1
|
He Y, Du G, Wang G, Guan H, Zhu S, Chen B, He X, Zhu Y. Autophagy is involved in the toxicity of the biocontrol agent GC16 against Tetranychus pueraricola (Acari: Tetranychidae) based on transcriptomic and proteomic analyses. BMC Genomics 2025; 26:119. [PMID: 39920625 PMCID: PMC11806590 DOI: 10.1186/s12864-025-11312-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 01/30/2025] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND GC16 is a novel pesticide with acaricidal properties against the spider mite Tetranychus pueraricola (Ehara & Gotoh). Its physiological mechanisms have been described previously, but its molecular mechanisms of action remain unclear. Thus, we aimed to explore the acaricidal mechanisms of GC16 through transcriptomic and proteomic analyses. The results were verified using transmission electron microscopy (TEM), immunofluorescence assay, and western blotting. RESULTS Transcriptomic and proteomic analyses revealed 2717 differentially expressed genes (DEGs) and 374 differentially expressed proteins (DEPs) between the GC16-treated and control mites. Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis indicated that the DEGs and DEPs were enriched in the autophagy pathway. TEM showed that the number of autophagosomes and autolysosomes was higher in the GC16-treated mites than in the control mites. Immunofluorescence assay and western blot results consistently indicated that GC16 treatment significantly enhanced the relative expression of the autophagy protein LC3 in insect Sf9 cells. The intracellular calcium concentration in the GC16-treated Sf9 cells was 2.30 times higher than that in the control cells, suggesting that GC16 disrupted calcium homeostasis and potentially acted as a calcium-driven nerve agent. CONCLUSIONS Autophagy is involved in the toxicity of GC16 against T. pueraricola and may be activated by elevated Ca2+ levels. This study reveals the molecular insecticidal mechanisms of GC16 and provides rationale for the field application of GC16 to control pest mites.
Collapse
Affiliation(s)
- Yanyan He
- School of Agriculture, Yunnan University, Kunming, 650500, China
- State Key Laboratory of Conservation and Utilization of Biological Resources of Yunnan, College of Plant Protection, Yunnan Agricultural University, Kunming, 650201, China
- Yunnan Provincial Center for Disease Control and Prevention, 650034, Kunming, China
| | - Guangzu Du
- State Key Laboratory of Conservation and Utilization of Biological Resources of Yunnan, College of Plant Protection, Yunnan Agricultural University, Kunming, 650201, China
| | - Guang Wang
- State Key Laboratory of Conservation and Utilization of Biological Resources of Yunnan, College of Plant Protection, Yunnan Agricultural University, Kunming, 650201, China
| | - Huiming Guan
- Inner Mongolia Wulanchabu Science and Technology Development Center, Wulanchabu, 012000, China
| | - Shusheng Zhu
- State Key Laboratory of Conservation and Utilization of Biological Resources of Yunnan, College of Plant Protection, Yunnan Agricultural University, Kunming, 650201, China
| | - Bin Chen
- State Key Laboratory of Conservation and Utilization of Biological Resources of Yunnan, College of Plant Protection, Yunnan Agricultural University, Kunming, 650201, China.
| | - Xiahong He
- Southwest Forestry University, Kunming, 650224, China.
| | - Youyong Zhu
- School of Agriculture, Yunnan University, Kunming, 650500, China.
- State Key Laboratory of Conservation and Utilization of Biological Resources of Yunnan, College of Plant Protection, Yunnan Agricultural University, Kunming, 650201, China.
| |
Collapse
|
2
|
de Andrade Borges A, Ouverney G, Arruda ATS, Ribeiro AV, Ribeiro RCB, de Souza AS, da Fonseca ACC, de Queiroz LN, de Almeida ECP, Pontes B, Rabelo VWH, Ferreira V, Abreu PA, de Carvalho da Silva F, da Silva Magalhaes Forezi L, Robbs BK. Determination of Inhibitory Effect of PKM2 Enzyme and Antitumoral Activity of Novel Coumarin-naphthoquinone Hybrids. Curr Med Chem 2025; 32:359-379. [PMID: 38877863 DOI: 10.2174/0109298673298471240605072658] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 04/08/2024] [Accepted: 05/06/2024] [Indexed: 02/19/2025]
Abstract
BACKGROUND Oral squamous cell carcinoma (OSCC) represents the primary form of oral cancer, posing a significant global health threat. The existing chemotherapy options are accompanied by notable side effects impacting patient treatment adherence. Consequently, the exploration and development of novel substances with enhanced anticancer effects and fewer side effects have become pivotal in the realms of biological and chemical science. OBJECTIVE This work presents the pioneering examples of naphthoquinone-coumarin hybrids as a new category of highly effective cytotoxic substances targeting oral squamous cell carcinoma (OSCC). METHODS Given the significance of both naphthoquinones and coumarins as essential pharmacophores/ privileged structures in the quest for anticancer compounds, this study focused on the synthesis and evaluation of novel naphthoquinones/coumarin hybrids against oral squamous cell carcinoma. RESULTS By several in vitro, in silico, and in vivo approaches, we demonstrated that compound 6e was highly cytotoxic against OSCC cells and several other cancer cell types and was more selective than current chemotherapeutic drugs (carboplatin) and the naphthoquinone lapachol. Furthermore, compound 6e was non-hemolytic and tolerated in vivo at 50 mg/kg with an LD50 of 62.5 mg/kg. Furthermore, compound 6e did not induce apoptosis and cell cycle arrest but led to intracellular vesicle formation with LC3 aggregation in autophagosomes, suggesting an autophagic cell death. Additionally, 6e had a high-affinity potential for PKM2 protein, higher than the known ligands, such as lapachol or shikonin, and was able to inhibit this enzyme activity in vitro. CONCLUSION We assert that compound 6e shows promise as a potential lead for a novel chemotherapeutic drug targeting OSCC, with potential applicability to other cancer types.
Collapse
Grants
- 1A 301873/2019-4, 301873/2019-4 CNPq, Conselho Nacional de Desenvolvimento Científico e Tecnológico
- E-26/010.101106/2018, E-26/202, 787/2019, E-26/10.002250/2019, E-26/210.085/2022, E-26/010.001318/2019, E-26/211.343/2021, E-26/210.068/2021, E-26/203.191/2017-JCNE, E-26 /202.800/2017-CNE, E-26/010.101106/2018, E-26/200 .870/2021-CNE, E-26/201.369/2021-JCNE, E-26/010/ 001687/2015, E-26/202.787/2019, E-26/210.514/2019, E-26/10.002250/2019, E-26/211.343/2021, E-26/210. 085/2022, E-26/210.068/2021 FAPERJ, Fundação de Amparo à Pesquisa do Estado do Rio de Janeiro
- 001 Coordination for the Improvement of Higher Education Personnel - Brazil (CAPES)
Collapse
Affiliation(s)
- Amanda de Andrade Borges
- Departamento de Química Orgânica, Instituto de Química, Universidade Federal Fluminense, CEP, Niterói, 24020-141, RJ, Brazil
| | - Gabriel Ouverney
- Programa de Pós-graduação em Ciências Aplicadas a Produtos para Saúde, Faculdade de Farmácia, Universidade Federal Fluminense, CEP, Niterói, 24020-141, RJ, Brazil
| | - Afonso Thales Sousa Arruda
- Departamento de Ciência Básica, Universidade Federal Fluminense, Campus Universitário de Nova Friburgo, CEP, Nova Friburgo, 28625-650, RJ, Brazil
| | - Amanda Vieira Ribeiro
- Departamento de Ciência Básica, Universidade Federal Fluminense, Campus Universitário de Nova Friburgo, CEP, Nova Friburgo, 28625-650, RJ, Brazil
| | - Ruan Carlos Busquet Ribeiro
- Departamento de Química Orgânica, Instituto de Química, Universidade Federal Fluminense, CEP, Niterói, 24020-141, RJ, Brazil
| | - Acacio Silva de Souza
- Departamento de Tecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal Fluminense, CEP, Niterói, 24020-141, RJ, Brazil
| | - Anna Carolina Carvalho da Fonseca
- Programa de Pós-graduação em Odontologia, Instituto de Saúde de Nova Friburgo, Universidade Federal Fluminense, CEP, Nova Friburgo, 28625-650, RJ, Brazil
| | - Lucas Nicolau de Queiroz
- Programa de Pós-graduação em Ciências Aplicadas a Produtos para Saúde, Faculdade de Farmácia, Universidade Federal Fluminense, CEP, Niterói, 24020-141, RJ, Brazil
| | - Elan Cardozo Paes de Almeida
- Departamento de Ciência Básica, Universidade Federal Fluminense, Campus Universitário de Nova Friburgo, CEP, Nova Friburgo, 28625-650, RJ, Brazil
| | - Bruno Pontes
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, CEP, Rio de Janeiro, 21941-902, RJ, Brazil
| | - Vitor Won-Held Rabelo
- Instituto de Biodiversidade e Sustentabilidade, Universidade Federal do Rio de Janeiro, CEP , Macaé, 27965-045, RJ, Brazil
| | - Vitor Ferreira
- Departamento de Tecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal Fluminense, CEP, Niterói, 24020-141, RJ, Brazil
| | - Paula Alvarez Abreu
- Instituto de Biodiversidade e Sustentabilidade, Universidade Federal do Rio de Janeiro, CEP , Macaé, 27965-045, RJ, Brazil
| | - Fernando de Carvalho da Silva
- Departamento de Química Orgânica, Instituto de Química, Universidade Federal Fluminense, CEP, Niterói, 24020-141, RJ, Brazil
| | - Luana da Silva Magalhaes Forezi
- Departamento de Química Orgânica, Instituto de Química, Universidade Federal Fluminense, CEP, Niterói, 24020-141, RJ, Brazil
| | - Bruno Kaufmann Robbs
- Departamento de Ciência Básica, Universidade Federal Fluminense, Campus Universitário de Nova Friburgo, CEP, Nova Friburgo, 28625-650, RJ, Brazil
| |
Collapse
|
3
|
Qi K, Li J, Hu Y, Qiao Y, Mu Y. Research progress in mechanism of anticancer action of shikonin targeting reactive oxygen species. Front Pharmacol 2024; 15:1416781. [PMID: 39076592 PMCID: PMC11284502 DOI: 10.3389/fphar.2024.1416781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 06/13/2024] [Indexed: 07/31/2024] Open
Abstract
Excessive buildup of highly reactive molecules can occur due to the generation and dysregulation of reactive oxygen species (ROS) and their associated signaling pathways. ROS have a dual function in cancer development, either leading to DNA mutations that promote the growth and dissemination of cancer cells, or triggering the death of cancer cells. Cancer cells strategically balance their fate by modulating ROS levels, activating pro-cancer signaling pathways, and suppressing antioxidant defenses. Consequently, targeting ROS has emerged as a promising strategy in cancer therapy. Shikonin and its derivatives, along with related drug carriers, can impact several signaling pathways by targeting components involved with oxidative stress to induce processes such as apoptosis, necroptosis, cell cycle arrest, autophagy, as well as modulation of ferroptosis. Moreover, they can increase the responsiveness of drug-resistant cells to chemotherapy drugs, based on the specific characteristics of ROS, as well as the kind and stage of cancer. This research explores the pro-cancer and anti-cancer impacts of ROS, summarize the mechanisms and research achievements of shikonin-targeted ROS in anti-cancer effects and provide suggestions for designing further anti-tumor experiments and undertaking further experimental and practical research.
