNFR1 TMH forms a defined trimeric complex in bicelles, and no evidences of higher-order clustering of trimers have been detected. Unexpectedly, a conserved proline, which is critical for Fas TMH trimerization, does not appear to play an important role in TNFR1 TMH trimerization, which is instead mediated by a glycine near the middle of the TMH. Further, TNFR1 TMH trimer shows a larger hydrophobic core than that of Fas or DR5, with four layers of hydrophobic interaction along the threefold axis. Comparison of the TNFR1 TMH structure with that of Fas and DR5 reveals reassuring similarities that have functional implications but also significant structural diversity that warrants systematic investigation of TMH oligomerization property for other members of the TNFRSF.
Collapse
Affiliation(s)
- Linlin Zhao
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, United States
| | - Qingshan Fu
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, United States
| | - Liqiang Pan
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, United States
| | - Alessandro Piai
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, United States
| | - James J Chou
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
58
|
Prooxidation and Cytotoxicity of Selenium Nanoparticles at Nonlethal Level in Sprague-Dawley Rats and Buffalo Rat Liver Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:7680276. [PMID: 32922654 PMCID: PMC7453254 DOI: 10.1155/2020/7680276] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 07/14/2020] [Accepted: 07/21/2020] [Indexed: 11/17/2022]
Abstract
The effects of selenium nanoparticles (SeNPs) on the antioxidant capacity in Sprague-Dawley (SD) rats were investigated. The rats were given intragastric administration of an SeNP suspension at doses of 0, 2, 4, and 8 mg Se/kg BW for two weeks. The antioxidant capacity in serum and organic tissues (liver, heart, and kidney) and the gene expression levels of glutathione peroxidase 1 (GPX1) and glutathione peroxidase 4 (GPX4) in the liver were measured. Buffalo rat liver (BRL) cell lines were further constructed to explore the cytotoxicity mechanism induced by SeNPs through the determination of antioxidant capacity; cell activity; apoptosis; and Caspase-3, Caspase-8, and Caspase-9 family activities. The results showed that SeNP administration over 4.0 mg Se/kg BW decreased the antioxidant capacities in the serum, liver, and heart and downregulated mRNA expression of GPX1 and GPX4 in the liver. The BRL cell line experiments showed that treatment with over 24 μM SeNPs decreased the viability of the cells and damaged the antioxidant capacity. Flow cytometry analysis showed that decreased cell viability induced by SeNPs is mainly due to apoptosis, rather than cell necrosis. Caspase-3 and Caspase-8 activities were also increased when BRL cells were treated with 24 μM and 48 μM SeNPs. Taken together, a nonlethal level of SeNPs could impair the antioxidant capacity in serum and organic tissues of rats, and the liver is the most sensitive to the toxicity of SeNPs. A pharmacological dose of SeNPs could lead to cytotoxicity and induce cell death through apoptosis and extrinsic pathways contributing to SeNP-induced apoptosis in BRL cells.
Collapse
|
59
|
Yu SH, Lee CM, Ha SH, Lee J, Jang KY, Park SH. Induction of cell cycle arrest and apoptosis by tomentosin in hepatocellular carcinoma HepG2 and Huh7 cells. Hum Exp Toxicol 2020; 40:231-244. [PMID: 32787465 DOI: 10.1177/0960327120943935] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Tomentosin, a sesquiterpene lactone, is known to possess various biological activities. However, its anticarcinogenic activity against human hepatocellular carcinoma (HCC) cells has not been investigated in detail. Thus, this study aimed to elucidate the cytotoxic mechanism of tomentosin in human HCC cell lines HepG2 and Huh7. WST-1, cell counting, and colony formation assay results showed that treatment with tomentosin decreased the viability and suppressed the proliferation rate of HepG2 and Huh7 cells in a dose- and time-dependent manner. Cell cycle analysis revealed increased population of cells at the SubG1 and G2/M stage, and decreased population of cells at the G0/1 stage in HepG2 and Huh7 cells treated with tomentosin. Annexin V/propidium iodide double staining and TUNEL assay results showed increased apoptotic cell population and DNA fragmentation in HepG2 and Huh7 cells treated with tomentosin. Western blotting analysis results showed that tomentosin treatment significantly increased the expression level of Bax, Bim (short form), cleaved PARP1, FOXO3, p53, pSer15p53, pSer20p53, pSer46p53, p21, and p27, but decreased the expression of Bcl2, caspase3, caspase7, caspase9, cyclin-dependent kinase 2 (CDK2), CDK4, CDK6, cyclinB1, cyclinD1, cyclinD2, cyclinD3, and cyclinE in a dose-dependent manner. Taken together, this study revealed that tomentosin, which acted through cell cycle arrest and apoptosis, may be a useful therapeutic option against HCC.
Collapse
Affiliation(s)
- S H Yu
- Department of Bio and Chemical Engineering, 65686Hongik University, Sejong, Republic of Korea
| | - C M Lee
- Department of Bio and Chemical Engineering, 65686Hongik University, Sejong, Republic of Korea
| | - S H Ha
- Division of Biotechnology, 26714Jeonbuk National University, Iksan, Republic of Korea
| | - J Lee
- Department of Integrative Biotechnology, 65666Sungkyunkwan University, Suwon, Republic of Korea
| | - K Y Jang
- Department of Pathology, 26714Jeonbuk National University Medical School, Jeonju, Republic of Korea.,Research Institute of Clinical Medicine of Jeonbuk National University, Jeonju, Republic of Korea.,Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Republic of Korea
| | - S H Park
- Department of Bio and Chemical Engineering, 65686Hongik University, Sejong, Republic of Korea
| |
Collapse
|
60
|
Volpin V, Michels T, Sorrentino A, Menevse AN, Knoll G, Ditz M, Milenkovic VM, Chen CY, Rathinasamy A, Griewank K, Boutros M, Haferkamp S, Berneburg M, Wetzel CH, Seckinger A, Hose D, Goldschmidt H, Ehrenschwender M, Witzens-Harig M, Szoor A, Vereb G, Khandelwal N, Beckhove P. CAMK1D Triggers Immune Resistance of Human Tumor Cells Refractory to Anti-PD-L1 Treatment. Cancer Immunol Res 2020; 8:1163-1179. [PMID: 32665263 DOI: 10.1158/2326-6066.cir-19-0608] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 03/16/2020] [Accepted: 07/09/2020] [Indexed: 11/16/2022]
Abstract
The success of cancer immunotherapy is limited by resistance to immune checkpoint blockade. We therefore conducted a genetic screen to identify genes that mediated resistance against CTLs in anti-PD-L1 treatment-refractory human tumors. Using PD-L1-positive multiple myeloma cells cocultured with tumor-reactive bone marrow-infiltrating CTL as a model, we identified calcium/calmodulin-dependent protein kinase 1D (CAMK1D) as a key modulator of tumor-intrinsic immune resistance. CAMK1D was coexpressed with PD-L1 in anti-PD-L1/PD-1 treatment-refractory cancer types and correlated with poor prognosis in these tumors. CAMK1D was activated by CTL through Fas-receptor stimulation, which led to CAMK1D binding to and phosphorylating caspase-3, -6, and -7, inhibiting their activation and function. Consistently, CAMK1D mediated immune resistance of murine colorectal cancer cells in vivo The pharmacologic inhibition of CAMK1D, on the other hand, restored the sensitivity toward Fas-ligand treatment in multiple myeloma and uveal melanoma cells in vitro Thus, rapid inhibition of the terminal apoptotic cascade by CAMK1D expressed in anti-PD-L1-refractory tumors via T-cell recognition may have contributed to tumor immune resistance.
Collapse
Affiliation(s)
- Valentina Volpin
- Regensburg Center for Interventional Immunology (RCI), University Regensburg, Regensburg, Germany.,German Cancer Research Center (DKFZ), Translational Immunology, Heidelberg, Germany
| | - Tillmann Michels
- Regensburg Center for Interventional Immunology (RCI), University Regensburg, Regensburg, Germany.,German Cancer Research Center (DKFZ), Translational Immunology, Heidelberg, Germany.,iOmx Therapeutics AG, Martinsried/Munich, Germany
| | - Antonio Sorrentino
- Regensburg Center for Interventional Immunology (RCI), University Regensburg, Regensburg, Germany.,German Cancer Research Center (DKFZ), Translational Immunology, Heidelberg, Germany
| | - Ayse N Menevse
- Regensburg Center for Interventional Immunology (RCI), University Regensburg, Regensburg, Germany
| | - Gertrud Knoll
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| | - Madlen Ditz
- Regensburg Center for Interventional Immunology (RCI), University Regensburg, Regensburg, Germany
| | - Vladimir M Milenkovic
- Department of Psychiatry and Psychotherapy, Molecular Neurosciences, University of Regensburg, Regensburg, Germany
| | - Chih-Yeh Chen
- Regensburg Center for Interventional Immunology (RCI), University Regensburg, Regensburg, Germany
| | - Anchana Rathinasamy
- Regensburg Center for Interventional Immunology (RCI), University Regensburg, Regensburg, Germany
| | - Klaus Griewank
- Department of Dermatology, University Hospital Essen, West German Cancer Center, University Duisburg-Essen and the German Cancer Consortium, Essen, Germany
| | - Michael Boutros
- German Cancer Research Center (DKFZ), Division Signalling and Functional Genomics, Heidelberg, Germany
| | - Sebastian Haferkamp
- Department of Dermatology, University Hospital Regensburg, Regensburg, Germany
| | - Mark Berneburg
- Department of Dermatology, University Hospital Regensburg, Regensburg, Germany
| | - Christian H Wetzel
- Department of Psychiatry and Psychotherapy, Molecular Neurosciences, University of Regensburg, Regensburg, Germany
| | - Anja Seckinger
- Labor für Myelomforschung, Medizinische Klinik V, Universitätsklinikum Heidelberg, Heidelberg, Germany
| | - Dirk Hose
- Department of Hematology and Immunology, Myeloma Center Brussels, Jette, Belgium
| | - Hartmut Goldschmidt
- Department of Internal Medicine V and National Center of Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany
| | - Martin Ehrenschwender
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| | - Mathias Witzens-Harig
- Department of Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Arpad Szoor
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Gyorgy Vereb
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | | | - Philipp Beckhove
- Regensburg Center for Interventional Immunology (RCI), University Regensburg, Regensburg, Germany. .,German Cancer Research Center (DKFZ), Translational Immunology, Heidelberg, Germany.,Department of Hematology, Oncology, Internal Medicine 3, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
61
|
Re-engineering Antimicrobial Peptides into Oncolytics Targeting Drug-Resistant Ovarian Cancers. Cell Mol Bioeng 2020; 13:447-461. [PMID: 33184577 DOI: 10.1007/s12195-020-00626-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 06/12/2020] [Indexed: 10/24/2022] Open
Abstract
Introduction Bacteria and cancer cells share a common trait-both possess an electronegative surface that distinguishes them from healthy mammalian counterparts. This opens opportunities to repurpose antimicrobial peptides (AMPs), which are cationic amphiphiles that kill bacteria by disrupting their anionic cell envelope, into anticancer peptides (ACPs). To test this assertion, we investigate the mechanisms by which a pathogen-specific AMP, originally designed to kill bacterial Tuberculosis, potentiates the lytic destruction of drug-resistant cancers and synergistically enhances chemotherapeutic potency. Materials and Methods De novo peptide design, paired with cellular assays, elucidate structure-activity relationships (SAR) important to ACP potency and specificity. Using the sequence MAD1, microscopy, spectrophotometry and flow cytometry identify the peptide's anticancer mechanisms, while parallel combinatorial screens define chemotherapeutic synergy in drug-resistant cell lines and patient derived ex vivo tumors. Results SAR investigations reveal spatial sequestration of amphiphilic regions increases ACP potency, but at the cost of specificity. Selecting MAD1 as a lead sequence, mechanistic studies identify that the peptide forms pore-like supramolecular assemblies within the plasma and nuclear membranes of cancer cells to potentiate death through lytic and apoptotic mechanisms. This diverse activity enables MAD1 to synergize broadly with chemotherapeutics, displaying remarkable combinatorial efficacy against drug-resistant ovarian carcinoma cells and patient-derived tumor spheroids. Conclusions We show that cancer-specific ACPs can be rationally engineered using nature's AMP toolbox as templates. Selecting the antimicrobial peptide MAD1, we demonstrate the potential of this strategy to open a wealth of synthetic biotherapies that offer new, combinatorial opportunities against drug resistant tumors.