Collapse
Affiliation(s)
- Ke Qi
- Department of Diagnostic Clinical Laboratory Science, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Jiayi Li
- Department of Clinical Test Center, Medical Laboratory, Peking University Cancer Hospital (Inner Mongolia Campus), Affiliated Cancer Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Yang Hu
- Department of Diagnostic Clinical Laboratory Science, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Yiyun Qiao
- Department of Clinical Test Center, Peking University Cancer Hospital (Inner Mongolia Campus), Affiliated Cancer Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Yongping Mu
- Department of Clinical Test Center, Peking University Cancer Hospital (Inner Mongolia Campus), Affiliated Cancer Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| |
Collapse
|
4
|
Borges AA, de Souza MP, da Fonseca ACC, Wermelinger GF, Ribeiro RCB, Amaral AAP, de Carvalho CJC, Abreu LS, de Queiroz LN, de Almeida ECP, Rabelo VW, Abreu PA, Pontes B, Ferreira VF, da Silva FDC, Forezi LDSM, Robbs BK. Chemoselective Synthesis of Mannich Adducts from 1,4-Naphthoquinones and Profile as Autophagic Inducers in Oral Squamous Cell Carcinoma. Molecules 2022; 28:molecules28010309. [PMID: 36615502 PMCID: PMC9822194 DOI: 10.3390/molecules28010309] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 12/23/2022] [Accepted: 12/24/2022] [Indexed: 01/03/2023] Open
Abstract
Oral squamous cell carcinoma (OSCC) is a worldwide public health problem, accounting for approximately 90% of all oral cancers, and is the eighth most common cancer in men. Cisplatin and carboplatin are the main chemotherapy drugs used in the clinic. However, in addition to their serious side effects, such as damage to the nervous system and kidneys, there is also drug resistance. Thus, the development of new drugs becomes of great importance. Naphthoquinones have been described with antitumor activity. Some of them are found in nature, but semi synthesis has been used as strategy to find new chemical entities for the treatment of cancer. In the present study, we promote a multiple component reaction (MCR) among lawsone, arylaldehydes, and benzylamine to produce sixteen chemoselectively derivated Mannich adducts of 1,4-naphthoquinones in good yield (up to 97%). The antitumor activities and molecular mechanisms of action of these compounds were investigated in OSCC models and the compound 6a induced cytotoxicity in three different tumor cell lines (OSCC4, OSCC9, and OSCC25) and was more selective (IS > 2) for tumor cells than the chemotropic drug carboplatin and the controls lapachol and shikonin, which are chemically similar compounds with cytotoxic effects. The 6a selectively and significantly reduced the amount of cell colony growth, was not hemolytic, and tolerable in mice with no serious side effects at a concentration of 100 mg/kg with a LD50 of 150 mg/kg. The new compound is biologically stable with a profile similar to carboplatin. Morphologically, 6a does not induce cell retraction or membrane blebs, but it does induce intense vesicle formation and late emergence of membrane bubbles. Exploring the mechanism of cell death induction, compound 6a does not induce ROS formation, and cell viability was not affected by inhibitors of apoptosis (ZVAD) and necroptosis (necrostatin 1). Autophagy followed by a late apoptosis process appears to be the death-inducing pathway of 6a, as observed by increased viability by the autophagy inhibitor (3-MA) and by the appearance of autophagosomes, later triggering a process of late apoptosis with the presence of caspase 3/7 and DNA fragmentation. Molecular modeling suggests the ability of the compound to bind to topoisomerase I and II and with greater affinity to hPKM2 enzyme than controls, which could explain the mechanism of cell death by autophagy. Finally, the in-silico prediction of drug-relevant properties showed that compound 6a has a good pharmacokinetic profile when compared to carboplatin and doxorubicin. Among the sixteen naphthoquinones tested, compound 6a was the most effective and is highly selective and well tolerated in animals. The induction of cell death in OSCC through autophagy followed by late apoptosis possibly via inhibition of the PKM2 enzyme points to a promising potential of 6a as a new preclinical anticancer candidate.
Collapse
Affiliation(s)
- Amanda A. Borges
- Departamento de Química Orgânica, Instituto de Química, Campus do Valonguinho, Universidade Federal Fluminense, Niterói CEP 24020-150, Brazil
| | - Michele P. de Souza
- Programa de Pós-Graduação em Ciências Aplicadas a Produtos para Saúde, Faculdade de Farmácia, Universidade Federal Fluminense, Niterói CEP 24241-000, Brazil
| | - Anna Carolina C. da Fonseca
- Programa de Pós-Graduação em Odontologia, Instituto de Saúde de Nova Friburgo, Universidade Federal Fluminense, Nova Friburgo CEP 28625-650, Brazil
| | - Guilherme F. Wermelinger
- Departamento de Ciência Básica, Campus Universitário de Nova Friburgo, Universidade Federal Fluminense, Nova Friburgo CEP 28625-650, Brazil
| | - Ruan C. B. Ribeiro
- Departamento de Química Orgânica, Instituto de Química, Campus do Valonguinho, Universidade Federal Fluminense, Niterói CEP 24020-150, Brazil
| | - Adriane A. P. Amaral
- Departamento de Química Orgânica, Instituto de Química, Campus do Valonguinho, Universidade Federal Fluminense, Niterói CEP 24020-150, Brazil
| | - Cláudio José C. de Carvalho
- Departamento de Química Orgânica, Instituto de Química, Campus do Valonguinho, Universidade Federal Fluminense, Niterói CEP 24020-150, Brazil
| | - Lucas S. Abreu
- Departamento de Química Orgânica, Instituto de Química, Campus do Valonguinho, Universidade Federal Fluminense, Niterói CEP 24020-150, Brazil
| | - Lucas Nicolau de Queiroz
- Programa de Pós-Graduação em Ciências Aplicadas a Produtos para Saúde, Faculdade de Farmácia, Universidade Federal Fluminense, Niterói CEP 24241-000, Brazil
| | - Elan C. P. de Almeida
- Departamento de Ciência Básica, Campus Universitário de Nova Friburgo, Universidade Federal Fluminense, Nova Friburgo CEP 28625-650, Brazil
| | - Vitor W. Rabelo
- Instituto de Biodiversidade e Sustentabilidade, Campus Macaé, Universidade Federal do Rio de Janeiro, Macaé CEP 27965-045, Brazil
| | - Paula A. Abreu
- Instituto de Biodiversidade e Sustentabilidade, Campus Macaé, Universidade Federal do Rio de Janeiro, Macaé CEP 27965-045, Brazil
| | - Bruno Pontes
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro CEP 21941-902, Brazil
| | - Vitor F. Ferreira
- Departamento de Tecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal Fluminense, Niterói CEP 24241-000, Brazil
| | - Fernando de C. da Silva
- Departamento de Química Orgânica, Instituto de Química, Campus do Valonguinho, Universidade Federal Fluminense, Niterói CEP 24020-150, Brazil
| | - Luana da S. M. Forezi
- Departamento de Química Orgânica, Instituto de Química, Campus do Valonguinho, Universidade Federal Fluminense, Niterói CEP 24020-150, Brazil
- Correspondence: (L.d.S.M.F.); (B.K.R.)
| | - Bruno K. Robbs
- Departamento de Química Orgânica, Instituto de Química, Campus do Valonguinho, Universidade Federal Fluminense, Niterói CEP 24020-150, Brazil
- Correspondence: (L.d.S.M.F.); (B.K.R.)