Collapse
|
62
|
De L, Yuan T, Yong Z. ST1926 inhibits glioma progression through regulating mitochondrial complex II. Biomed Pharmacother 2020; 128:110291. [PMID: 32526455 DOI: 10.1016/j.biopha.2020.110291] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 05/12/2020] [Accepted: 05/16/2020] [Indexed: 10/24/2022] Open
Abstract
The antitumor activity of atypical adamantyl retinoid ST1926 has been frequently reported in cancer studies; nevertheless, its effect on glioma has not been fully understood. Mitochondria are critical in regulating tumorigenesis and are defined as a promising target for anti-tumor therapy. In the present study, we found that ST1926 might be a mitochondria-targeting anti-glioma drug. ST1926 showed significantly inhibitory role in the viability of glioma cells mainly through inducing apoptosis and autophagy. The results showed that ST1926 alleviated mitochondria-regulated bioenergetics in glioma cells via reducing ATP production and promoting reactive oxygen species production. Importantly, ST1926 significantly impaired complex II (CII) function, which was associated with the inhibition of succinate dehydrogenase (SDH) activity. In addition, the effects of ST1926 on the induction of apoptosis and ROS were further promoted by the treatment of CII inhibitors, including TTFA and 3-NPA. Furthermore, the in vivo experiments confirmed the role of ST1926 in suppressing xenograft tumor growth with few toxicity. Therefore, ST1926 might be an effective anti-glioma drug through targeting CII.
Collapse
Affiliation(s)
- Liu De
- Department of Neurosurgery, Liaocheng Third People's Hospital, Shandong Province, 252000, China
| | - Tang Yuan
- Department of Neurosurgery, Liaocheng Third People's Hospital, Shandong Province, 252000, China
| | - Zheng Yong
- Department of Neurosurgery, The Second Affiliated Hospital of Shenzhen University (People's Hospital of Shenzhen Baoan District), Shenzhen, Guangdong, 518101, China.
| |
Collapse
|
63
|
El-Huneidi W, Shehab NG, Bajbouj K, Vinod A, El-Serafi A, Shafarin J, Bou Malhab LJ, Abdel-Rahman WM, Abu-Gharbieh E. Micromeria fruticosa Induces Cell Cycle Arrest and Apoptosis in Breast and Colorectal Cancer Cells. Pharmaceuticals (Basel) 2020; 13:115. [PMID: 32503209 PMCID: PMC7345572 DOI: 10.3390/ph13060115] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 06/01/2020] [Accepted: 06/01/2020] [Indexed: 11/16/2022] Open
Abstract
Micromeria fruticosa (L.) Druce subsp. Serpyllifolia (Lamiaceae) has been used widely in folk medicine to alleviate various ailments such as abdominal pains, diarrhea, colds, eye infections, heart disorders and wounds. A few reports have confirmed different therapeutic potentialities of its extracts, including the anti-inflammatory, gastroprotective, analgesic, antiobesity and antidiabetic activities. This study aimed to investigate the mechanistic pathway of the antiproliferative activity of the ethanolic extract of M. fruticose on two different cancer cell lines, namely human breast (mammary carcinoma F7 (MCF-7)) and human colorectal (human colon tumor cells (HCT-116)) cell lines. The 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide tetrazolium (MTT) assay, Annexin V-FITC/PI, caspases 8/9 and cell cycle analyses, qRT-PCR and Western blot were used to assess the effect of M. fruticosa on cytotoxicity, apoptosis, cell cycle, cell cycle-related genes and protein expression profiles in MCF-7 and HCT-116. The extract inhibits cell proliferation in a time- and dose-dependent manner. The half-maximal inhibitory concentration (IC50) for both cell lines was found to be 100 μg/mL. Apoptosis induction was confirmed by Annexin V-FITC/PI, that was related to caspases 8 and 9 activities induction. Furthermore, the cell cycle analysis revealed arrest at G2/M phase. The underlying mechanism involved in the G2/M arrest was found to be associated with the downregulation of CDK1, cyclin B1 and survivin that was confirmed by qRT-PCR and Western blotting.
Collapse
Affiliation(s)
- Waseem El-Huneidi
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, UAE; (W.E.-H.); (K.B.)
| | - Naglaa G. Shehab
- Department of Pharmaceutical Chemistry and Natural Products, Dubai Pharmacy College, Dubai 19099, UAE;
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Giza 12613, Egypt
| | - Khuloud Bajbouj
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, UAE; (W.E.-H.); (K.B.)
| | - Arya Vinod
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, UAE; (A.V.); (J.S.); (L.J.B.M.); (W.M.A.-R.)
| | - Ahmed El-Serafi
- Department of Biomedical and Clinical Sciences, Linköping University, SE-581 85 Linköping, Sweden;
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Jasmin Shafarin
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, UAE; (A.V.); (J.S.); (L.J.B.M.); (W.M.A.-R.)
| | - Lara J. Bou Malhab
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, UAE; (A.V.); (J.S.); (L.J.B.M.); (W.M.A.-R.)
| | - Wael M. Abdel-Rahman
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, UAE; (A.V.); (J.S.); (L.J.B.M.); (W.M.A.-R.)
- Department of Medical Laboratory Sciences, College of Health Sciences, University of Sharjah, Sharjah 27272, UAE
| | - Eman Abu-Gharbieh
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, UAE
| |
Collapse
|
64
|
Ran L, Chen F, Zhang J, Mi J, Lu L, Yan Y, Cao Y. Antitumor effects of pollen polysaccharides from Chinese wolfberry on DU145 cells via the PI3K/AKT pathway in vitro and in vivo. Int J Biol Macromol 2020; 152:1164-1173. [DOI: 10.1016/j.ijbiomac.2019.10.206] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 10/13/2019] [Accepted: 10/24/2019] [Indexed: 12/15/2022]
|
65
|
Dittmann J, Haydn T, Metzger P, Ward GA, Boerries M, Vogler M, Fulda S. Next-generation hypomethylating agent SGI-110 primes acute myeloid leukemia cells to IAP antagonist by activating extrinsic and intrinsic apoptosis pathways. Cell Death Differ 2020; 27:1878-1895. [PMID: 31831875 PMCID: PMC7244748 DOI: 10.1038/s41418-019-0465-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 11/21/2019] [Accepted: 11/21/2019] [Indexed: 12/16/2022] Open
Abstract
Therapeutic efficacy of first-generation hypomethylating agents (HMAs) is limited in elderly acute myeloid leukemia (AML) patients. Therefore, combination strategies with targeted therapies are urgently needed. Here, we discover that priming with SGI-110 (guadecitabine), a next-generation HMA, sensitizes AML cells to ASTX660, a novel antagonist of cellular inhibitor of apoptosis protein 1 and 2 (cIAP1/2) and X-linked IAP (XIAP). Importantly, SGI-110 and ASTX660 synergistically induced cell death in a panel of AML cell lines as well as in primary AML samples while largely sparing normal CD34+ human progenitor cells, underlining the translational relevance of this combination. Unbiased transcriptome analysis revealed that SGI-110 alone or in combination with ASTX660 upregulated the expression of key regulators of both extrinsic and intrinsic apoptosis signaling pathways such as TNFRSF10B (DR5), FAS, and BAX. Individual knockdown of the death receptors TNFR1, DR5, and FAS significantly reduced SGI-110/ASTX660-mediated cell death, whereas blocking antibodies for tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) or FAS ligand (FASLG) failed to provide protection. Also, TNFα-blocking antibody Enbrel had little protective effect on SGI-110/ASTX660-induced cell death. Further, SGI-110 and ASTX660 acted in concert to promote cleavage of caspase-8 and BID, thereby providing a link between extrinsic and intrinsic apoptotic pathways. Consistently, sequential treatment with SGI-110 and ASTX660-triggered loss of mitochondrial membrane potential (MMP) and BAX activation which contributes to cell death, as BAX silencing significantly protected from SGI-110/ASTX660-mediated apoptosis. Together, these events culminated in the activation of caspases-3/-7, nuclear fragmentation, and cell death. In conclusion, SGI-110 and ASTX660 cooperatively induced apoptosis in AML cells by engaging extrinsic and intrinsic apoptosis pathways, highlighting the therapeutic potential of this combination for AML.
Collapse
Affiliation(s)
- Jessica Dittmann
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University Frankfurt, Frankfurt am Main, Germany
| | - Tinka Haydn
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University Frankfurt, Frankfurt am Main, Germany
| | - Patrick Metzger
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center, Faculty of Medicine, University Freiburg, Freiburg im Breisgau, Germany
- Institute of Molecular Medicine and Cell Research (IMMZ), Albert Ludwigs-University Freiburg, Freiburg im Breisgau, Germany
- Faculty of Biology, University Freiburg, Freiburg im Breisgau, Germany
| | | | - Melanie Boerries
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center, Faculty of Medicine, University Freiburg, Freiburg im Breisgau, Germany
- Institute of Molecular Medicine and Cell Research (IMMZ), Albert Ludwigs-University Freiburg, Freiburg im Breisgau, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg im Breisgau, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Meike Vogler
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University Frankfurt, Frankfurt am Main, Germany
| | - Simone Fulda
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University Frankfurt, Frankfurt am Main, Germany.
- German Cancer Research Center (DKFZ), Heidelberg, Germany.
- German Cancer Consortium (DKTK), Partner Site Frankfurt, Frankfurt am Main, Germany.
| |
Collapse
|
66
|
Kim SM, Vetrivel P, Ha SE, Kim HH, Kim JA, Kim GS. Apigetrin induces extrinsic apoptosis, autophagy and G2/M phase cell cycle arrest through PI3K/AKT/mTOR pathway in AGS human gastric cancer cell. J Nutr Biochem 2020; 83:108427. [PMID: 32559585 DOI: 10.1016/j.jnutbio.2020.108427] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 05/06/2020] [Accepted: 05/15/2020] [Indexed: 12/24/2022]
Abstract
Apigetrin is a flavonoid glycoside phytonutrient derived from fruits and vegetables that is well known for a variety of biological activities such as antioxidant and anti-inflammatory activities. In the current study, we determined the effect of apigetrin on AGS gastric cancer cell. Apigetrin reduced cancer cell proliferation and induced G2/M phase cell cycle arrest by regulating cyclin B1, cdc25c and cdk1 protein expression in AGS cell. Apigetrin treatment caused apoptotic cell death in AGS cells, characterized by the accumulation of apoptosis portion, cleavage of caspase-3 and poly ADP-ribose polymerase (PARP). Apigetrin-treated cells increased the expression of extrinsic apoptosis pathway proteins and mRNA. However, intrinsic apoptosis pathway related proteins were not altered. In addition, AGS cells treated with apigetrin increased autophagic cell death, featured by the formation of autophagic vacuole and acidic vesicular organelles. Autophagy marker proteins, such as LC3B-II and beclin-1, were increased, and p62, an autophagy flux marker protein, was also increased by endoplasmic reticulum stress. Also, the phosphorylation of PI3K/AKT/mTOR pathway proteins and its downstream targets in apigetrin-treated AGS cells was identified to be decreased. Taken together, these data suggest that apigetrin-treated AGS cells induced G2/M phase cell cycle arrest, extrinsic apoptosis and autophagic cell death through PI3K/AKT/mTOR pathway, which can lead to the inhibition of gastric cancer development. Thus, our findings strongly indicate that apigetrin is a basic natural derived compound that could be used as a nutrient source with potential anticancer activities against gastric cancer.
Collapse
Affiliation(s)
- Seong Min Kim
- Research Institute of Life science and College of Veterinary Medicine, Gyeongsang National University, Gazwa, Jinju, 52828, Republic, Republic of Korea.
| | - Preethi Vetrivel
- Research Institute of Life science and College of Veterinary Medicine, Gyeongsang National University, Gazwa, Jinju, 52828, Republic, Republic of Korea.
| | - Sang Eun Ha
- Research Institute of Life science and College of Veterinary Medicine, Gyeongsang National University, Gazwa, Jinju, 52828, Republic, Republic of Korea.
| | - Hun Hwan Kim
- Research Institute of Life science and College of Veterinary Medicine, Gyeongsang National University, Gazwa, Jinju, 52828, Republic, Republic of Korea.
| | - Jin-A Kim
- Department of Physical Therapy, International University of Korea, Jinju, 52833, Republic of Korea.
| | - Gon Sup Kim
- Research Institute of Life science and College of Veterinary Medicine, Gyeongsang National University, Gazwa, Jinju, 52828, Republic, Republic of Korea.
| |
Collapse
|
67
|
Gao C, Sun X, Wu Z, Yuan H, Han H, Huang H, Shu Y, Xu M, Gao R, Li S, Zhang J, Tian J. A Novel Benzofuran Derivative Moracin N Induces Autophagy and Apoptosis Through ROS Generation in Lung Cancer. Front Pharmacol 2020; 11:391. [PMID: 32477104 PMCID: PMC7235196 DOI: 10.3389/fphar.2020.00391] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 03/16/2020] [Indexed: 12/15/2022] Open
Abstract
Introduction The leaves of Morus alba L is a traditional Chinese medicine widely applied in lung diseases. Moracin N (MAN), a secondary metabolite extracted form the leaves of Morus alba L, is a potent anticancer agent. But its molecular mechanism remains unveiled. Objective In this study, we aimed to examine the effect of MAN on human lung cancer and reveal the underlying molecular mechanism. Methods MTT assay was conducted to measure cell viability. Annexin V-FITC/PI staining was used to detect cell apoptosis. Confocal microscope was performed to determine the formation of autophagosomes and autolysosomes. Flow cytometry was performed to quantify cell death. Western blotting was used to determine the related-signaling pathway. Results In the present study, we demonstrated for the first time that MAN inhibitd cell proliferation and induced cell apoptosis in human non-small-cell lung carcinoma (NSCLC) cells. We found that MAN treatment dysregulated mitochondrial function and led to mitochondrial apoptosis in A549 and PC9 cells. Meanwhile, MAN enhanced autophagy flux by the increase of autophagosome formation, the fusion of autophagsomes and lysosomes and lysosomal function. Moreover, mTOR signaling pathway, a classical pathway regualting autophagy, was inhibited by MAN in a time- and dose-dependent mannner, resulting in autophagy induction. Interestingly, autophagy inhibition by CQ or Atg5 knockdown attenuated cell apoptosis by MAN, indicating that autophagy serves as cell death. Furthermore, autophagy-mediated cell death by MAN can be blocked by reactive oxygen species (ROS) scavenger NAC, indicating that ROS accumulation is the inducing factor of apoptosis and autophagy. In summary, we revealed the molecular mechanism of MAN against lung cancer through apoptosis and autophagy, suggesting that MAN might be a novel therapeutic agent for NSCLC treatment.
Collapse
Affiliation(s)
- Chengcheng Gao
- College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China.,Clinical Research Institute, Key Laboratory of Tumor Molecular Diagnosis and Individual Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Xin Sun
- Clinical Research Institute, Key Laboratory of Tumor Molecular Diagnosis and Individual Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China.,Department of Oncology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Zhipan Wu
- College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Huahua Yuan
- College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Haote Han
- College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Hongliang Huang
- School of Biosciences & Biopharmaceutics and Center for Bioresources & Drug Discovery, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yuhan Shu
- College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Mengting Xu
- College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Ruilan Gao
- Institution of Hematology Research, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Shouxin Li
- College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China.,Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang-Malaysia Joint Research Center for Traditional Medicine, Zhejiang University, Hangzhou, China
| | - Jianbin Zhang
- Clinical Research Institute, Key Laboratory of Tumor Molecular Diagnosis and Individual Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China.,Department of Oncology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Jingkui Tian
- College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China.,Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang-Malaysia Joint Research Center for Traditional Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
68
|
Mahendra CK, Tan LTH, Lee WL, Yap WH, Pusparajah P, Low LE, Tang SY, Chan KG, Lee LH, Goh BH. Angelicin-A Furocoumarin Compound With Vast Biological Potential. Front Pharmacol 2020; 11:366. [PMID: 32372949 PMCID: PMC7176996 DOI: 10.3389/fphar.2020.00366] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 03/10/2020] [Indexed: 12/14/2022] Open
Abstract
Angelicin, a member of the furocoumarin group, is related to psoralen which is well known for its effectiveness in phototherapy. The furocoumarins as a group have been studied since the 1950s but only recently has angelicin begun to come into its own as the subject of several biological studies. Angelicin has demonstrated anti-cancer properties against multiple cell lines, exerting effects via both the intrinsic and extrinsic apoptotic pathways, and also demonstrated an ability to inhibit tubulin polymerization to a higher degree than psoralen. Besides that, angelicin too demonstrated anti-inflammatory activity in inflammatory-related respiratory and neurodegenerative ailments via the activation of NF-κB pathway. Angelicin also showed pro-osteogenesis and pro-chondrogenic effects on osteoblasts and pre-chondrocytes respectively. The elevated expression of pro-osteogenic and chondrogenic markers and activation of TGF-β/BMP, Wnt/β-catenin pathway confirms the positive effect of angelicin bone remodeling. Angelicin also increased the expression of estrogen receptor alpha (ERα) in osteogenesis. Other bioactivities, such as anti-viral and erythroid differentiating properties of angelicin, were also reported by several researchers with the latter even displaying an even greater aptitude as compared to the commonly prescribed drug, hydroxyurea, which is currently on the market. Apart from that, recently, a new application for angelicin against periodontitis had been studied, where reduction of bone loss was indirectly caused by its anti-microbial properties. All in all, angelicin appears to be a promising compound for further studies especially on its mechanism and application in therapies for a multitude of common and debilitating ailments such as sickle cell anaemia, osteoporosis, cancer, and neurodegeneration. Future research on the drug delivery of angelicin in cancer, inflammation and erythroid differentiation models would aid in improving the bioproperties of angelicin and efficacy of delivery to the targeted site. More in-depth studies of angelicin on bone remodeling, the pro-osteogenic effect of angelicin in various bone disease models and the anti-viral implications of angelicin in periodontitis should be researched. Finally, studies on the binding of angelicin toward regulatory genes, transcription factors, and receptors can be done through experimental research supplemented with molecular docking and molecular dynamics simulation.
Collapse
Affiliation(s)
- Camille Keisha Mahendra
- Biofunctional Molecule Exploratory Research Group, School of Pharmacy, Monash University Malaysia, Subang Jaya, Malaysia
- Novel Bacteria and Drug Discovery Research Group, Microbiome and Bioresource Research Strength Jeffrey Cheah School of Medicine and Health Sciences, Monash University, Subang Jaya, Malaysia
| | - Loh Teng Hern Tan
- Novel Bacteria and Drug Discovery Research Group, Microbiome and Bioresource Research Strength Jeffrey Cheah School of Medicine and Health Sciences, Monash University, Subang Jaya, Malaysia
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China
| | - Wai Leng Lee
- School of Science, Monash University Malaysia, Subang Jaya, Malaysia
| | - Wei Hsum Yap
- School of Biosciences, Taylor's University, Subang Jaya, Malaysia
| | - Priyia Pusparajah
- Medical Health and Translational Research Group, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Subang Jaya, Malaysia
| | - Liang Ee Low
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Key Laboratory of Biomedical Engineering of the Ministry of Education, College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, China
| | - Siah Ying Tang
- Chemical Engineering Discipline, School of Engineering, Monash University Malaysia, Subang Jaya, Malaysia
- Advanced Engineering Platform, Monash University Malaysia, Subang Jaya, Malaysia
| | - Kok Gan Chan
- International Genome Centre, Jiangsu University, Zhenjiang, China
- Division of Genetics and Molecular Biology, Faculty of Science, Institute of Biological Sciences, University of Malaya, Kuala Lumpur, Malaysia
| | - Learn Han Lee
- Novel Bacteria and Drug Discovery Research Group, Microbiome and Bioresource Research Strength Jeffrey Cheah School of Medicine and Health Sciences, Monash University, Subang Jaya, Malaysia
| | - Bey Hing Goh
- Biofunctional Molecule Exploratory Research Group, School of Pharmacy, Monash University Malaysia, Subang Jaya, Malaysia
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Health and Well-Being Cluster, Global Asia in the 21st Century (GA21) Platform, Monash University Malaysia, Subang Jaya, Malaysia
| |
Collapse
|
69
|
Mansouri RA, Percival SS. Cranberry extract initiates intrinsic apoptosis in HL-60 cells by increasing BAD activity through inhibition of AKT phosphorylation. BMC Complement Med Ther 2020; 20:71. [PMID: 32143616 PMCID: PMC7076838 DOI: 10.1186/s12906-020-2870-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 02/26/2020] [Indexed: 12/17/2022] Open
Abstract
Background Cranberry has been studied as a potential anticancer agent as it is capable of inducing apoptosis within cancer cells. The aim of this study was to better define the mechanism by which cranberry triggers apoptosis in HL-60 cells. Methods The study was carried on cranberry extracts (CB). Anti-apoptotic B-cell lymphoma-2 (BCL-2) and pro-apoptotic BCL-2-associated death promoter death (BAD) proteins in cell lysates were detected through Western blotting techniques. Equivalent protein loading was confirmed through anti-α-tubulin antibody. Results The results showed that treatment of HL-60 cells with CB causes a significant increase in the levels of caspase-9 and caspases-3/7 and increased mitochondrial outer membrane permeability, leading to the release of cytochrome C and Smac. These apoptotic events were associated with a significant decrease in protein kinase B (AKT) phosphorylation, which caused significant increase in BAD de-phosphorylation and promoted a sequence of events that led to intrinsic apoptosis. Conclusion The study findings have described a molecular framework for CB-initiated apoptosis in HL-60 cells and suggested a direction for future in vivo studies investigating the anticancer effect of cranberry.
Collapse
Affiliation(s)
- Rasha A Mansouri
- Department of Biochemistry, King Abdul Aziz University, Jeddah, Saudi Arabia.
| | - Susan S Percival
- Food Science and Human Nutrition Department, University of Florida, Gainesville, FL, 32611, USA
| |
Collapse
|
70
|
Ahmad B, Khan S, Liu Y, Xue M, Nabi G, Kumar S, Alshwmi M, Qarluq AW. Molecular Mechanisms of Anticancer Activities of Puerarin. Cancer Manag Res 2020; 12:79-90. [PMID: 32021425 PMCID: PMC6956866 DOI: 10.2147/cmar.s233567] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Accepted: 12/16/2019] [Indexed: 12/12/2022] Open
Abstract
Medicinal plants are a vital source of natural products (NPs) that can cure cancer through modulation of different pathways, including oxidative stress, extrinsic and intrinsic apoptosis, cell cycle, inflammation, NF-kB, PI3K/AKT/mTOR, AMPK (JNK), MEK/ERK (Raf)-MEK-ERK and autophagy. Puerarin (Pue), an important NP belonging to the isoflavone glycoside group, is derived from Pueraria lobata (Willd.) Ohwi, Pueraria thomsonii Benth, and Pueraria tuberosa (Willd.). This NP was approved by the Chinese Ministry of Health for the treatment of different diseases in 1993, but it was also later reported to exhibit anticancer activity. Pue causes cancer cells death through modulation of different mechanisms including oxidative stress, intrinsic and extrinsic, Survivin and XIAP, PI3K/AKT/mTOR, Ras-Raf-MEK-ERK, JNK, cell cycle, AMPK, NF-kB, inflammation and autophagy pathways. Therefore, this review compiles for the first time the studies about the anticancer mechanism of Pue and provides comprehensive information about the anticancer effects of Pue. This review may serve as a basis for future research and clinical treatment.
Collapse
Affiliation(s)
- Bashir Ahmad
- Department of Pathology and Pathophysiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, Liaoning 116044, People's Republic of China
| | - Suliman Khan
- The Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China.,Henan Medical Key Laboratory of Translational Cerebrovascular Diseases, Zhengzhou, People's Republic of China
| | - Yang Liu
- The Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China.,Henan Medical Key Laboratory of Translational Cerebrovascular Diseases, Zhengzhou, People's Republic of China
| | - Mengzhou Xue
- The Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China.,Henan Medical Key Laboratory of Translational Cerebrovascular Diseases, Zhengzhou, People's Republic of China
| | - Ghulam Nabi
- Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, Shijiazhuang 050024, People's Republic of China
| | - Sunjeet Kumar
- The Key Laboratory of Aquatic Biodiversity and Conservation, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, People's Republic of China
| | - Mohammed Alshwmi
- Department of Clinical Laboratory, The First Affiliated Hospital, Dalian Medical University, Dalian, Liaoning 116044, People's Republic of China
| | - Abdul Wakeel Qarluq
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian, Liaoning 116044, People's Republic of China
| |
Collapse
|
71
|
Abosheasha MA, Abd El Khalik EAM, El-Gowily AH. Indispensable Role of Protein Turnover in Autophagy, Apoptosis and Ubiquitination Pathways. HEAT SHOCK PROTEINS 2020:447-468. [DOI: 10.1007/7515_2020_31] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
|
72
|
Wan Mohd Tajuddin WNB, Lajis NH, Abas F, Othman I, Naidu R. Mechanistic Understanding of Curcumin's Therapeutic Effects in Lung Cancer. Nutrients 2019; 11:E2989. [PMID: 31817718 PMCID: PMC6950067 DOI: 10.3390/nu11122989] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 11/22/2019] [Accepted: 11/30/2019] [Indexed: 12/24/2022] Open
Abstract
Lung cancer is among the most common cancers with a high mortality rate worldwide. Despite the significant advances in diagnostic and therapeutic approaches, lung cancer prognoses and survival rates remain poor due to late diagnosis, drug resistance, and adverse effects. Therefore, new intervention therapies, such as the use of natural compounds with decreased toxicities, have been considered in lung cancer therapy. Curcumin, a natural occurring polyphenol derived from turmeric (Curcuma longa) has been studied extensively in recent years for its therapeutic effects. It has been shown that curcumin demonstrates anti-cancer effects in lung cancer through various mechanisms, including inhibition of cell proliferation, invasion, and metastasis, induction of apoptosis, epigenetic alterations, and regulation of microRNA expression. Several in vitro and in vivo studies have shown that these mechanisms are modulated by multiple molecular targets such as STAT3, EGFR, FOXO3a, TGF-β, eIF2α, COX-2, Bcl-2, PI3KAkt/mTOR, ROS, Fas/FasL, Cdc42, E-cadherin, MMPs, and adiponectin. In addition, limitations, strategies to overcome curcumin bioavailability, and potential side effects as well as clinical trials were also reviewed.