| |
Collapse
|
5
|
Rahman MM, Islam MR, Akash S, Shohag S, Ahmed L, Supti FA, Rauf A, Aljohani ASM, Al Abdulmonem W, Khalil AA, Sharma R, Thiruvengadam M. Naphthoquinones and derivatives as potential anticancer agents: An updated review. Chem Biol Interact 2022; 368:110198. [PMID: 36179774 DOI: 10.1016/j.cbi.2022.110198] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/01/2022] [Accepted: 09/12/2022] [Indexed: 11/03/2022]
Abstract
One of the leading global causes of death is cancer; even though several treatment methods have improved survival rates, the incidence and fatality rates remain high. Naphthoquinones are a type of quinone that is found in nature and has vital biological roles. These chemicals have anticancer (antineoplastic), analgesic, anti-inflammatory, antimalarial, antifungal, antiviral, antitrypanosomal, antischistosomal, leishmanicidal, and anti-ulcerative effects. Direct addition of a substituent group to the 1,4-naphthoquinone ring can alter the naphthoquinone's oxidation/reduction and acid/base characteristics, and the activity can be altered. Because of their pharmacological properties, such as anticancer activity and probable therapeutic application, naphthoquinones have greatly interested the scientific community. Some chemicals having a quinone ring in malignant cells have been found to have antiproliferative effects. Naphthoquinones' deadly impact is connected with the inhibition of electron transporters, the uncoupling of oxidative phosphorylation, the creation of ROS, and the formation of protein adducts, notably with -SH enzyme groups. This review article aims to discuss naphthoquinones and their derivatives, which act against cancer and their future perspectives. This review covers several studies highlighting the potent anticancer properties of naphthoquinones. Further, various proposed mechanisms of anticancer actions of naphthoquinones have been summarized in this review.
Collapse
Affiliation(s)
- Md Mominur Rahman
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, 1207, Dhaka, Bangladesh
| | - Md Rezaul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, 1207, Dhaka, Bangladesh
| | - Shopnil Akash
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, 1207, Dhaka, Bangladesh
| | - Sheikh Shohag
- Department of Genetic Engineering and Biotechnology, Faculty of Earth and Ocean Science, Bangabandhu Sheikh Mujibur Rahman Maritime University, Mirpur 12, Dhaka, 1216, Bangladesh
| | - Limon Ahmed
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, 1207, Dhaka, Bangladesh
| | - Fatema Akter Supti
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, 1207, Dhaka, Bangladesh
| | - Abdur Rauf
- Department of Chemistry, University of Swabi, Anbar 23561, Khyber Pakhtunkhwa, Pakistan.
| | - Abdullah S M Aljohani
- Department of Veterinary Medicine, College of Agriculture and Veterinary Medicine, Qassim University Buraydah, 52571, Saudi Arabia
| | - Waleed Al Abdulmonem
- Department of Pathology, College of Medicine Qassim University, Buraydah, Saudi Arabia
| | - Anees Ahmed Khalil
- University Institute of Diet and Nutritional Sciences, Faculty of Allied Health Sciences, The University of Lahore-Pakistan, Pakistan
| | - Rohit Sharma
- Department of Rasa Shastra & Bhaishajya Kalpana, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India
| | - Muthu Thiruvengadam
- Department of Crop Science, College of Sanghuh Life Sciences, Konkuk University, Seoul 05029, South Korea; Department of Microbiology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai 600077, India.
| |
Collapse
|
6
|
Cao Z, Guo C, Chen G, Liu J, Ni H, Liu F, Xiong G, Liao X, Lu H. Shikonin Inhibits Fin Regeneration in Zebrafish Larvae. Cells 2022; 11:cells11203187. [PMID: 36291055 PMCID: PMC9601185 DOI: 10.3390/cells11203187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/15/2022] [Accepted: 09/28/2022] [Indexed: 11/23/2022] Open
Abstract
Shikonin is a naphthoquinone compound extracted from Chinese comfrey for treating cancer. However, there are few reports on its research on vertebrate tissue regeneration. Zebrafish is an ideal model for studying organ regeneration. In this study, we found that 3-dpf of zebrafish larvae exposed to shikonin at concentrations of 0.2, 0.3, and 0.4 mg/L showed increasingly inhibited regeneration of the tail fin. Immunohistochemical staining showed that shikonin exposure from 6 to 12 hpa increased the number of apoptotic cells in the caudal fin wound of larvae and decreased the number of proliferating cells. Shikonin exposure was found to up-regulate oxidative stress, increase ROS levels, and reduce neutrophil recruitment in the early stage of wound repair. Moreover, shikonin exposure caused disordered expression of fin regeneration blastemal-related genes. The use of astaxanthin to down-regulate oxidative stress was found to significantly reduce the inhibition of caudal fin regeneration. Mixed exposure of AMPK inhibitors or fullerenes (C60) with shikonin also showed the similar rescue effect. Collectively, our study showed that shikonin inhibited fin regeneration in zebrafish larvae by the upregulation of oxidative stress level and AMPK signaling pathway. This research provides valuable information on the mechanism of action of shikonin for its safe application.
Collapse
Affiliation(s)
- Zigang Cao
- Correspondence: (Z.C.); (H.L.); Tel./Fax: +86-796-8116182 (Z.C.)
| | | | | | | | | | | | | | | | - Huiqiang Lu
- Correspondence: (Z.C.); (H.L.); Tel./Fax: +86-796-8116182 (Z.C.)
| |
Collapse
|
7
|
Li Y, Li Y, Zhang J, Ji L, Li M, Sun X, Feng H, Yu Z, Gao Y. Current Perspective of Traditional Chinese Medicines and Active Ingredients in the Therapy of Hepatocellular Carcinoma. J Hepatocell Carcinoma 2022; 9:41-56. [PMID: 35178363 PMCID: PMC8843800 DOI: 10.2147/jhc.s346047] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Accepted: 01/27/2022] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the leading lethal tumors worldwide, and the treatment remains a great medical challenge. Surgery and chemotherapy are current standard curative methods for patients with HCC, but the prognosis is still dismal. Based on unique medical theories and rich practical experience, traditional Chinese medicine (TCM) has been broadly employed to effectively treat HCC for a long history. Recently, systematic clinical trials have been well designed to study the efficacy of TCMs in the treatment of HCC, and the underlying antitumor mechanisms were also deeply explored. Here, we reviewed the published clinical evaluation of some commonly used TCMs in the treatment of HCC, and the related anti-HCC mechanisms through in vitro and in vivo study, promoting the modernization of TCM study in oncology for achieving a substantial reduction of HCC burden in the future.
Collapse
Affiliation(s)
- Yuyao Li
- Institute of Infectious Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Yue Li
- Institute of Infectious Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Jinghao Zhang
- Department of Liver Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Longshan Ji
- Laboratory of Cellular Immunity, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Man Li
- Laboratory of Cellular Immunity, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Xuehua Sun
- Department of Liver Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Hai Feng
- Institute of Infectious Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Zhuo Yu
- Department of Liver Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Correspondence: Zhuo Yu; Yueqiu Gao, Department of Liver Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, No. 528 Zhangheng Road, Pudong New District, Shanghai, 201203, People’s Republic of China, Tel +86 21 2025 6507, Fax +86 21 20256699, Email ;
| | - Yueqiu Gao
- Institute of Infectious Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Department of Liver Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| |
Collapse
|
8
|
Kaur K, Singh A, Sharma H, Punj S, Bedi N. Formulation Strategies and Therapeutic Applications of Shikonin and Related Derivatives. RECENT ADVANCES IN DRUG DELIVERY AND FORMULATION 2022; 16:55-67. [PMID: 35236278 DOI: 10.2174/2667387816666220302112201] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 01/11/2022] [Accepted: 01/11/2022] [Indexed: 06/14/2023]
Abstract
Shikonin and its derivatives are excellent representatives of biologically active naphthoquinones. A wide range of investigations carried out in the last few decades validated their pharmacological efficacy. Besides having magnificent therapeutic potential, shikonin and its derivatives suffer from various pharmacokinetic, toxicity, and stability issues like poor bioavailability, nephrotoxicity, photodegradation, etc. Recently, various research groups have developed an extensive range of formulations to tackle these issues to ease their path to clinical practice. The latest formulation approaches have been focused on exploiting the unique features of novel functional excipients, which in turn escalate the therapeutic effect of shikonin. Moreover, the codelivery approach in various drug delivery systems has been taken into consideration in a recent while to reduce toxicity associated with shikonin and its derivatives. This review sheds light on the essential reports and patents published related to the array of formulations containing shikonin and its derivatives.