Collapse
Affiliation(s)
- Wan Nur Baitty Wan Mohd Tajuddin
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Selangor Darul Ehsan 47500, Malaysia; (W.N.B.W.M.T.); (I.O.)
| | - Nordin H. Lajis
- Laboratory of Natural Products, Faculty of Science, Universiti Putra Malaysia, UPM, Serdang 43400, Malaysia; (N.H.L.); (F.A.)
| | - Faridah Abas
- Laboratory of Natural Products, Faculty of Science, Universiti Putra Malaysia, UPM, Serdang 43400, Malaysia; (N.H.L.); (F.A.)
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, UPM, Serdang 43400, Malaysia
| | - Iekhsan Othman
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Selangor Darul Ehsan 47500, Malaysia; (W.N.B.W.M.T.); (I.O.)
| | - Rakesh Naidu
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Selangor Darul Ehsan 47500, Malaysia; (W.N.B.W.M.T.); (I.O.)
| |
Collapse
|
73
|
Zhuang Q, Dai F, Zhao X, Shao Y, Guo M, Lv Z, Li C, Zhang W. Cloning and characterization of the virulence factor Hop from Vibrio splendidus. Microb Pathog 2019; 139:103900. [PMID: 31790795 DOI: 10.1016/j.micpath.2019.103900] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 11/11/2019] [Accepted: 11/28/2019] [Indexed: 10/25/2022]
Abstract
BACKGROUND Vibrio splendidus is an aquaculture pathogen that can cause skin ulcer syndrome (SUS) in Apostichopus japonicus. HopPmaJ is a type III system effector (T3SE) that has been reported to be an important virulence factor. In this study, a gene named hop, which encodes HopPmaJ in V. splendidus was cloned and its cytotoxicity to coelomocytes and its effects on the expression of immune-related genes in A. japonicus were characterized. METHODS Real time reverse transcription PCR (RT-PCR) was used to determine the expression of the hop gene under various conditions. To obtain the purified Hop, hop gene was conditionally expressed in Escherichia coli BL21(DE3) and was purified by GST tag. The cytotoxicity of Hop to coelomocyte was determined using MTT method, and the effect of Hop on the expression of immune-related genes was determined using real time RT-PCR. RESULTS The deduced amino acid sequence of Hop from V. splendidus shared 84%-96% homology with those of Hops from other Vibrio spp. The expression of hop gene was induced not only by host-pathogen contact but also by high cell density. Purified recombinant Hop (rHop) showed cytotoxicity to the coelomocyte of A. japonicus. The cell viability decreased to approximately 42%, 26%, 32%, 30% and 20%, when 30, 50, 60, 80 and 100 μL of purified rHop was added, respectively. After being injected with rHop, the expression levels of immune-related genes that encode complement component (C1q) and caspase were significantly increased, and the production of reactive oxygen species were also increased in A. japonicus. CONCLUSION Our results indicated that Hop not only contributed to the cytotoxicity to coelomocyte, but also caused immune response in A. japonicus.
Collapse
Affiliation(s)
- Qiuting Zhuang
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, PR China
| | - Fa Dai
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, PR China
| | - Xuelin Zhao
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, PR China
| | - Yina Shao
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, PR China
| | - Ming Guo
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, PR China
| | - Zhimeng Lv
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, PR China
| | - Chenghua Li
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, PR China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266071, PR China
| | - Weiwei Zhang
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, PR China.
| |
Collapse
|
74
|
Zong H, Zhou H, Xiang Y, Wu G. miR-125b suppresses cellular proliferation by targeting c-FLIP in gallbladder carcinoma. Oncol Lett 2019; 18:6822-6828. [PMID: 31788125 DOI: 10.3892/ol.2019.11000] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 08/13/2019] [Indexed: 01/07/2023] Open
Abstract
Gallbladder carcinoma (GBC) is the most common malignant tumor of the biliary tract. The incidence rate of gallbladder cancer ranks sixth among gastrointestinal types of cancer, and its incidence is increasing each year. Further clarification of the pathogenesis of GBC is essential, and identification of novel effective treatments is required. It has been previously demonstrated that high expression of the anti-apoptotic protein cellular Fas-associated death domain-like interleukin-1-converting enzyme inhibitory protein (c-FLIP) in GBC inhibited apoptosis in gallbladder cancer cells. In subsequent experiments, it was observed that microRNA (miR)-125b could target c-FLIP and inhibit the protein expression of c-FLIP by binding to the 3'untranslated regions of c-FLIP mRNA. In addition, the expression of miR-125b in GBC was significantly decreased, and the growth of gallbladder cancer cells was inhibited by the overexpression of miR-125b. The present study demonstrated that miR-125b could suppress the proliferation of gallbladder cancer cells by targeting c-FLIP. c-FLIP enriched the target gene pathway of miR-125b and may serve as a novel target for the treatment of GBC.
Collapse
Affiliation(s)
- Huajie Zong
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Huading Zhou
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Yang Xiang
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Gang Wu
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| |
Collapse
|
75
|
Yu XH, Hong XQ, Mao QC, Chen WH. Biological effects and activity optimization of small-molecule, drug-like synthetic anion transporters. Eur J Med Chem 2019; 184:111782. [DOI: 10.1016/j.ejmech.2019.111782] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 10/08/2019] [Accepted: 10/09/2019] [Indexed: 12/27/2022]
|
76
|
Kim JH, Nam GS, Kim SH, Ryu DS, Lee DS. Orostachys japonicus exerts antipancreatic cancer activity through induction of apoptosis and cell cycle arrest in PANC-1 cells. Food Sci Nutr 2019; 7:3549-3559. [PMID: 31763005 PMCID: PMC6848830 DOI: 10.1002/fsn3.1207] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 07/30/2019] [Accepted: 08/12/2019] [Indexed: 12/11/2022] Open
Abstract
Targeted therapy at the molecular level is important for pancreatic cancer treatment. This study looked over the anticancer activity of Orostachys japonicus in a human pancreatic cancer cell line, PANC-1. An ethyl acetate fraction containing quercetin, kaempferol, and flavonol glycosides from O. japonicus (OJE) exhibited significant anticancer activity against the PANC-1. OJE activated caspase-3, caspase-8, and caspase-9, leading to the induction of both intrinsic and extrinsic apoptosis pathways. It also inhibited cyclin D1, cyclin B1, and cyclin-dependent kinase 4, representing cell cycle arrest at both G1/S and G2/M phases. In addition, OJE phosphorylated MAPKs such as p38, JNK, and ERK, which are important upstream signaling factors in apoptosis and arrest of cell cycle inducing system. In conclusion, OJE effectively exerted antipancreatic cancer activity via induction of apoptosis directed by both intrinsic and extrinsic pathways and arrest of cell cycle regulated at both G1/S and G2/M stages, which is activated by MAPKs, p38, JNK, and ERK.
Collapse
Affiliation(s)
- Ji Hyun Kim
- Department of Smart Foods and DrugsGraduate School of Inje UniversityGimhaeKorea
| | - Gi Suk Nam
- Department of Smart Foods and DrugsGraduate School of Inje UniversityGimhaeKorea
| | - Sung Hyun Kim
- Department of Smart Foods and DrugsGraduate School of Inje UniversityGimhaeKorea
| | - Deok Seon Ryu
- Department of Biomedical Laboratory ScienceSoonchunhyang UniversityAsanKorea
| | - Dong Seok Lee
- Department of Smart Foods and DrugsGraduate School of Inje UniversityGimhaeKorea
| |
Collapse
|
77
|
Lee MH, Liu KH, Thomas JL, Chen JR, Lin HY. Immunotherapy of Hepatocellular Carcinoma with Magnetic PD-1 Peptide-Imprinted Polymer Nanocomposite and Natural Killer Cells. Biomolecules 2019; 9:biom9110651. [PMID: 31731492 PMCID: PMC6920774 DOI: 10.3390/biom9110651] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 10/23/2019] [Accepted: 10/23/2019] [Indexed: 12/11/2022] Open
Abstract
Programmed cell death protein 1 (PD-1) is a biomarker on the surface of cells with a role in promoting self-tolerance by suppressing the inflammatory activity of T cells. In this work, one peptide of PD-1 was used as the template for molecular imprinting to form magnetic peptide-imprinted poly(ethylene-co-vinyl alcohol) composite nanoparticles (MPIP NPs). The nanoparticles were characterized by dynamic light scattering (DLS), high-performance liquid chromatography (HPLC), Brunauer–Emmett–Teller (BET) analysis, and superconducting quantum interference device (SQUID) analysis. Natural killer 92 (NK-92) cells were added to these composite nanoparticles and then incubated with human hepatoma (HepG2) cells. The viability and the apoptosis pathway of HepG2 were then studied using cell counting kit-8 (CCK8) and quantitative real-time polymerase chain reaction (qRT-PCR), respectively. These nanoparticles were found to significantly enhance the activity of natural killer cells toward HepG2 cells by increasing the expression of nuclear factor kappa B (NF-κB), caspase 8, and especially caspase 3.
Collapse
Affiliation(s)
- Mei-Hwa Lee
- Department of Materials Science and Engineering, I-Shou University, Kaohsiung 84001, Taiwan;
| | - Kai-Hsi Liu
- Department of Internal Medicine, Division of Cardiology, Zuoying Branch of Kaohsiung Armed Forces General Hospital, Kaohsiung 813, Taiwan;
- Department of Chemical and Materials Engineering, National University of Kaohsiung, Kaohsiung 81148, Taiwan;
| | - James L. Thomas
- Department of Physics and Astronomy, University of New Mexico, Albuquerque, NM 87131, USA;
| | - Jyun-Ren Chen
- Department of Chemical and Materials Engineering, National University of Kaohsiung, Kaohsiung 81148, Taiwan;
| | - Hung-Yin Lin
- Department of Chemical and Materials Engineering, National University of Kaohsiung, Kaohsiung 81148, Taiwan;
- Correspondence: ; Tel.: +886-(7)-591-9455 or +886-(912)-178-751
| |
Collapse
|
78
|
Sun HR, Wang S, Yan SC, Zhang Y, Nelson PJ, Jia HL, Qin LX, Dong QZ. Therapeutic Strategies Targeting Cancer Stem Cells and Their Microenvironment. Front Oncol 2019; 9:1104. [PMID: 31709180 PMCID: PMC6821685 DOI: 10.3389/fonc.2019.01104] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 10/07/2019] [Indexed: 12/12/2022] Open
Abstract
Cancer stem cells (CSCs) have been demonstrated in a variety of tumors and are thought to act as a clonogenic core for the genesis of new tumor growth. This small subpopulation of cancer cells has been proposed to help drive tumorigenesis, metastasis, recurrence and conventional therapy resistance. CSCs show self-renewal and flexible clonogenic properties and help define specific tumor microenvironments (TME). The interaction between CSCs and TME is thought to function as a dynamic support system that fosters the generation and maintenance of CSCs. Investigation of the interaction between CSCs and the TME is shedding light on the biologic mechanisms underlying the process of tumor malignancy, metastasis, and therapy resistance. We summarize recent advances in CSC biology and their environment, and discuss the challenges and future strategies for targeting this biology as a new therapeutic approach.
Collapse
Affiliation(s)
- Hao-Ran Sun
- Department of General Surgery, Cancer Metastasis Institute, Institutes of Biomedical Sciences, Huashan Hospital, Fudan University, Shanghai, China
| | - Shun Wang
- Department of General Surgery, Cancer Metastasis Institute, Institutes of Biomedical Sciences, Huashan Hospital, Fudan University, Shanghai, China
| | - Shi-Can Yan
- Department of General Surgery, Cancer Metastasis Institute, Institutes of Biomedical Sciences, Huashan Hospital, Fudan University, Shanghai, China
| | - Yu Zhang
- Department of General Surgery, Cancer Metastasis Institute, Institutes of Biomedical Sciences, Huashan Hospital, Fudan University, Shanghai, China
| | - Peter J. Nelson
- Medizinische Klinik und Poliklinik IV, Ludwig-Maximilian-University (LMU), Munich, Germany
| | - Hu-Liang Jia
- Department of General Surgery, Cancer Metastasis Institute, Institutes of Biomedical Sciences, Huashan Hospital, Fudan University, Shanghai, China
| | - Lun-Xiu Qin
- Department of General Surgery, Cancer Metastasis Institute, Institutes of Biomedical Sciences, Huashan Hospital, Fudan University, Shanghai, China
| | - Qiong-Zhu Dong
- Department of General Surgery, Cancer Metastasis Institute, Institutes of Biomedical Sciences, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
79
|
Zheng H, Zhang Y, Zhan Y, Liu S, Lu J, Wen Q, Fan S. Expression of DR5 and c‑FLIP proteins as novel prognostic biomarkers for non‑small cell lung cancer patients treated with surgical resection and chemotherapy. Oncol Rep 2019; 42:2363-2370. [PMID: 31638235 PMCID: PMC6859453 DOI: 10.3892/or.2019.7355] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 09/12/2019] [Indexed: 12/16/2022] Open
Abstract
TRAIL-R2 (DR5), one of the death receptors, can activate the extrinsic apoptosis pathway, while cellular FLICE-inhibitory protein (c-FLIP) can inhibit this pathway. Both of them play important roles in the occurrence and development of most tumors. To date, there is no relevant report concerning the relationship between expression of DR5 and c-FLIP protein and clinicopathological/prognostic implications in patients with non-small cell lung cancer (NSCLC) treated with surgical resection and chemotherapy. Thus, the aim of the present study was to investigate the potential prognostic significance of DR5 and c-FLIP in NSCLC patients and their predictive roles in the chemotherapeutic response. In the present study, DR5 and c-FLIP were detected by immunohistochemistry (IHC) in tissue microarrays of NSCLC. The results showed that the expression levels of DR5 and c-FLIP were significantly higher in lung squamous cell carcinoma (SCC) and lung adenocarcinoma (ADC) tissues compared with levels noted in the non-cancerous control lung tissues (all P<0.05). In addition, DR5 expression was significantly increased in lung ADC (P<0.001), whereas, c-FLIP was higher in lung SCC (P<0.001) and smoker patients with clinical stage III (P=0.019, P=0.016, respectively). In addition, NSCLC patients with overexpression of DR5 and loss of c-FLIP expression exhibited a higher overall survival (OS) rate as determined by Kaplan-Meier analysis (P=0.029, P=0.038, respectively). Multivariate analysis confirmed that high expression of DR5 and loss of c-FLIP expression were independent favorable prognostic factors for NSCLC patients (P=0.016, P=0.035, respectively). In conclusion, overexpression of DR5 and loss of c-FLIP expression may serve as novel favorable prognostic biomarkers for NSCLC patients treated with chemotherapy after radical resection and used as predictors for tumor response to chemotherapy drugs.
Collapse
Affiliation(s)
- Hongmei Zheng
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Yuting Zhang
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Yuting Zhan
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Sile Liu
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Junmi Lu
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Qiuyuan Wen
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Songqing Fan
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| |
Collapse
|
80
|
Sadeghnezhad G, Romão E, Bernedo-Navarro R, Massa S, Khajeh K, Muyldermans S, Hassania S. Identification of New DR5 Agonistic Nanobodies and Generation of Multivalent Nanobody Constructs for Cancer Treatment. Int J Mol Sci 2019; 20:E4818. [PMID: 31569768 PMCID: PMC6801735 DOI: 10.3390/ijms20194818] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 09/24/2019] [Accepted: 09/25/2019] [Indexed: 12/24/2022] Open
Abstract
Current cancer therapeutics suffer from a lack of specificity in targeting tumor cells and cause severe side effects. Therefore, the design of highly specialized drugs comprising antibody derivatives inducing apoptosis in targeted cancer cells is considered to be a promising strategy. Drugs acting on death receptor 5 (DR5) such as DR5 agonist antibodies replacing "TNF-related apoptosis-inducing ligand" (TRAIL) offer feasible opportunities in this direction. Although such agonists provided good antitumor activity in preclinical studies, they were less effective in clinical studies, possibly due to a disturbed Fc interaction with Fc-γ receptors. Thus, multimerized antigen binding fragments without Fc have been proposed to increase their efficacy. We generated nanobodies (Nbs), recombinant variable domains of heavy chain-only antibodies of camelids, against the DR5 ectodomain. Nb24 and Nb28 had an affinity in the nM and sub-nM range, but only Nb28 competes with TRAIL for binding to DR5. Bivalent, trivalent, and tetravalent constructs were generated, as well as an innovative pentameric Nb complex, to provoke avidity effects. In our cellular assays, these trimeric, tetrameric, and pentameric Nbs have a higher apoptotic capacity than monomeric Nbs and seem to mimic the activity of the natural TRAIL ligand on various cancer cells.
Collapse
Affiliation(s)
- Golnaz Sadeghnezhad
- Faculty of Biological Sciences, Tarbiat Modares University, Teheran 14115-331, Iran.
- Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels 1050, Belgium.
| | - Ema Romão
- Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels 1050, Belgium.
| | - Robert Bernedo-Navarro
- Laboratory of Environmental and Life Sciences, University of Nova Gorica, Nova Gorica 5000, Slovenia.
| | - Sam Massa
- Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels 1050, Belgium.
| | - Khosro Khajeh
- Faculty of Biological Sciences, Tarbiat Modares University, Teheran 14115-331, Iran.
| | - Serge Muyldermans
- Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels 1050, Belgium.
| | - Sadegh Hassania
- Faculty of Biological Sciences, Tarbiat Modares University, Teheran 14115-331, Iran.
| |
Collapse
|
81
|
Wong SHM, Kong WY, Fang CM, Loh HS, Chuah LH, Abdullah S, Ngai SC. The TRAIL to cancer therapy: Hindrances and potential solutions. Crit Rev Oncol Hematol 2019; 143:81-94. [PMID: 31561055 DOI: 10.1016/j.critrevonc.2019.08.008] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 08/29/2019] [Accepted: 08/29/2019] [Indexed: 12/15/2022] Open
Abstract
Apoptosis is an ordered and orchestrated cellular process that occurs in physiological and pathological conditions. Resistance to apoptosis is a hallmark of virtually all malignancies. Despite being a cause of pathological conditions, apoptosis could be a promising target in cancer treatment. Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL), also known as Apo-2 ligand (Apo2L), is a member of TNF cytokine superfamily. It is a potent anti-cancer agent owing to its specific targeting towards cancerous cells, while sparing normal cells, to induce apoptosis. However, resistance occurs either intrinsically or after multiple treatments which may explain why cancer therapy fails. This review summarizes the apoptotic mechanisms via extrinsic and intrinsic apoptotic pathways, as well as the apoptotic resistance mechanisms. It also reviews the current clinically tested recombinant human TRAIL (rhTRAIL) and TRAIL receptor agonists (TRAs) against TRAIL-Receptors, TRAIL-R1 and TRAIL-R2, in which the outcomes of the clinical trials have not been satisfactory. Finally, this review discusses the current strategies in overcoming resistance to TRAIL-induced apoptosis in pre-clinical and clinical settings.
Collapse
Affiliation(s)
- Sonia How Ming Wong
- School of Biosciences, Faculty of Science and Engineering, University of Nottingham Malaysia, 43500, Semenyih, Selangor, Malaysia
| | - Wei Yang Kong
- School of Biosciences, Faculty of Science and Engineering, University of Nottingham Malaysia, 43500, Semenyih, Selangor, Malaysia
| | - Chee-Mun Fang
- Division of Biomedical Sciences, School of Pharmacy, University of Nottingham Malaysia, 43500, Semenyih, Selangor, Malaysia
| | - Hwei-San Loh
- School of Biosciences, Faculty of Science and Engineering, University of Nottingham Malaysia, 43500, Semenyih, Selangor, Malaysia
| | - Lay-Hong Chuah
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia; Advanced Engineering Platform, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia
| | - Syahril Abdullah
- Medical Genetics Laboratory, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, 43400 UPM, Malaysia; UPM-MAKNA Cancer Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Selangor, 43400 UPM, Malaysia
| | - Siew Ching Ngai
- School of Biosciences, Faculty of Science and Engineering, University of Nottingham Malaysia, 43500, Semenyih, Selangor, Malaysia.
| |
Collapse
|
82
|
Antiproliferative and apoptotic activities of 8-prenylnaringenin against human colon cancer cells. Life Sci 2019; 232:116633. [DOI: 10.1016/j.lfs.2019.116633] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 06/26/2019] [Accepted: 07/03/2019] [Indexed: 11/22/2022]
|
83
|
Emdad L, Bhoopathi P, Talukdar S, Pradhan AK, Sarkar D, Wang XY, Das SK, Fisher PB. Recent insights into apoptosis and toxic autophagy: The roles of MDA-7/IL-24, a multidimensional anti-cancer therapeutic. Semin Cancer Biol 2019; 66:140-154. [PMID: 31356866 DOI: 10.1016/j.semcancer.2019.07.013] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 06/21/2019] [Accepted: 07/19/2019] [Indexed: 12/18/2022]
Abstract
Apoptosis and autophagy play seminal roles in maintaining organ homeostasis. Apoptosis represents canonical type I programmed cell death. Autophagy is viewed as pro-survival, however, excessive autophagy can promote type II cell death. Defective regulation of these two obligatory cellular pathways is linked to various diseases, including cancer. Biologic or chemotherapeutic agents, which can reprogram cancer cells to undergo apoptosis- or toxic autophagy-mediated cell death, are considered effective tools for treating cancer. Melanoma differentiation associated gene-7 (mda-7) selectively promotes these effects in cancer cells. mda-7 was identified more than two decades ago by subtraction hybridization showing elevated expression during induction of terminal differentiation of metastatic melanoma cells following treatment with recombinant fibroblast interferon and mezerein (a PKC activating agent). MDA-7 was classified as a member of the IL-10 gene family based on its chromosomal location, and the presence of an IL-10 signature motif and a secretory sequence, and re-named interleukin-24 (MDA-7/IL-24). Multiple studies have established MDA-7/IL-24 as a potent anti-cancer agent, which when administered at supra-physiological levels induces growth arrest and cell death through apoptosis and toxic autophagy in a wide variety of tumor cell types, but not in corresponding normal/non-transformed cells. Furthermore, in a phase I/II clinical trial, MDA-7/IL-24 administered by means of a non-replicating adenovirus was well tolerated and displayed significant clinical activity in patients with multiple advanced cancers. This review examines our current comprehension of the role of MDA-7/IL-24 in mediating cancer-specific cell death via apoptosis and toxic autophagy.
Collapse
Affiliation(s)
- Luni Emdad
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA.
| | - Praveen Bhoopathi
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA
| | - Sarmistha Talukdar
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA
| | - Anjan K Pradhan
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA
| | - Devanand Sarkar
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA
| | - Xiang-Yang Wang
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA
| | - Swadesh K Das
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA
| | - Paul B Fisher
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA.
| |
Collapse
|
84
|
Mortezaei Z, Tavallaei M, Hosseini SM. Considering smoking status, coexpression network analysis of non-small cell lung cancer at different cancer stages, exhibits important genes and pathways. J Cell Biochem 2019; 120:19172-19185. [PMID: 31271232 DOI: 10.1002/jcb.29246] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 05/23/2019] [Indexed: 02/01/2023]
Abstract
Non-small cell lung cancer (NSCLC) is the most common subtype of lung cancer among smokers, nonsmokers, women, and young individuals. Tobacco smoking and different stages of the NSCLC have important roles in cancer evolution and require different treatments. Existence of poorly effective therapeutic options for the NSCLC brings special attention to targeted therapies by considering genetic alterations. In this study, we used RNA-Seq data to compare expression levels of RefSeq genes and to find some genes with similar expression levels. We utilized the "Weighted Gene Co-expression Network Analysis" method for three different datasets to create coexpressed genetic modules having relations with the smoking status and different stages of the NSCLC. Our results indicate seven important genetic modules having important associations with the smoking status and cancer stages. Based on investigated genetic modules and their biological explanation, we then identified 13 newly candidate genes and 7 novel transcription factors in association with the NSCLC, the smoking status, and cancer stages. We then examined those results using other datasets and explained our results biologically to illustrate some important genes in relation with the smoking status and metastatic stage of the NSCLC that can bring some crucial information about cancer evolution. Our genetic findings also can be used as some therapeutic targets for different clinical conditions of the NSCLC.