Collapse
Affiliation(s)
- Kirandeep Kaur
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, 143005, India
| | - Atamjit Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, 143005, India
| | - Hamayal Sharma
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Sanha Punj
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, 143005, India
| | - Neena Bedi
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, 143005, India
| |
Collapse
|
9
|
Sun Q, Gong T, Liu M, Ren S, Yang H, Zeng S, Zhao H, Chen L, Ming T, Meng X, Xu H. Shikonin, a naphthalene ingredient: Therapeutic actions, pharmacokinetics, toxicology, clinical trials and pharmaceutical researches. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 94:153805. [PMID: 34749177 DOI: 10.1016/j.phymed.2021.153805] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 09/15/2021] [Accepted: 10/17/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Shikonin is one of the major phytochemical components of Lithospermum erythrorhizon (Purple Cromwell), which is a type of medicinal herb broadly utilized in traditional Chinese medicine. It is well established that shikonin possesses remarkable therapeutic actions on various diseases, with the underlying mechanisms, pharmacokinetics and toxicological effects elusive. Also, the clinical trial and pharmaceutical study of shikonin remain to be comprehensively delineated. PURPOSE The present review aimed to systematically summarize the updated knowledge regarding the therapeutic actions, pharmacokinetics, toxicological effects, clinical trial and pharmaceutical study of shikonin. METHODS The information contained in this review article were retrieved from some authoritative databases including Web of Science, PubMed, Google scholar, Chinese National Knowledge Infrastructure (CNKI), Wanfang Database and so on, till August 2021. RESULTS Shikonin exerts multiple therapeutic efficacies, such as anti-inflammation, anti-cancer, cardiovascular protection, anti-microbiomes, analgesia, anti-obesity, brain protection, and so on, mainly by regulating the NF-κB, PI3K/Akt/MAPKs, Akt/mTOR, TGF-β, GSK3β, TLR4/Akt signaling pathways, NLRP3 inflammasome, reactive oxygen stress, Bax/Bcl-2, etc. In terms of pharmacokinetics, shikonin has an unfavorable oral bioavailability, 64.6% of the binding rate of plasma protein, and enhances some metabolic enzymes, particularly including cytochrome P450. In regard to the toxicological effects, shikonin may potentially cause nephrotoxicity and skin allergy. The above pharmacodynamics and pharmacokinetics of shikonin have been validated by few clinical trials. In addition, pharmaceutical innovation of shikonin with novel drug delivery system such as nanoparticles, liposomes, microemulsions, nanogel, cyclodextrin complexes, micelles and polymers are beneficial to the development of shikonin-based drugs. CONCLUSIONS Shikonin is a promising phytochemical for drug candidates. Extensive and intensive explorations on shikonin are warranted to expedite the utilization of shikonin-based drugs in the clinical setting.
Collapse
Affiliation(s)
- Qiang Sun
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Ting Gong
- Department of Ultrasound, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 611731, Sichuan, China
| | - Maolun Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Shan Ren
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Han Yang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Sha Zeng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Hui Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Li Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Tianqi Ming
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xianli Meng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Haibo Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| |
Collapse
|
10
|
Zhu L, Li K, Liu M, Liu K, Ma S, Cai W. Anti-cancer Research on Arnebiae Radix-derived Naphthoquinone in Recent Five Years. Recent Pat Anticancer Drug Discov 2021; 17:218-230. [PMID: 34886780 DOI: 10.2174/1574892816666211209164745] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 09/13/2021] [Accepted: 09/21/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND In recent years, many naphthoquinone compounds with anticancer activity have been identified in Arnebiae Radix, and some of them have the potential to be developed into anticancer drugs. OBJECTIVE This article aimed to provide a comprehensive overview of the anticancer effects of naphthoquinone compounds through a detailed review of literature and Chinese patents, and discuss their potential to be developed as anticancer drugs for clinical application. METHODS Research papers were collected through the databases of PubMed, Cnki and SciDirect using keyword searches "naphthoquinone compounds" and "anticancer". The keywords of "shikonin" and "shikonin derivatives" were also used in PubMed, Cnki and SciDirect databases to collect research articles. The Chinese patents were collected using the Cnki patent database. RESULTS Naphthoquinone compounds have been found to possess anti-cancer activity, and their modes of action are associated with inducing apoptosis, inhibiting cancer cell proliferation, promoting autophagy in cancer cells, anti-cancer angiogenesis and inhibition of cell adhesion, invasion and metastasis, inhibiting glycolysis and inhibiting DNA topoisomerase activity. CONCLUSION Most of the naphthoquinone compounds show effective anti-cancer activity in vitro. The structure modification of naphthoquinone aims to develop anti-cancer drugs with high efficacy and low toxicity.
Collapse
Affiliation(s)
- Lian Zhu
- School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua 41800. China
| | - Kailin Li
- School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua 41800. China
| | - Mingjuan Liu
- School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua 41800. China
| | - Kexin Liu
- School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua 41800. China
| | - Shengjun Ma
- School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua 41800. China
| | - Wei Cai
- School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua 41800. China
| |
Collapse
|
11
|
Wang Q, Wang J, Wang J, Ju X, Zhang H. Molecular mechanism of shikonin inhibiting tumor growth and potential application in cancer treatment. Toxicol Res (Camb) 2021; 10:1077-1084. [PMID: 34956612 PMCID: PMC8692723 DOI: 10.1093/toxres/tfab107] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 09/13/2021] [Accepted: 10/27/2021] [Indexed: 12/20/2022] Open
Abstract
Shikonin is one of the major bioactive components of Lithospermum erythrorhizon. It has a good killing effect in a variety of tumor cells. Its antitumor effect involves multiple targets and pathways and has received extensive attention and study in recent years. In this review, we systematically review recent progress in determining the antitumor mechanism of shikonin and its derivatives, specifically their induction of reactive oxygen species production, inhibition of EGFR and PI3K/AKT signaling pathway activation, inhibition of angiogenesis and induction of apoptosis and necroptosis. We also discuss the application of nanoparticles loaded with shikonin in the targeted therapy of various cancers. Finally, we suggest new strategies for the clinical application of shikonin and its derivatives.
Collapse
Affiliation(s)
- Qiang Wang
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu, 212013, PR China
| | - Jing Wang
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu, 212013, PR China
| | - Jiayou Wang
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu, 212013, PR China
| | - Xiaoli Ju
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Heng Zhang
- Department of General Surgery, Nanjing Lishui District People’s Hospital, Zhongda Hospital Lishui Branch, Southeast University, Nanjing, China
| |
Collapse
|
12
|
Tsai MF, Chen SM, Ong AZ, Chung YH, Chen PN, Hsieh YH, Kang YT, Hsu LS. Shikonin Induced Program Cell Death through Generation of Reactive Oxygen Species in Renal Cancer Cells. Antioxidants (Basel) 2021; 10:antiox10111831. [PMID: 34829701 PMCID: PMC8615048 DOI: 10.3390/antiox10111831] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 11/09/2021] [Accepted: 11/16/2021] [Indexed: 01/26/2023] Open
Abstract
Shikonin mitigated tumor cell proliferation by elevating reactive oxygen species (ROS) levels. Herein, we investigated the effects of shikonin on renal cancer cell (RCC) cell proliferation. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay indicated that shikonin dose-dependently reduced the proliferation of Caki-1 and ACHN cells. Shikonin remarkably triggered necrosis and apoptosis in Caki-1 and ACHN cells in proportion to its concentration. Moreover, necrostatin-1 recovered cell viability in the presence of shikonin. Elevated ROS levels and mitochondrial dysfunction were also found in shikonin treatment groups. Pretreatment with N-acetyl cysteine remarkably mitigated shikonin-induced cell death and ROS generation. Western blot analysis revealed that shikonin reduced pro-PARP, pro-caspase-3, and Bcl-2 expression and increased cleavage PARP expression. Enhanced autophagy was also found in the shikonin-treated group as evidenced by acridine orange staining. Moreover, light chain 3B (LC3B)-II accumulation and enhanced p62 expression indicated that autophagy occurred in the shikonin-treated group. LC3B knockdown considerably recovered cell viability in the presence of shikonin. Shikonin treatment elevated p38 activity in a dose-dependent manner. In conclusion, our results revealed that shikonin triggered programmed cell death via the elevation of ROS level and p38 activity in different types of RCC cells. These findings suggested that shikonin may be a potential anti-RCC agent.
Collapse
Affiliation(s)
- Ming-Feng Tsai
- Department of Nephrology, Antai Medical Care Cooperation Antai Tian-Sheng Memorial Hospital, Pingtung 92842, Taiwan;
| | - Shih-Ming Chen
- Bachelor Program in Health Care and Social Work for Indigenous Students, Providence University, Taichung 43301, Taiwan;
| | - Ann-Zhi Ong
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan; (A.-Z.O.); (Y.-H.C.); (P.-N.C.); (Y.-H.H.); (Y.-T.K.)
| | - Yi-Hsuan Chung
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan; (A.-Z.O.); (Y.-H.C.); (P.-N.C.); (Y.-H.H.); (Y.-T.K.)
| | - Pei-Ni Chen
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan; (A.-Z.O.); (Y.-H.C.); (P.-N.C.); (Y.-H.H.); (Y.-T.K.)
| | - Yi-Hsien Hsieh
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan; (A.-Z.O.); (Y.-H.C.); (P.-N.C.); (Y.-H.H.); (Y.-T.K.)
| | - Yu-Ting Kang
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan; (A.-Z.O.); (Y.-H.C.); (P.-N.C.); (Y.-H.H.); (Y.-T.K.)
| | - Li-Sung Hsu
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan; (A.-Z.O.); (Y.-H.C.); (P.-N.C.); (Y.-H.H.); (Y.-T.K.)
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
- Correspondence:
| |
Collapse
|
13
|
Wang T, Ma M, Chen C, Yang X, Qian Y. Three widely used pesticides and their mixtures induced cytotoxicity and apoptosis through the ROS-related caspase pathway in HepG2 cells. Food Chem Toxicol 2021; 152:112162. [PMID: 33813062 DOI: 10.1016/j.fct.2021.112162] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 03/01/2021] [Accepted: 03/28/2021] [Indexed: 02/06/2023]
Abstract
Difenoconazole, cypermethrin and triazophos are widely used pesticides in agricultural production and frequently detected in foods. The aim of this study was to determine the effect of these pesticides and their mixtures on cell viability, reactive oxygen species (ROS), lactate dehydrogenase (LDH) content, apoptosis rate and DNA fragmentation and synthesis in human hepatocellular carcinoma cells (HepG2). The order of inhibitory effects for the individual pesticides was ranked as difenoconazole > cypermethrin > triazophos. The enhanced expression of caspase-3, caspase-7 and PARP activity was observed in HepG2 cells, which was 1.7, 1.3 and 1.6-fold higher than the control, respectively, along with significant protein cleavage; and induced apoptosis in a concentration-dependent manner. Further, the pesticide mixtures significantly increased ROS level (up to 1.3-fold), induced DNA fragmentation (up to 1.8-fold), inhibited DNA synthesis (up to 53%), and damaged the cells by destroying the cell membrane and producing a large amount of LDH at concentration range of 10-30 μM. Specifically, mixtures containing difenoconazole showed stronger toxicities than individual pesticides, implying higher health risks associated with mixtures. Our results show that three widely used pesticides exhibited cytotoxicity and apoptosis through the ROS-related caspase pathway, providing a basis for evaluation of health risks from pesticide mixtures via food consumption.
Collapse
Affiliation(s)
- Tiancai Wang
- Key Laboratory of Argo-Product Quality and Safety of Ministry of Agriculture, Institute of Quality Standards and Testing Technology for Argo-Products, Chinese Academy of Agricultural Sciences, NO.12 Zhong-guan-cun South Street, Haidian District, Beijing, 100081, People's Republic of China
| | - Mengmeng Ma
- Key Laboratory of Argo-Product Quality and Safety of Ministry of Agriculture, Institute of Quality Standards and Testing Technology for Argo-Products, Chinese Academy of Agricultural Sciences, NO.12 Zhong-guan-cun South Street, Haidian District, Beijing, 100081, People's Republic of China
| | - Chen Chen
- Key Laboratory of Argo-Product Quality and Safety of Ministry of Agriculture, Institute of Quality Standards and Testing Technology for Argo-Products, Chinese Academy of Agricultural Sciences, NO.12 Zhong-guan-cun South Street, Haidian District, Beijing, 100081, People's Republic of China.
| | - Xi Yang
- Key Laboratory of Argo-Product Quality and Safety of Ministry of Agriculture, Institute of Quality Standards and Testing Technology for Argo-Products, Chinese Academy of Agricultural Sciences, NO.12 Zhong-guan-cun South Street, Haidian District, Beijing, 100081, People's Republic of China
| | - Yongzhong Qian
- Key Laboratory of Argo-Product Quality and Safety of Ministry of Agriculture, Institute of Quality Standards and Testing Technology for Argo-Products, Chinese Academy of Agricultural Sciences, NO.12 Zhong-guan-cun South Street, Haidian District, Beijing, 100081, People's Republic of China
| |
Collapse
|
14
|
Shen XB, Wang Y, Han XZ, Sheng LQ, Wu FF, Liu X. Design, synthesis and anticancer activity of naphthoquinone derivatives. J Enzyme Inhib Med Chem 2020; 35:773-785. [PMID: 32200656 PMCID: PMC7144209 DOI: 10.1080/14756366.2020.1740693] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Basis on molecular docking and pharmacophore analysis of naphthoquinone moiety, a total of 23 compounds were designed and synthesised. With the help of reverse targets searching, anti-cancer activity was preliminarily evaluated, most of them are effective against some tumour cells, especially compound 12: 1-(5,8-dihydroxy-1,4-dioxo-1,4-dihydronaphthalen-2-yl)-4-methylpent-3-en-1-yl-4-oxo-4-((4-phenoxyphenyl)amino) butanoate whose IC50 against SGC-7901 was 4.1 ± 2.6 μM. Meanwhile the anticancer mechanism of compound 12 had been investigated by AnnexinV/PI staining, immunofluorescence, Western blot assay and molecular docking. The results indicated that this compound might induce cell apoptosis and cell autophagy through regulating the PI3K signal pathway.
Collapse
Affiliation(s)
- Xiao-Bao Shen
- School of Pharmacy, Anhui Medical University, Hefei, PR China.,Engineering Research Center of Biomass Conversion and Pollution Prevention of Anhui Educational Institutions, Fuyang Normal University, Fuyang, PR China
| | - Yang Wang
- School of Pharmacy, Anhui Medical University, Hefei, PR China
| | - Xuan-Zhen Han
- Engineering Research Center of Biomass Conversion and Pollution Prevention of Anhui Educational Institutions, Fuyang Normal University, Fuyang, PR China
| | - Liang-Quan Sheng
- Engineering Research Center of Biomass Conversion and Pollution Prevention of Anhui Educational Institutions, Fuyang Normal University, Fuyang, PR China
| | - Fu-Fang Wu
- School of Pharmacy, Anhui Medical University, Hefei, PR China.,Engineering Research Center of Biomass Conversion and Pollution Prevention of Anhui Educational Institutions, Fuyang Normal University, Fuyang, PR China
| | - Xinhua Liu
- School of Pharmacy, Anhui Medical University, Hefei, PR China
| |
Collapse
|
15
|
Liao X, Bu Y, Jia Q. Traditional Chinese medicine as supportive care for the management of liver cancer: Past, present, and future. Genes Dis 2020; 7:370-379. [PMID: 32884991 PMCID: PMC7452431 DOI: 10.1016/j.gendis.2019.10.016] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 10/23/2019] [Accepted: 10/25/2019] [Indexed: 12/24/2022] Open
Abstract
Liver cancer is the sixth most commonly diagnosed cancer and the fourth leading cause of cancer death worldwide. Western medicine and therapies are the primary treatment strategies of hepatocellular carcinoma (HCC), but the general prognosis for HCC patients is still dismal. Under these circumstances, HCC prevention is particularly important. Traditional Chinese medicine (TCM) encompasses a wealth of documented therapeutic resources, and "preventative treatment" is the principle of TCM. In China, TCM has been used for HCC prevention for thousands of years, and has also been demonstrated to be effective for the treatment of HCC in modern China. However, the TCM theory for prevention and treatment of HCC is more widely accepted in China than abroad. In this review, we first summarize the herbs and ancient formulas with therapeutic effects on HCC. We also review the research status of TCM in modern medicine as well as the current obstacles in its development. Finally, we discuss the future of TCM in the context of precision and integrated medicine. After reviewing the literature, we believe that TCM, through ancient development, is an advanced method of cancer treatment with positive curative effects, despite its surrounding controversy. Furthermore, precise analyses and systematic research methods provides novel approaches to modernize TCM for the future.
Collapse
Affiliation(s)
- Xia Liao
- Department of Nutrition, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Yang Bu
- Department of Hepatobiliary Surgery, General Hospital, Ningxia Medical University, Yinchuan, 750001, China
| | - Qingan Jia
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| |
Collapse
|
16
|
Ahmadi ES, Tajbakhsh A, Iranshahy M, Asili J, Kretschmer N, Shakeri A, Sahebkar A. Naphthoquinone Derivatives Isolated from Plants: Recent Advances in Biological Activity. Mini Rev Med Chem 2020; 20:2019-2035. [PMID: 32811411 DOI: 10.2174/1389557520666200818212020] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 05/22/2020] [Accepted: 05/29/2020] [Indexed: 11/22/2022]
Abstract
Naturally occurring naphthoquinones (NQs) comprising highly reactive small molecules are the subject of increasing attention due to their promising biological activities such as antioxidant, antimicrobial, apoptosis-inducing activities, and especially anticancer activity. Lapachol, lapachone, and napabucasin belong to the NQs and are in phase II clinical trials for the treatment of many cancers. This review aims to provide a comprehensive and updated overview on the biological activities of several new NQs isolated from different species of plants reported from January 2013 to January 2020, their potential therapeutic applications and their clinical significance.
Collapse
Affiliation(s)
- Esmaeil Sheikh Ahmadi
- Department of Pharmacognosy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Tajbakhsh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Milad Iranshahy
- Department of Pharmacognosy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Javad Asili
- Department of Pharmacognosy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Nadine Kretschmer
- Department of Pharmacognosy, Institute of Pharmaceutical Sciences, University of Graz, Graz, Austria
| | - Abolfazl Shakeri
- Department of Pharmacognosy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
17
|
Hao Y, Huang Y, Chen J, Li J, Yuan Y, Wang M, Han L, Xin X, Wang H, Lin D, Peng F, Yu F, Zheng C, Shen C. Exopolysaccharide from Cryptococcus heimaeyensis S20 induces autophagic cell death in non-small cell lung cancer cells via ROS/p38 and ROS/ERK signalling. Cell Prolif 2020; 53:e12869. [PMID: 32597573 PMCID: PMC7445402 DOI: 10.1111/cpr.12869] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 06/03/2020] [Accepted: 06/13/2020] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVES Cryptococcus heimaeyensis S20 is found in Antarctica and can produce exopolysaccharides (CHEPS). Here, we explore the anti-tumour effects of CHEPS on non-small cell lung cancer (NSCLC). MATERIALS AND METHODS Cell viability was assessed by CCK8 and colony formation assays. Flow cytometry was used to analyse the cell cycle, cell apoptosis and reactive oxygen species (ROS). Cell autophagy was detected by EGFP-LC3 puncta assay, Lyso-Tracker Red staining and transmission electron microscopy. mRNA and protein levels were analysed by qRT-PCR and Western blot. Related mechanisms were confirmed using appropriate inhibitors or shRNA. In vitro results were further confirmed by a tumour xenograft study. RESULTS CHEPS inhibited the proliferation of NSCLC cells by inducing S- and G2/M-phase arrest and autophagic cell death, but not apoptosis. CHEPS was less toxic to normal human embryonic lung fibroblasts. CHEPS activated the MAPK pathway in NSCLC cells, and p38 and ERK promoted CHEPS-induced cell death. Further studies showed that p38 and ERK promoted CHEPS-induced NSCLC cell autophagy and ERK promoted CHEPS-induced S- and G2/M-phase arrest. ROS were induced by CHEPS. A ROS scavenger attenuated CHEPS-induced p38 and ERK activation, autophagy and cell death. Finally, CHEPS reduced orthotopic lung tumour growth without organ-related toxicity. CHEPS also induced ROS, activated p38 and ERK, and triggered autophagy in vivo. CONCLUSIONS CHEPS induces autophagic cell death and S- and G2/M-phase arrest in NSCLC cells via ROS/p38 and ROS/ERK signalling.