Collapse
Affiliation(s)
- Zahra Mortezaei
- Human Genetic Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mahmood Tavallaei
- Human Genetic Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Sayed Mostafa Hosseini
- Human Genetic Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
85
|
Antitumor activity of methyl (Z)-2-(isothioureidomethyl)-2-pentenoate hydrobromide against leukemia cell lines via mitotic arrest and apoptotic pathways. Biochim Biophys Acta Gen Subj 2019; 1863:1332-1342. [PMID: 31170497 DOI: 10.1016/j.bbagen.2019.05.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 05/28/2019] [Accepted: 05/29/2019] [Indexed: 01/07/2023]
Abstract
In a previous study, we described a series of 28 aryl- and alkyl-substituted isothiouronium salts with antitumor activity and selectivity toward a leukemia cell line. Among the synthesized compounds, methyl (Z)-2-(isothioureidomethyl)-2-pentenoate hydrobromide (IS-MF08) showed conspicuous activity. In the present study, we investigated the mechanism of action of IS-MF08. Our results showed that its mechanism most likely is related with the membrane receptor Fas and subsequent activation of the extrinsic cell death pathway, triggered by a decrease in the levels of the anti-apoptotic protein Bcl-2 and caspase-8 and -3 cascade activation, causing DNA damage and mitotic arrest. IS-MF08 also caused an increase in intracellular ROS, endoplasmic reticulum (ER) stress, and mitochondrial membrane permeabilization, resulting in organelle degradation as an attempt to reestablish cell homeostasis. Furthermore, cells exposed to IS-MF08 combined to an autophagy inhibitor were less susceptible to compound's cytotoxicity, suggesting that autophagy makes part of its mechanism of action. These data support the hypothesis that IS-MF08 acts by the apoptosis extrinsic pathway and possibly by autophagy as mechanisms of cell death.
Collapse
|
86
|
Sakanashi F, Shintani M, Tsuneyoshi M, Ohsaki H, Kamoshida S. Apoptosis, necroptosis and autophagy in colorectal cancer: Associations with tumor aggressiveness and p53 status. Pathol Res Pract 2019; 215:152425. [PMID: 31097354 DOI: 10.1016/j.prp.2019.04.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 04/05/2019] [Accepted: 04/26/2019] [Indexed: 01/07/2023]
Abstract
OBJECTIVE Cleaved caspase-3 (CC3), phosphorylated-mixed-lineage kinase domain-like protein (p-MLKL), and microtubule-associated protein-1 light chain-3B (LC3B) have pivotal functions in apoptosis, necroptosis, and autophagy, respectively. In vitro studies have shown that interaction of these proteins are complex and their roles in cancer can be influenced by many factors. However, these findings are not adequately assessed in human tissues. Here, we determined CC3, p-MLKL, and LC3B expression in colorectal cancers (CRCs), and assessed their associations with clinicopathological parameters, and with KRAS and p53 status. METHODS We immunohistochemically assessed 113 CRC specimens for levels of CC3, p-MLKL, LC3B, and p53. KRAS gene status was analyzed using the Scorpion- amplification refractory mutation system. RESULTS High levels of CC3 (CC3High) and LC3B (LC3BHigh) were detected in 38% and 35% of the 113 CRCs, respectively, but no or only a few p-MLKL-positive cells were observed in any of the tumors. CC3High was significantly associated with high pT status (P = 0.03), vascular invasion (P = 0.03) and high pStage (P = 0.04) and was marginally associated with lymph node (P = 0.06) and distant metastases (P = 0.06). LC3BHigh was also significantly associated with high pT status (P = 0.02) and lymphatic invasion (P = 0.002), and was marginally associated with nerve plexus invasion (P = 0.06). In combined analysis, compared with CC3Low/LC3BLow tumors, tumors that were either CC3High, LC3BHigh, or both were significantly associated with high pT status (P = 0.0007), lymphatic invasion (P = 0.03), vascular invasion (P = 0.003), distant metastasis (P = 0.04) and high pStage (P = 0.04). LC3BHigh was significantly associated with a mutant-type expression pattern of p53 (P = 0.003). CONCLUSION To the best of our knowledge, this is the first study to examine the combination of CC3/LC3B and p-MLKL expression in clinical CRC samples and to correlate these expression data with clinicopathological parameters and EGFR and p53 status. Our results suggest that necroptosis is a rare process in CRC, apoptosis and autophagy are upregulated in aggressive CRCs, and p53 mutation may lead to the upregulation of autophagy.
Collapse
Affiliation(s)
- Fuminori Sakanashi
- Laboratory of Pathology, Department of Medical Biophysics, Kobe University Graduate School of Health Sciences, 7-10-2 Tomogaoka, Suma, Kobe, Hyogo 654-0142, Japan; Department of Diagnostic Pathology, Fukuoka Sanno Hospital, 3-6-45 Momochihama, Sawara, Fukuoka 814-0001, Japan.
| | - Michiko Shintani
- Department of Medical Technology, Kobe Tokiwa University, 2-6-2 Ohtani, Nagata, Kobe, Hyogo 653-0838, Japan
| | - Masazumi Tsuneyoshi
- Department of Diagnostic Pathology, Fukuoka Sanno Hospital, 3-6-45 Momochihama, Sawara, Fukuoka 814-0001, Japan
| | - Hiroyuki Ohsaki
- Laboratory of Pathology, Department of Medical Biophysics, Kobe University Graduate School of Health Sciences, 7-10-2 Tomogaoka, Suma, Kobe, Hyogo 654-0142, Japan
| | - Shingo Kamoshida
- Laboratory of Pathology, Department of Medical Biophysics, Kobe University Graduate School of Health Sciences, 7-10-2 Tomogaoka, Suma, Kobe, Hyogo 654-0142, Japan
| |
Collapse
|
87
|
Ahmad B, Khan S, Nabi G, Gamallat Y, Su P, Jamalat Y, Duan P, Yao L. Natural gypenosides: targeting cancer through different molecular pathways. Cancer Manag Res 2019; 11:2287-2297. [PMID: 31114315 PMCID: PMC6497488 DOI: 10.2147/cmar.s185232] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 02/08/2019] [Indexed: 01/22/2023] Open
Abstract
The second foremost cause of mortality around the word is cancer. Conventional therapies, such as radiation, surgery, and chemotherapy have limited accessibility owing to secondary resistance. Therefore, convenient, safe, and nonresistant drugs are urgently needed. Plant-derived natural products have attracted considerable interest owing to their high efficacy, low toxicity, and convenience. Gypenosides (Gyp) inhibit invasion, migration, metastasis, and proliferation and induce apoptosis in different cancers, including oral, lung, colorectal, hepatocellular, and leukemic cancers through different molecular pathways. This review summarizes Gyp studies on cancer to serve as a reference for further research and clinical trials.
Collapse
Affiliation(s)
- Bashir Ahmad
- Henan Provincial Engineering Research Center for Health Products of Livestock and Poultry, Nanyang Normal University, Nanyang, People's Republic of China.,College of Basic Medical Sciences, Dalian Medical University, Dalian City, People's Republic of China
| | - Suliman Khan
- Henan Provincial Engineering Research Center for Health Products of Livestock and Poultry, Nanyang Normal University, Nanyang, People's Republic of China.,Collaborative Innovation Centre of Water Security for Water Source Region of Mid-line of South-to-North Diversion Project of Henan Province/Key Laboratory of Ecological Security for Water Source Region of Mid-line of South-to-North Diversion Project of Henan Province, School of Agricultural Engineering, Nanyang Normal University, Nanyang, People's Republic of China.,University of Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Ghulam Nabi
- University of Chinese Academy of Sciences, Beijing, People's Republic of China.,Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei, People's Republic of China
| | - Yaser Gamallat
- College of Basic Medical Sciences, Dalian Medical University, Dalian City, People's Republic of China
| | - Pengyu Su
- College of Basic Medical Sciences, Dalian Medical University, Dalian City, People's Republic of China
| | - Yazeed Jamalat
- College of Basic Medical Sciences, Dalian Medical University, Dalian City, People's Republic of China
| | - Pengfei Duan
- Henan Provincial Engineering Research Center for Health Products of Livestock and Poultry, Nanyang Normal University, Nanyang, People's Republic of China.,Collaborative Innovation Centre of Water Security for Water Source Region of Mid-line of South-to-North Diversion Project of Henan Province/Key Laboratory of Ecological Security for Water Source Region of Mid-line of South-to-North Diversion Project of Henan Province, School of Agricultural Engineering, Nanyang Normal University, Nanyang, People's Republic of China
| | - Lunguang Yao
- Henan Provincial Engineering Research Center for Health Products of Livestock and Poultry, Nanyang Normal University, Nanyang, People's Republic of China.,Collaborative Innovation Centre of Water Security for Water Source Region of Mid-line of South-to-North Diversion Project of Henan Province/Key Laboratory of Ecological Security for Water Source Region of Mid-line of South-to-North Diversion Project of Henan Province, School of Agricultural Engineering, Nanyang Normal University, Nanyang, People's Republic of China
| |
Collapse
|
88
|
Carfilzomib enhances cisplatin-induced apoptosis in SK-N-BE(2)-M17 human neuroblastoma cells. Sci Rep 2019; 9:5039. [PMID: 30911132 PMCID: PMC6434076 DOI: 10.1038/s41598-019-41527-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 03/06/2019] [Indexed: 12/14/2022] Open
Abstract
Neuroblastoma is a solid malignant tumor of the sympathetic nervous system, which accounts for 8–10% of childhood cancers. Considering the overall high risk and poor prognosis associated with neuroblastoma, effective therapeutics should be developed to improve patient survival and quality of life. A recent study showed that a proteasome inhibitor, carfilzomib (CFZ), reduced cell viability of SK-N-BE(2)-M17 neuroblastoma cells. Therefore, we investigated the molecular mechanisms by which CFZ lower the cell viability of neuroblastoma cells. CFZ reduced cell viability via cell cycle arrest at G2/M and apoptosis, which involved caspase activation (caspases-8, 9, 4, and 3), endoplasmic reticulum stress, reactive oxygen species production, mitochondrial membrane potential loss, and autophagy in a dose- and time-dependent manner. The effect of CFZ was additive to that of cisplatin (Cis), a well-known chemotherapeutic drug, in terms of cell viability reduction, cell cycle arrest, and apoptosis. Importantly, the additive effect of CFZ was maintained in Cis-resistant neuroblastoma cells. These results suggest that CFZ can be used in combination therapy for patients with neuroblastoma to overcome the resistance and adverse side effects of Cis.
Collapse
|
89
|
Poondla N, Chandrasekaran AP, Heese K, Kim KS, Ramakrishna S. CRISPR-mediated upregulation of DR5 and downregulation of cFLIP synergistically sensitize HeLa cells to TRAIL-mediated apoptosis. Biochem Biophys Res Commun 2019; 512:60-65. [PMID: 30862357 DOI: 10.1016/j.bbrc.2019.03.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 03/03/2019] [Indexed: 12/17/2022]
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has received attention as an anticancer therapy because it mediates apoptosis of several cancer cell types but not normal human cell types. In this study, we implemented genome editing techniques to upregulate DR5 and downregulate cFLIP in HeLa cells to stimulate TRAIL-induced apoptosis. We designed and validated sgRNAs to enrich the endogenous level of DR5 by dead Cas9 (dCas9). Similarly, we designed two sgRNAs to disrupt the cFLIP gene by CRISPR/Cas9. We analyzed the effect of TRAIL on tumor cells by co-transfecting HeLa cells with the best combinations of sgRNAs regulating DR5 and cFLIP genes. TRAIL-induced apoptosis in HeLa cells was evaluated by the γH2AX foci formation assay to check for double-strand break and propidium iodide and Annexin V staining to quantify apoptotic cells. Viable cells were identified by CCK-8 assay, and cleaved-PARP level was evaluated by Western blot. This is the first study to demonstrate that genome editing techniques can be used as an effective combinatorial treatment strategy to induce apoptosis of cancer cells. In particular, enhancement of DR5 expression and inhibition of cFLIP expression by genome editing had a synergistic effect of inhibiting proliferation and inducing apoptosis in TRAIL-resistant HeLa cells. These results suggest that combinatorial treatment strategies mediated by the CRISPR/Cas9 system may be effective for design of other human TRAIL-resistant cell types.