Collapse
Affiliation(s)
- Yao Hao
- State Key Laboratory of VirologyCollege of Life SciencesWuhan UniversityWuhanChina
| | - Yao Huang
- College of Life SciencesWuhan UniversityWuhanChina
| | - Jingyi Chen
- State Key Laboratory of VirologyCollege of Life SciencesWuhan UniversityWuhanChina
| | - Jiadai Li
- State Key Laboratory of VirologyCollege of Life SciencesWuhan UniversityWuhanChina
| | - Yuncong Yuan
- State Key Laboratory of VirologyCollege of Life SciencesWuhan UniversityWuhanChina
| | - Mingzhen Wang
- State Key Laboratory of VirologyCollege of Life SciencesWuhan UniversityWuhanChina
| | - Lingling Han
- State Key Laboratory of VirologyCollege of Life SciencesWuhan UniversityWuhanChina
| | - Xiu Xin
- Institute of Pathogenic Microorganism and College of Bioscience and EngineeringJiangxi Agricultural UniversityNanchangChina
| | - Hailong Wang
- State Key Laboratory of VirologyCollege of Life SciencesWuhan UniversityWuhanChina
| | - Danqing Lin
- College of Life SciencesWuhan UniversityWuhanChina
| | - Fang Peng
- College of Life SciencesWuhan UniversityWuhanChina
- China Center for Type Culture CollectionWuhan UniversityWuhanChina
| | - Fang Yu
- Department of PathologyZhongnan HospitalWuhan University
| | - Congyi Zheng
- State Key Laboratory of VirologyCollege of Life SciencesWuhan UniversityWuhanChina
- China Center for Type Culture CollectionWuhan UniversityWuhanChina
| | - Chao Shen
- State Key Laboratory of VirologyCollege of Life SciencesWuhan UniversityWuhanChina
- China Center for Type Culture CollectionWuhan UniversityWuhanChina
| |
Collapse
|
18
|
Lan XO, Wang HX, Qi RQ, Xu YY, Yu YJ, Yang Y, Guo H, Gao XH, Geng L. Shikonin inhibits CEBPD downregulation in IL‑17‑treated HaCaT cells and in an imiquimod‑induced psoriasis model. Mol Med Rep 2020; 22:2263-2272. [PMID: 32705251 PMCID: PMC7411367 DOI: 10.3892/mmr.2020.11315] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Accepted: 06/08/2020] [Indexed: 01/16/2023] Open
Abstract
Psoriasis is a chronic inflammatory skin disease characterized by well-defined scaly papules and plaques. Interleukin (IL)-17 is involved in its pathogenesis and promotes the proliferation of epidermal keratinocytes through signal transducer and activator of transcription 3 (STAT3) activation. Shikonin, a natural naphthoquinone isolated from Lithospermum erythrorhizon, possesses anti-inflammatory and immunosuppressive properties and can suppress IL-17-induced vascular endothelial growth factor expression by inhibiting the JAK/STAT3 pathway. In the present study, MTS, iCELLigence and RT-qPCR were used to determine the optimal concentration and duration of IL-17 or shikonin acting on HaCaT cells. The changes in the expression levels of genes associated with the IL-6/STAT3 pathway in differentially treated cells were analyzed via RT2Profiler™ PCR Array. Small interfering RNA was used to silence the expression levels of the target gene CCAAT/enhancer-binding protein δ (CEBPD). Western blotting and immunohistochemistry were used to evaluate the effect of shikonin on imiquimod-induced psoriasis in mice and the expression levels of CEBPD. Shikonin reversed IL-17-mediated downregulation of the tumor suppressor CEBPD in HaCaT cells. Moreover, low levels of CEBPD in the imiquimod-induced mouse model of psoriasis were restored by shikonin treatment, which ameliorated excessive keratinocyte proliferation. Taken together, these findings suggest that CEBPD plays a key role in the pathogenesis of psoriasis and can be targeted by shikonin as a potential therapeutic strategy.
Collapse
Affiliation(s)
- Xiao-Ou Lan
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - He-Xiao Wang
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Rui-Qun Qi
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Yuan-Yuan Xu
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Ya-Jie Yu
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Yang Yang
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Hao Guo
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Xing-Hua Gao
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Long Geng
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
19
|
Wang Y, Zhu F, Zhang Y, Chen C, Lai Y, Sun J, Chen S, Qiu P, Gao J, Deng G. Shikonin suppresses trophoblast cell growth via regulation of GLI1, and p62 mediated caspase 8 activation. Reprod Toxicol 2020; 95:104-112. [PMID: 32461113 DOI: 10.1016/j.reprotox.2020.05.011] [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: 01/10/2020] [Revised: 05/10/2020] [Accepted: 05/18/2020] [Indexed: 10/24/2022]
Abstract
Unruptured ectopic pregnancy (UEP) is a common cause of morbidity and, occasionally, of mortality in women of reproductive age. Pharmacological intervention is a common therapeutic approach for early-stage UEP. Herein, we investigated the cytotoxic effect and novel mechanism of shikonin, a natural naphthoquinone pigment purified from Lithospermum erythrorhizon, in human trophoblast cells. These data demonstrated that shikonin suppressed proliferation and induced apoptosis in a time-dependent manner in HTR-8/SVneo cells. Shikonin blocked autophagic flux and promoted p62 interaction with caspase 8, resulting in caspase 8 activation. Moreover, shikonin suppressed GLI1 expression, and GLI1 overexpression attenuated shikonin-induced cell apoptosis. Although silencing GLI1 slightly promoted cell apoptosis, p62 overexpression enhanced GLI1 silencing-induced cell apoptosis by activating caspase 8. Furthermore, rapamycin increased shikonin-induced cell apoptosis in HTR-8/SVneo cells, whereas 3-MA attenuated the cytotoxic effect of shikonin. In conclusion, shikonin suppressed trophoblast cell growth by silencing GLI1 and increasing p62 co-mediated activation of caspase 8, which suggested a potential novel therapeutic target for UEP.
Collapse
Affiliation(s)
- Yanxi Wang
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Fangfang Zhu
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Yingxuan Zhang
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Chunlin Chen
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Yuling Lai
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Jianhua Sun
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Si Chen
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Pin Qiu
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Jie Gao
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| | - Gaopi Deng
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| |
Collapse
|
20
|
Cao H, Zhang W, Liu D, Hou M, Liu S, He W, Lin J, Shao M. Identification, in vitro evaluation and modeling studies of the constituents from the roots of Arnebia euchroma for antitumor activity and STAT3 inhibition. Bioorg Chem 2020; 96:103655. [DOI: 10.1016/j.bioorg.2020.103655] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 01/16/2020] [Accepted: 02/05/2020] [Indexed: 12/24/2022]
|
21
|
Luo H, Vong CT, Chen H, Gao Y, Lyu P, Qiu L, Zhao M, Liu Q, Cheng Z, Zou J, Yao P, Gao C, Wei J, Ung COL, Wang S, Zhong Z, Wang Y. Naturally occurring anti-cancer compounds: shining from Chinese herbal medicine. Chin Med 2019; 14:48. [PMID: 31719837 PMCID: PMC6836491 DOI: 10.1186/s13020-019-0270-9] [Citation(s) in RCA: 318] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 10/23/2019] [Indexed: 12/24/2022] Open
Abstract
Numerous natural products originated from Chinese herbal medicine exhibit anti-cancer activities, including anti-proliferative, pro-apoptotic, anti-metastatic, anti-angiogenic effects, as well as regulate autophagy, reverse multidrug resistance, balance immunity, and enhance chemotherapy in vitro and in vivo. To provide new insights into the critical path ahead, we systemically reviewed the most recent advances (reported since 2011) on the key compounds with anti-cancer effects derived from Chinese herbal medicine (curcumin, epigallocatechin gallate, berberine, artemisinin, ginsenoside Rg3, ursolic acid, silibinin, emodin, triptolide, cucurbitacin B, tanshinone I, oridonin, shikonin, gambogic acid, artesunate, wogonin, β-elemene, and cepharanthine) in scientific databases (PubMed, Web of Science, Medline, Scopus, and Clinical Trials). With a broader perspective, we focused on their recently discovered and/or investigated pharmacological effects, novel mechanism of action, relevant clinical studies, and their innovative applications in combined therapy and immunomodulation. In addition, the present review has extended to describe other promising compounds including dihydroartemisinin, ginsenoside Rh2, compound K, cucurbitacins D, E, I, tanshinone IIA and cryptotanshinone in view of their potentials in cancer therapy. Up to now, the evidence about the immunomodulatory effects and clinical trials of natural anti-cancer compounds from Chinese herbal medicine is very limited, and further research is needed to monitor their immunoregulatory effects and explore their mechanisms of action as modulators of immune checkpoints.
Collapse
Affiliation(s)
- Hua Luo
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Chi Teng Vong
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Hanbin Chen
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Yan Gao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Peng Lyu
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Ling Qiu
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Mingming Zhao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Qiao Liu
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Zehua Cheng
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Jian Zou
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Peifen Yao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Caifang Gao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Jinchao Wei
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Carolina Oi Lam Ung
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Shengpeng Wang
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Zhangfeng Zhong
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Yitao Wang
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| |
Collapse
|
22
|
Liu Y, Kang X, Niu G, He S, Zhang T, Bai Y, Li Y, Hao H, Chen C, Shou Z, Li B. Shikonin induces apoptosis and prosurvival autophagy in human melanoma A375 cells via ROS-mediated ER stress and p38 pathways. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:626-635. [PMID: 30873870 DOI: 10.1080/21691401.2019.1575229] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Shikonin, a botanical drug extracted from Lithospermum erythrorhizon, exhibits anti-cancer effects in various cancer cell lines. However, the mechanisms underlying these effects have not been completely elucidated yet. Here, we showed that Shikonin induces apoptosis and autophagy in A375 cells and inhibits their proliferation. Shikonin caused G2/M phase arrest through upregulation of p21 and downregulation of cyclin B1. Shikonin significantly triggered ER stress-mediated apoptosis by upregulating the expression of p-eIF2α, CHOP, and cleaved caspase-3. It also induced protective autophagy by activating the p38 pathway, followed by an increase in the levels of p-p38, LC3B-II, and Beclin 1. Upon suppression of autophagy by 3-methyladenine, Shikonin-induced apoptosis was enhanced in A375 cells. Moreover, after pretreatment with N-acetyl-cysteine, Shikonin increased the production of reactive oxygen species that are involved in regulating ER stress-mediated apoptosis and p38-activated autophagy, as evidenced by the reversion of cell viability and apoptosis and a decrease in p-eIF2α, CHOP, p-p38, LC3B-II, and Beclin 1 levels. Thus, we demonstrated that Shikonin induced apoptosis and autophagy in A375 cells via the activation of ROS-mediated ER stress and p38 pathways, indicating that Shikonin can serve as a potential agent for human melanoma therapy.
Collapse
Affiliation(s)
- Yongkang Liu
- a Ministry of Education, Key Laboratory of Resource Biology and Biotechnology in Western China , Northwest University , Xi'an , China.,b School of Life Sciences , Northwest University , Xi'an , Shaanxi , China.,c National Engineering Research Center for Miniaturized Detection Systems, Northwest University , Xi'an , China
| | - Xing Kang
- a Ministry of Education, Key Laboratory of Resource Biology and Biotechnology in Western China , Northwest University , Xi'an , China.,b School of Life Sciences , Northwest University , Xi'an , Shaanxi , China.,c National Engineering Research Center for Miniaturized Detection Systems, Northwest University , Xi'an , China
| | - Geng Niu
- a Ministry of Education, Key Laboratory of Resource Biology and Biotechnology in Western China , Northwest University , Xi'an , China.,b School of Life Sciences , Northwest University , Xi'an , Shaanxi , China.,c National Engineering Research Center for Miniaturized Detection Systems, Northwest University , Xi'an , China
| | - Senlin He
- a Ministry of Education, Key Laboratory of Resource Biology and Biotechnology in Western China , Northwest University , Xi'an , China.,b School of Life Sciences , Northwest University , Xi'an , Shaanxi , China.,c National Engineering Research Center for Miniaturized Detection Systems, Northwest University , Xi'an , China
| | - Tingting Zhang
- a Ministry of Education, Key Laboratory of Resource Biology and Biotechnology in Western China , Northwest University , Xi'an , China.,b School of Life Sciences , Northwest University , Xi'an , Shaanxi , China.,c National Engineering Research Center for Miniaturized Detection Systems, Northwest University , Xi'an , China
| | - Yuwei Bai
- a Ministry of Education, Key Laboratory of Resource Biology and Biotechnology in Western China , Northwest University , Xi'an , China.,b School of Life Sciences , Northwest University , Xi'an , Shaanxi , China.,c National Engineering Research Center for Miniaturized Detection Systems, Northwest University , Xi'an , China
| | - Yi Li
- d School of Computer Science , Xi'an Polytechnic University , Xi'an , China
| | - Houyan Hao
- a Ministry of Education, Key Laboratory of Resource Biology and Biotechnology in Western China , Northwest University , Xi'an , China
| | - Chao Chen
- b School of Life Sciences , Northwest University , Xi'an , Shaanxi , China.,c National Engineering Research Center for Miniaturized Detection Systems, Northwest University , Xi'an , China
| | - Zhexing Shou
- e Department of Integrated Traditional Chinese and Western Medicine , Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| | - Bin Li
- a Ministry of Education, Key Laboratory of Resource Biology and Biotechnology in Western China , Northwest University , Xi'an , China.,b School of Life Sciences , Northwest University , Xi'an , Shaanxi , China.,c National Engineering Research Center for Miniaturized Detection Systems, Northwest University , Xi'an , China
| |
Collapse
|
23
|
ROS -mediated p53 activation by juglone enhances apoptosis and autophagy in vivo and in vitro. Toxicol Appl Pharmacol 2019; 379:114647. [PMID: 31283929 DOI: 10.1016/j.taap.2019.114647] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Revised: 07/03/2019] [Accepted: 07/04/2019] [Indexed: 02/07/2023]
Abstract
Juglone (JG) exhibits a broad-spectrum of cytotoxicity against some cancer cells. However, its molecular mechanisms have not been investigated well. Here, the present results showed that JG significantly inhibited tumor growth in vivo. CCK-8 assays, flow cytometric analysis, western blotting and immunohistochemistry revealed that JG effectively inhibited cell proliferation and induced apoptosis through extrinsic pathways. We also observed that JG treatment induced autophagy flux via activiting the AMPK-mTOR signaling pathway. In addition, we found that JG enhanced p53 activation by increasing down-regulation of ubiquitin-mediated degradation. Inhibition of p53 by siRNA attenuated JG-induced cell death and autophagy. Moreover, JG enhanced the generation of hydrogen peroxide (H2O2) and superoxide anion radical (O2• -). Further experiments proved that H2O2 was a major factor since the H2O2 scavenger catalase (CAT) reduced both autophagy and cell death to a greater extent than the O2• - scavenger SOD. Overall, our results illustrated that JG caused apoptosis and autophagy via activating the ROS-mediated p53 pathway in human liver cancer cells in vitro and in vivo, which provided basic scientific evidence that JG serves as a potential anti-cancer agent.
Collapse
|
24
|
Mbaveng AT, Bitchagno GTM, Kuete V, Tane P, Efferth T. Cytotoxicity of ungeremine towards multi-factorial drug resistant cancer cells and induction of apoptosis, ferroptosis, necroptosis and autophagy. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 60:152832. [PMID: 31031043 DOI: 10.1016/j.phymed.2019.152832] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 01/09/2019] [Accepted: 01/12/2019] [Indexed: 05/27/2023]
Abstract
BACKGROUND Successful cancer chemotherapy is hampered by resistance of cancer cells to established anticancer drugs. Numerous natural products reveal cytotoxicity towards tumor cells. PURPOSE The present study was aimed to determine the cytotoxicity of a betaine-type alkaloid, ungeremine, towards 9 cancer cell lines including various sensitive and drug-resistant phenotypes. The mode of action of this compound was further investigated. METHODS The cytotoxicity, ferroptotic and necroptotic cell death were determined by the resazurin reduction assay. Caspase activation was evaluated using the caspase-Glo assay. Flow cytometry was applied for the analysis of cell cycle analysis (PI staining), apoptosis (annexin V/PI staining), mitochondrial membrane potential (MMP) (JC-1) and reactive oxygen species (ROS) (H2DCFH-DA). Apoptotic, necroptotic and autophagic markers were determined by Western blotting. CCRF-CEM leukemia cells were used for all mechanistic studies. RESULTS Ungeremine displayed cytotoxic activity towards the 9 cancer cell lines tested, including drug-sensitive and MDR phenotypes. The IC50values obtained varied from 3.67 µM (in MDA-MB-231-BCRP breast carcinoma cells) to 75.24 µM (against in CEM/ADR5000 leukemia cells) for ungeremine and from 0.02 µM (against CCRF-CEM cells) to 122.96 µM (against CEM/ADR5000 cells) for doxorubicin (control drug). Ungeremine induced ferroptosis, necroptosis, autophagy as well as apoptosis mediated by caspase activation, MMP alteration and increase ROS production. CONCLUSION The present investigation showed that ungeremine is a promising cytotoxic compoundthat could be further explored in the future to develop new anticancer drugs to fight sensitive and resistant phenotypes.