Collapse
Affiliation(s)
- Naresh Poondla
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, South Korea
| | | | - Klaus Heese
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, South Korea; College of Medicine, Hanyang University, Seoul, South Korea
| | - Kye-Seong Kim
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, South Korea; College of Medicine, Hanyang University, Seoul, South Korea.
| | - Suresh Ramakrishna
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, South Korea; College of Medicine, Hanyang University, Seoul, South Korea.
| |
Collapse
|
90
|
Zhang S, Cao L, Wang ZR, Li Z, Ma J. Anti-cancer effect of toosendanin and its underlying mechanisms. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2019; 21:270-283. [PMID: 29629572 DOI: 10.1080/10286020.2018.1451516] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 03/08/2018] [Accepted: 03/08/2018] [Indexed: 06/08/2023]
Abstract
Toosendanin (TSN) is a triterpenoid purified from the medicinal herb Melia toosendan Sieb. et Zucc and has been used as an insecticide for decades. Recent studies have attracted increasing interest of TSN due to its novel anti-cancer effect in diverse cancer models. The broad spectrum anti-cancer activity suggests that TSN inhibits multiple pathways/targets that are critical for cancer cell survival and proliferation. Our recent study indicated that TSN has anti-cancer effect in glioblastoma through induction of estrogen receptor β (ERβ) and p53. This review highlights the anti-cancer efficacy of TSN and provides proof-of-principle insight into the underlying mechanisms.
Collapse
Affiliation(s)
- Sha Zhang
- a Department of Basic Medicine , Shaanxi University of Chinese Medicine , Xianyang 712046 , China
| | - Liang Cao
- b Department of Traditional Chinese Medicine, Xijing Hospital , Fourth Military Medical University , Xi'an 710032 , China
| | - Zong-Ren Wang
- b Department of Traditional Chinese Medicine, Xijing Hospital , Fourth Military Medical University , Xi'an 710032 , China
| | - Zhe Li
- c Second Clinical Medical College , Shaanxi University of Chinese Medicine , Xianyang 712046 , China
| | - Jing Ma
- b Department of Traditional Chinese Medicine, Xijing Hospital , Fourth Military Medical University , Xi'an 710032 , China
| |
Collapse
|
91
|
Pan L, Fu TM, Zhao W, Zhao L, Chen W, Qiu C, Liu W, Liu Z, Piai A, Fu Q, Chen S, Wu H, Chou JJ. Higher-Order Clustering of the Transmembrane Anchor of DR5 Drives Signaling. Cell 2019; 176:1477-1489.e14. [PMID: 30827683 DOI: 10.1016/j.cell.2019.02.001] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 11/14/2018] [Accepted: 01/29/2019] [Indexed: 12/18/2022]
Abstract
Receptor clustering on the cell membrane is critical in the signaling of many immunoreceptors, and this mechanism has previously been attributed to the extracellular and/or the intracellular interactions. Here, we report an unexpected finding that for death receptor 5 (DR5), a receptor in the tumor necrosis factor receptor superfamily, the transmembrane helix (TMH) alone in the receptor directly assembles a higher-order structure to drive signaling and that this structure is inhibited by the unliganded ectodomain. Nuclear magnetic resonance structure of the TMH in bicelles shows distinct trimerization and dimerization faces, allowing formation of dimer-trimer interaction networks. Single-TMH mutations that disrupt either trimerization or dimerization abolish ligand-induced receptor activation. Surprisingly, proteolytic removal of the DR5 ectodomain can fully activate downstream signaling in the absence of ligand. Our data suggest a receptor activation mechanism in which binding of ligand or antibodies to overcome the pre-ligand autoinhibition allows TMH clustering and thus signaling.
Collapse
Affiliation(s)
- Liqiang Pan
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute at Harvard Medical School, 250 Longwood Avenue, Boston, MA 02115, USA
| | - Tian-Min Fu
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute at Harvard Medical School, 250 Longwood Avenue, Boston, MA 02115, USA; Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA
| | - Wenbin Zhao
- Institute of Drug Metabolism and Pharmaceutical Analysis and Zhejiang Province Key Laboratory of Anti-cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, 310058 Hangzhou, China
| | - Linlin Zhao
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute at Harvard Medical School, 250 Longwood Avenue, Boston, MA 02115, USA
| | - Wen Chen
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute at Harvard Medical School, 250 Longwood Avenue, Boston, MA 02115, USA
| | - Chixiao Qiu
- Institute of Drug Metabolism and Pharmaceutical Analysis and Zhejiang Province Key Laboratory of Anti-cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, 310058 Hangzhou, China
| | - Wenhui Liu
- Institute of Drug Metabolism and Pharmaceutical Analysis and Zhejiang Province Key Laboratory of Anti-cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, 310058 Hangzhou, China
| | - Zhijun Liu
- National Facility for Protein Science in Shanghai, ZhangJiang Lab, Chinese Academy of Sciences, 201210 Shanghai, China
| | - Alessandro Piai
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute at Harvard Medical School, 250 Longwood Avenue, Boston, MA 02115, USA
| | - Qingshan Fu
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute at Harvard Medical School, 250 Longwood Avenue, Boston, MA 02115, USA
| | - Shuqing Chen
- Institute of Drug Metabolism and Pharmaceutical Analysis and Zhejiang Province Key Laboratory of Anti-cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, 310058 Hangzhou, China.
| | - Hao Wu
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute at Harvard Medical School, 250 Longwood Avenue, Boston, MA 02115, USA; Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA.
| | - James J Chou
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute at Harvard Medical School, 250 Longwood Avenue, Boston, MA 02115, USA.
| |
Collapse
|
92
|
da Silva PAV, Dos Anjos LMJ, Abduch TF, Pereira R, da Fonseca ADS, de Paoli F. Photobiomodulation can alter mRNA levels cell death-related. Lasers Med Sci 2019; 34:1373-1380. [DOI: 10.1007/s10103-019-02732-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Accepted: 01/21/2019] [Indexed: 11/27/2022]
|
93
|
Malsy M, Bitzinger D, Graf B, Bundscherer A. Staurosporine induces apoptosis in pancreatic carcinoma cells PaTu 8988t and Panc-1 via the intrinsic signaling pathway. Eur J Med Res 2019; 24:5. [PMID: 30686270 PMCID: PMC6348604 DOI: 10.1186/s40001-019-0365-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 01/18/2019] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Cancer, one of the leading causes of death worldwide, develops when the normal balance between mitosis and apoptosis is disrupted. The subsequently increased proliferation rate or decreased apoptosis rate of cells leads to uncontrolled cellular growth. Thus, the current aim of cancer research is to increase the apoptosis rate in tumor cells-while limiting the concurrent death of healthy cells-and to induce controlled apoptosis in abnormal cells. Staurosporine is a very potent inducer of apoptosis because it inhibits many different kinases. So far, many different kinase pathways of staurosporine-induced apoptosis have been discussed for various tumor entities. AIMS To identify the effect of staurosporine in pancreatic and colorectal carcinoma cells and its apoptosis-inducing signaling pathway. METHODS The apoptosis rate in pancreatic and colorectal carcinoma cells was analyzed by annexin V staining after staurosporine administration. Staurosporine stimulation and its effects on the expression of Bcl2, BAX, Bad, caspase-8, and caspase-9 were investigated with immunoblot. RESULTS Staurosporine significantly increased apoptosis in pancreatic carcinoma cells. Western blot analysis showed activation of caspase-9 in PaTu 8988t and Panc-1 cells with 1 µM staurosporine. In addition, expression of Bcl2 and Bad was decreased in PaTu 8988t cells. In colorectal carcinoma cells SW 480, staurosporine stimulation did not induce apoptosis. CONCLUSION Modern therapeutic strategies for tumor diseases target the efficient modulation of specific signaling and transcription pathways. In this respect, the therapeutic potential of protein kinase inhibitors has been repeatedly discussed. Our study showed that staurosporine induces apoptosis in pancreatic carcinoma cells via the intrinsic signaling pathway. Thus, staurosporine is a suitable positive control for in vitro apoptosis tests for the pancreatic cancer cell lines PaTu 8988t and Panc-1. Further clinical studies should analyze the impact of this finding on cancer treatment.
Collapse
Affiliation(s)
- Manuela Malsy
- Department of Anesthesiology, University Medical Center Regensburg, Franz Josef Strauss Allee 11, 93053 Regensburg, Germany
| | - Diane Bitzinger
- Department of Anesthesiology, University Medical Center Regensburg, Franz Josef Strauss Allee 11, 93053 Regensburg, Germany
| | - Bernhard Graf
- Department of Anesthesiology, University Medical Center Regensburg, Franz Josef Strauss Allee 11, 93053 Regensburg, Germany
| | - Anika Bundscherer
- Department of Anesthesiology, University Medical Center Regensburg, Franz Josef Strauss Allee 11, 93053 Regensburg, Germany
| |
Collapse
|
94
|
Qu D, Jiang M, Huang D, Zhang H, Feng L, Chen Y, Zhu X, Wang S, Han J. Synergistic Effects of The Enhancements to Mitochondrial ROS, p53 Activation and Apoptosis Generated by Aspartame and Potassium Sorbate in HepG2 Cells. Molecules 2019; 24:E457. [PMID: 30696035 PMCID: PMC6384600 DOI: 10.3390/molecules24030457] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 01/23/2019] [Accepted: 01/25/2019] [Indexed: 01/26/2023] Open
Abstract
The safety of food additives has been widely concerned. Using single additives in the provisions of scope is safe, but the combination of additives, may induce additive, synergy, antagonism and other joint effects. This study investigated the cytotoxicity of aspartame (AT) together with potassium sorbate (PS). Thiazolyl Blue Tetrazolium Bromide (MTT) assay indicated that AT and PS had IC50 values of 0.48 g/L and 1.25 g/L at 24 h, respectively. High content analysis (HCA) showed that both AT and PS had a negative effect on mitochondrial membrane potential (MMP), reactive oxygen species (ROS) and DNA damage while the joint group behaved more obviously. The biochemical assays revealed typical cell morphological changes and the activation of cytochrome c and caspase-3 verified apoptosis induced by AT together with PS. With dissipation of MMP and increase of cell membrane permeability (CMP), it indicated AT together with PS-induced apoptosis was mediated by mitochondrial pathway. Meanwhile, p53 were involved in DNA damage, and the ratio of Bax/Bcl-2 was increased. Moreover, excessive ROS induced by AT together with PS is a key initiating factor for apoptosis. All these results proved that p53 was involved in apoptosis via mitochondria-mediated pathway and the process was regulated by ROS.
Collapse
Affiliation(s)
- Daofeng Qu
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310035, China.
| | - Mengxue Jiang
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310035, China.
| | - Dongping Huang
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310035, China.
| | - Hui Zhang
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310035, China.
| | - Lifang Feng
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310035, China.
| | - Yuewen Chen
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310035, China.
| | - Xuan Zhu
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310035, China.
| | - Suhua Wang
- Wenzhou Entry-exit Inspection and Quarantine Bureau, Wenzhou 325027, China.
| | - Jianzhong Han
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310035, China.
| |
Collapse
|
95
|
Aliwaini S, Lubbad AM, Shourfa A, Hamada HAA, Ayesh B, Abu Tayem HEM, Abu Mustafa A, Abu Rouk F, Redwan MM, Al-Najjar M. Overexpression of TBX3 transcription factor as a potential diagnostic marker for breast cancer. Mol Clin Oncol 2019; 10:105-112. [PMID: 30655984 DOI: 10.3892/mco.2018.1761] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 10/26/2018] [Indexed: 11/06/2022] Open
Abstract
The T-box 3 (TBX3) transcription factor has been shown to serve multiple roles in normal development. Recent findings have revealed that TBX3 is overexpressed in different types of carcinomas, including breast, cervical, ovarian, melanoma, pancreatic, lung, liver, bladder, head and neck. Therefore, the present study investigated the significance of TBX3 as a diagnostic marker of breast cancer. To achieve this aim, breast cancer samples and their adjacent normal tissues were collected from 51 breast cancer patients from the European Gaza hospital during 2015-2016. Sections from each sample were immune-stained by anti-TBX3 and suitable secondary and tertiary antibodies. TBX3 levels were evaluated in cancerous and normal samples. Clinicopathological data for each patient were documented. The correlation between TBX3 levels and the clinicopathological parameters were statistically tested. The results revealed that TBX3 is significantly overexpressed in breast cancer tissues when compared with normal tissues. Furthermore, TBX3 was mainly a cytoplasmic protein in normal and breast cancer tissues. Notably, TBX3 levels exhibited a sensitivity of 78.4%, specificity of 79.6%, accuracy of 79% and area under the curve of 0.791 (0.700-0.882) at a cut-off value=9 as breast cancer marker. However, no significant associations were observed between TBX3 levels and other breast cancer markers including oestrogen receptor, progesterone receptor, human epidermal growth factor receptor 2, cancer antigen 15-3 and breast cancer stages. Altogether, these results suggested that TBX3 overexpression may be a potential biomarker for breast cancer.
Collapse
Affiliation(s)
- Saeb Aliwaini
- Department of Biological Sciences and Biotechnology, Faculty of Sciences, Islamic University of Gaza, 108 Gaza, Palestine
| | - Abdel Monem Lubbad
- Department of Pathology, Faculty of Medicine, Islamic University of Gaza, 108 Gaza, Palestine
| | - Ahmed Shourfa
- Department of Oncology, European Gaza Hospital, 7049 Gaza, Palestine
| | | | - Basim Ayesh
- Department of Laboratory Medical Sciences, Alaqsa University, 4051 Gaza, Palestine
| | - Husam Eddeen M Abu Tayem
- Department of Biological Sciences and Biotechnology, Faculty of Sciences, Islamic University of Gaza, 108 Gaza, Palestine
| | - Ayman Abu Mustafa
- Nursing Department, Palestine College of Nursing, 7049 Gaza, Palestine
| | - Fayek Abu Rouk
- Department of Oncology, European Gaza Hospital, 7049 Gaza, Palestine
| | - Moen M Redwan
- Department of Pathology, Alshefa Hospital, 1016 Gaza, Palestine
| | - Mohamed Al-Najjar
- Department of Oncology, European Gaza Hospital, 7049 Gaza, Palestine
| |
Collapse
|
96
|
Chen Y, Wu Y, Shi H, Wang J, Zheng Z, Chen J, Chen X, Zhang Z, Xu D, Wang X, Xiao J. Melatonin ameliorates intervertebral disc degeneration via the potential mechanisms of mitophagy induction and apoptosis inhibition. J Cell Mol Med 2019; 23:2136-2148. [PMID: 30609271 PMCID: PMC6378230 DOI: 10.1111/jcmm.14125] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 12/05/2018] [Accepted: 12/08/2018] [Indexed: 12/23/2022] Open
Abstract
Intervertebral disc degeneration (IDD) is a complicated disease in patients. The pathogenesis of IDD encompasses cellular oxidative stress, mitochondrion dysfunction and apoptosis. Melatonin eliminates oxygen free radicals, regulates mitochondrial homoeostasis and function, stimulates mitophagy and protects against cellular apoptosis. Therefore, we hypothesize that melatonin has beneficial effect on IDD by mitophagy stimulation and inhibition of apoptosis. The effects of melatonin on IDD were investigated in vitro and in vivo. For the former, melatonin diminished cellular apoptosis caused by tert‐butyl hydroperoxide in nucleus pulposus (NP) cells. Mitophagy, as well as its upstream regulator Parkin, was activated by melatonin in both a dose and time‐dependent manner. Mitophagy inhibition by cyclosporine A (CsA) partially eliminated the protective effects of melatonin against NP cell apoptosis, suggesting that mitophagy is involved in the protective effect of melatonin on IDD. In addition, melatonin was demonstrated to preserve the extracellular matrix (ECM) content of Collagen II, Aggrecan and Sox‐9, while inhibiting the expression of matrix degeneration enzymes, including MMP‐13 and ADAMTS‐5. In vivo, our results demonstrated that melatonin treatment ameliorated IDD in a puncture‐induced rat model. To conclude, our results suggested that melatonin protected NP cells against apoptosis via mitophagy induction and ameliorated disc degeneration, providing the potential therapy for IDD.
Collapse
Affiliation(s)
- Yu Chen
- Department of Orthopaedic Surgery, The Second Afliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,School of Pharmacy, Key Laboratory of Biotechnology and Pharmaceutical Engineering, Wenzhou Medical University, Wenzhou, China
| | - Yanqing Wu
- Institute of Life Sciences, Wenzhou University, Wenzhou, China
| | - Hongxue Shi
- School of Pharmacy, Key Laboratory of Biotechnology and Pharmaceutical Engineering, Wenzhou Medical University, Wenzhou, China
| | - Jianle Wang
- Department of Orthopaedic Surgery, The Second Afliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zengming Zheng
- Department of Orthopaedic Surgery, The Second Afliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jian Chen
- Department of Orthopaedic Surgery, The Second Afliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xibang Chen
- Department of Orthopaedic Surgery, The Second Afliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zengjie Zhang
- Department of Orthopaedic Surgery, The Second Afliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Daoliang Xu
- Department of Orthopaedic Surgery, The Second Afliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiangyang Wang
- Department of Orthopaedic Surgery, The Second Afliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jian Xiao
- Department of Orthopaedic Surgery, The Second Afliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,School of Pharmacy, Key Laboratory of Biotechnology and Pharmaceutical Engineering, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
97
|
Mo Y, Fan Y, Fu W, Xu W, Chen S, Wen Y, Liu S, Peng L, Xiao Y. Acute immune stress improves cell resistance to chemical poison damage in SP600125-induced polyploidy of fish cells in vitro. FISH & SHELLFISH IMMUNOLOGY 2019; 84:656-663. [PMID: 30393156 DOI: 10.1016/j.fsi.2018.10.063] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Revised: 10/15/2018] [Accepted: 10/24/2018] [Indexed: 06/08/2023]
Abstract
Previous research has indicated that the small compound, SP600125, could induce polyploidy of fish cells, and has established a stable tetraploid cell line from diploid fish cells. In order to explore how fish cells maintain homeostasis under SP600125-stress in vitro, this study investigates impacts of SP600125-stress on intracellular pathways, as well as on regulation of the cellular homeostasis feedback in fish cells. Transcriptomes are obtained from the SP600125-treated cells. Compared with unigenes expressed in control group (crucial carp fin cells), a total of 2670 and 1846 unigenes are significantly upregulated and downregulated in these cells, respectively. Differentially expressed genes are found, which are involved in innate defense, inflammatory pathways and cell adhesion molecules-related pathways. The SP600125-stress enhances cell-mediated immunity, characterized by significantly increasing expression of multiple immune genes. These enhanced immune genes include the pro-inflammatory cytokines (IL-1β, TNF-ɑ, IL-6R), the adaptor signal transducers (STAT, IκBɑ), and the integrins (ɑ2β1, ɑMβ2). Furthermore, mitochondria are contributed to the cellular homeostasis regulation upon the SP600125-stress. The results show that acute inflammation is an adaptive and controlled response to the SP600125-stress, which is beneficial for alleviating toxicity by SP600125. They provide a potential way of breeding fish polyploidy induced by SP600125 in the future research.
Collapse
Affiliation(s)
- Yanxiu Mo
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, Hunan, 410081, PR China; School of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, PR China; School of Basic Medical Science, Xiangnan University, Chenzhou, Hunan, 423000, PR China
| | - Yunpeng Fan
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, Hunan, 410081, PR China; School of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, PR China
| | - Wen Fu
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, Hunan, 410081, PR China; School of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, PR China
| | - Wenting Xu
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, Hunan, 410081, PR China; School of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, PR China
| | - Shujuan Chen
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, Hunan, 410081, PR China; School of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, PR China
| | - Yuanhui Wen
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, Hunan, 410081, PR China; School of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, PR China
| | - Shaojun Liu
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, Hunan, 410081, PR China; School of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, PR China
| | - Liangyue Peng
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, Hunan, 410081, PR China; School of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, PR China.
| | - Yamei Xiao
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, Hunan, 410081, PR China; School of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, PR China.
| |
Collapse
|
98
|
Nerguizian V, Stiharu I, Al-Azzam N, Yassine-Diab B, Alazzam A. The effect of dielectrophoresis on living cells: crossover frequencies and deregulation in gene expression. Analyst 2019; 144:3853-3860. [DOI: 10.1039/c9an00320g] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
This article documents the effect of dielectrophoresis on living cells.
Collapse
Affiliation(s)
- Vahé Nerguizian
- Department of Electrical Engineering
- École de technologie supérieure (ÉTS)
- Montreal
- Canada
| | - Ion Stiharu
- Department of Mechanical and Industrial Engineering
- Concordia University
- Montreal
- Canada
| | - Nosayba Al-Azzam
- Department of Physiology and Biochemistry
- Jordan University of Science and Technology
- Irbid
- Jordan
| | | | - Anas Alazzam
- Department of Mechanical Engineering
- Khalifa University
- Abu Dhabi
- United Arab Emirates
- Department of Electrical Engineering
| |
Collapse
|
99
|
Wang Y, Wang C, Liu N, Hou J, Xiao W, Wang H. HOXC6 promotes cervical cancer progression via regulation of Bcl-2. FASEB J 2018; 33:3901-3911. [PMID: 30509141 DOI: 10.1096/fj.201801099rr] [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] [Indexed: 12/12/2022]
Abstract
Homeobox C6 ( HOXC6) is a transcription factor that plays an important role in the development of several cancers. However, it is unknown whether HOXC6 regulates cervical cancer progression. In this study, we used quantitative PCR and Western blots to demonstrate that HOXC6 overexpression is associated with cervical cancer progression. These results are also supported by the public RNA-seq data showing that HOXC6 overexpression is correlated with poor patient survival. In vitro and in vivo experiments revealed that enhanced HOXC6 expression results in cervical cancer cell proliferation, cell cycle progression, anchorage colony formation, and xenograft tumor growth. Mechanistically, we show that HOXC6 up-regulates BCL2 expression through transcription regulation. Together, these results led us to conclude that overexpression of HOXC6 expression can enhance BCL2-mediated antiapoptotic effects to promote cervical cancer cell cycle progression and proliferation.-Wang, Y., Wang, C., Liu, N., Hou, J., Xiao, W., Wang, H. HOXC6 promotes cervical cancer progression via regulation of Bcl-2.
Collapse
Affiliation(s)
- Yongsheng Wang
- Department of Obstetrics and Gynecology, People's Hospital of Linyi City, Linyi City, China; and
| | - Cong Wang
- Department of Obstetrics and Gynecology, People's Hospital of Linyi City, Linyi City, China; and
| | - Na Liu
- Department of Obstetrics and Gynecology, People's Hospital of Linyi City, Linyi City, China; and
| | - Jun Hou
- Department of Pharmacy, Chengdu Military General Hospital, Chengdu, China
| | - Wenjing Xiao
- Department of Pharmacy, Chengdu Military General Hospital, Chengdu, China
| | - Hui Wang
- Department of Obstetrics and Gynecology, People's Hospital of Linyi City, Linyi City, China; and
| |
Collapse
|
100
|
Qu C, Yang W, Xu Q, Sun J, Lu M, Wang Y, Liu C, Wang W, Wang L, Song L. A novel effector caspase (Caspase-3/7-1) involved in the regulation of immune homeostasis in Chinese mitten crab Eriocheir sinensis. FISH & SHELLFISH IMMUNOLOGY 2018; 83:76-83. [PMID: 30195917 DOI: 10.1016/j.fsi.2018.09.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 08/26/2018] [Accepted: 09/05/2018] [Indexed: 06/08/2023]
Abstract
Caspases are a conserved family of cysteine proteases characterized by specificity for aspartic acid and play an essential role in cell apoptosis. In the present study, a novel effector caspase (designated as EsCaspase-3/7-1) was identified from Chinese mitten crab Eriocheir sinensis. The open reading frame of EsCaspase-3/7-1 cDNA was of 972 bp, encoding a polypeptide of 323 amino acids. EsCaspase-3/7-1 contained an N-terminal prodomain and a conservative C-terminal CASc domain, with the conserved active site "QACRG". The mRNA transcripts of EsCaspase-3/7-1 were constitutively expressed in all the examined tissues with high expression level in hemocytes, hepatopancreas and gill. The EsCaspase-3/7-1 protein was mainly distributed in the cytoplasm of hemocytes. After Aeromonas hydrophila and lipopolysaccharide (LPS) stimulations, the mRNA expression level of EsCaspase-3/7-1 in hemocytes increased significantly. The mRNA expression level of EsCaspase-3/7-1 in hemocytes was significantly up-regulated after H2O2 treatment in vitro. The recombinant EsCaspase-3/7-1 protein (rEsCaspase-3/7-1) was capable of hydrolyzing the substrate Ac-DEVD-pNA rather than Ac-YVAD-pNA and Ac-VEID-pNA in vitro, and exhibited binding activity to LPS. These results demonstrated that EsCaspase-3/7-1 might act as an LPS receptor, and play an important role in the regulation of immune homeostasis of E. sinensis.
Collapse
Affiliation(s)
- Chen Qu
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology & Disease Control, Dalian Ocean University, Dalian, 116023, China
| | - Wen Yang
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China
| | - Qingsong Xu
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China
| | - Jiejie Sun
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China
| | - Mengmeng Lu
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China
| | - Ying Wang
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China
| | - Chao Liu
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China
| | - Weilin Wang
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China
| | - Lingling Wang
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Laboratory of Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China
| | - Linsheng Song
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Laboratory of Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China; Liaoning Key Laboratory of Marine Animal Immunology & Disease Control, Dalian Ocean University, Dalian, 116023, China.
| |
Collapse
|