Collapse
Affiliation(s)
- Armelle T Mbaveng
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, University of Mainz, Staudinger Weg 5, 55128 Mainz, Germany; Department of Biochemistry, Faculty of Science, University of Dschang, P.O. Box 67, Dschang, Cameroon
| | - Gabin T M Bitchagno
- Department of Chemistry, Faculty of Science, University of Dschang, P.O. Box 67, Dschang, Cameroon
| | - Victor Kuete
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, University of Mainz, Staudinger Weg 5, 55128 Mainz, Germany; Department of Biochemistry, Faculty of Science, University of Dschang, P.O. Box 67, Dschang, Cameroon
| | - Pierre Tane
- Department of Chemistry, Faculty of Science, University of Dschang, P.O. Box 67, Dschang, Cameroon
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, University of Mainz, Staudinger Weg 5, 55128 Mainz, Germany.
| |
Collapse
|
25
|
Shikonin derivatives for cancer prevention and therapy. Cancer Lett 2019; 459:248-267. [PMID: 31132429 DOI: 10.1016/j.canlet.2019.04.033] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 04/15/2019] [Accepted: 04/26/2019] [Indexed: 12/25/2022]
Abstract
Phytochemicals gained considerable interest during the past years as source to develop new treatment options for chemoprevention and cancer therapy. Motivated by the fact that a majority of established anticancer drugs are derived in one way or another from natural resources, we focused on shikonin, a naphthoquinone with high potentials to be further developed as preventive or therapeutic drug to fight cancer. Shikonin is the major chemical component of Lithospermum erythrorhizon (Purple Cromwell) roots. Traditionally, the root extract has been applied to cure dermatitis, burns, and wounds. Over the past three decades, the anti-inflammatory and anticancer effects of root extracts, isolated shikonin as well as semi-synthetic and synthetic derivatives and nanoformulations have been described. In vitro and in vivo experiments were conducted to understand the effect of shikonin at cellular and molecular levels. Preliminary clinical trials indicate the potential of shikonin for translation into clinical oncology. Shikonin exerts additive and synergistic interactions in combination with established chemotherapeutics, immunotherapeutic approaches, radiotherapy and other treatment modalities, which further underscores the potential of this phytochemical to be integrated into standard treatment regimens.
Collapse
|
26
|
Jiang GM, Tan Y, Wang H, Peng L, Chen HT, Meng XJ, Li LL, Liu Y, Li WF, Shan H. The relationship between autophagy and the immune system and its applications for tumor immunotherapy. Mol Cancer 2019; 18:17. [PMID: 30678689 PMCID: PMC6345046 DOI: 10.1186/s12943-019-0944-z] [Citation(s) in RCA: 258] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 01/14/2019] [Indexed: 12/15/2022] Open
Abstract
Autophagy is a genetically well-controlled cellular process that is tightly controlled by a set of core genes, including the family of autophagy-related genes (ATG). Autophagy is a “double-edged sword” in tumors. It can promote or suppress tumor development, which depends on the cell and tissue types and the stages of tumor. At present, tumor immunotherapy is a promising treatment strategy against tumors. Recent studies have shown that autophagy significantly controls immune responses by modulating the functions of immune cells and the production of cytokines. Conversely, some cytokines and immune cells have a great effect on the function of autophagy. Therapies aiming at autophagy to enhance the immune responses and anti-tumor effects of immunotherapy have become the prospective strategy, with enhanced antigen presentation and higher sensitivity to CTLs. However, the induction of autophagy may also benefit tumor cells escape from immune surveillance and result in intrinsic resistance against anti-tumor immunotherapy. Increasing studies have proven the optimal use of either ATG inducers or inhibitors can restrain tumor growth and progression by enhancing anti-tumor immune responses and overcoming the anti-tumor immune resistance in combination with several immunotherapeutic strategies, indicating that induction or inhibition of autophagy might show us a prospective therapeutic strategy when combined with immunotherapy. In this article, the possible mechanisms of autophagy regulating immune system, and the potential applications of autophagy in tumor immunotherapy will be discussed.
Collapse
Affiliation(s)
- Guan-Min Jiang
- Department of Clinical laboratory, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China. .,Central Laboratory, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China.
| | - Yuan Tan
- Department of Clinical Laboratory, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Hao Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of University of Science and Technology of China, Hefei, Anhui, China
| | - Liang Peng
- Department of Clinical laboratory, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Hong-Tao Chen
- Department of Clinical laboratory, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Xiao-Jun Meng
- Department of Endocrinology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Ling-Ling Li
- Central Laboratory, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Yan Liu
- Department of Clinical laboratory, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Wen-Fang Li
- Department of Clinical laboratory, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Hong Shan
- Key Laboratory of Biomedical Imaging of Guangdong Province, Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China.
| |
Collapse
|
27
|
Autophagy and its potent modulators from phytochemicals in cancer treatment. Cancer Chemother Pharmacol 2018; 83:17-26. [PMID: 30353226 DOI: 10.1007/s00280-018-3707-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 10/15/2018] [Indexed: 12/12/2022]
Abstract
Autophagy is a ubiquitous catabolic process by which damaged or harmful intracellular components are delivered to the lysosomes for self-digestion and recycling. It is critical in cancer treatment. Therapy-induced autophagy predominantly acts as a pro-survival mechanism, but progressive autophagy can lead to non-apoptotic cell death, also known as autophagic cell death. Plants or herbs contain various natural compounds that are widely used in the treatment of many types of malignancies. Emerging evidence indicates that phytochemicals targeting the autophagic pathway are promising agents for cancer treatment. However, these compounds play different roles in autophagy. In this review, we discussed the role of autophagy in cancer development and therapy, and focussed on elucidating the anti-cancer activities of autophagic modulators, especially phytochemicals. Notably, we described a novel premise that the dynamic role of phytochemicals should be evaluated in regulation of autophagy in cancer.
Collapse
|
28
|
Huang C, Hu G. Shikonin suppresses proliferation and induces apoptosis in endometrioid endometrial cancer cells via modulating miR-106b/PTEN/AKT/mTOR signaling pathway. Biosci Rep 2018; 38:BSR20171546. [PMID: 29449346 PMCID: PMC5897745 DOI: 10.1042/bsr20171546] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 02/10/2018] [Accepted: 02/15/2018] [Indexed: 12/22/2022] Open
Abstract
Shikonin, a natural naphthoquinone isolated from a traditional Chinese medicinal herb, which exerts anticancer effects in various cancers. However, the molecular mechanisms underlying the therapeutic effects of shikonin against endometrioid endometrial cancer (EEC) have not yet been fully elucidated. Herein, we investigated anticancer effects of shikonin on EEC cells and explored the underlying molecular mechanism. We observed that shikonin inhibits proliferation in human EEC cell lines in a dose-dependent manner. Moreover, shikonin-induced apoptosis was characterized by the up-regulation of the pro-apoptotic proteins cleaved-Caspase-3 and Bax, and the down-regulation of the anti-apoptotic protein Bcl-2. Microarray analyses demonstrated that shikonin induces many miRNAs' dysregulation, and miR-106b was one of the miRNAs being most significantly down-regulated. miR-106b was identified to exert procancer effect in various cancers, but in EEC remains unclear. We first confirmed that miR-106b is up-regulated in EEC tissues and cells, and knockdown of miR-106b suppresses proliferation and promotes apoptosis. Meanwhile, our results validated that the restored expression of miR-106b abrogates the antiproliferative and pro-apoptotic effects of shikonin. We also identified that miR-106b targets phosphatase and tensin homolog (PTEN), a tumor suppressor gene, which in turn modulates AKT/mTOR signaling pathway. Our findings indicated that shikonin inhibits proliferation and promotes apoptosis in human EEC cells by modulating the miR-106b/PTEN/AKT/mTOR signaling pathway, suggesting shikonin could act a potential therapeutic agent in the EEC treatment.
Collapse
Affiliation(s)
- Caimei Huang
- Department of Traditional Chinese Medicine, Shanghai Changning Maternity and Infant Health Hospital, Shanghai 200051, China
| | - Guohua Hu
- Gynecology of Traditional Chinese Medicine, Shanghai Municipal Hospital of Traditional Chinese Medicine Affiliated to Shanghai TCM University, Shanghai 200071, China
| |
Collapse
|
29
|
Moosavi MA, Haghi A, Rahmati M, Taniguchi H, Mocan A, Echeverría J, Gupta VK, Tzvetkov NT, Atanasov AG. Phytochemicals as potent modulators of autophagy for cancer therapy. Cancer Lett 2018; 424:46-69. [PMID: 29474859 DOI: 10.1016/j.canlet.2018.02.030] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Revised: 02/18/2018] [Accepted: 02/19/2018] [Indexed: 02/07/2023]
Abstract
The dysregulation of autophagy is involved in the pathogenesis of a broad range of diseases, and accordingly universal research efforts have focused on exploring novel compounds with autophagy-modulating properties. While a number of synthetic autophagy modulators have been identified as promising cancer therapy candidates, autophagy-modulating phytochemicals have also attracted attention as potential treatments with minimal side effects. In this review, we firstly highlight the importance of autophagy and its relevance in the pathogenesis and treatment of cancer. Subsequently, we present the data on common phytochemicals and their mechanism of action as autophagy modulators. Finally, we discuss the challenges associated with harnessing the autophagic potential of phytochemicals for cancer therapy.
Collapse
Affiliation(s)
- Mohammad Amin Moosavi
- Department of Molecular Medicine, National Institute of Genetic Engineering and Biotechnology, P.O Box:14965/161, Tehran, Iran.
| | - Atousa Haghi
- Young Researchers & Elite Club, Pharmaceutical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Marveh Rahmati
- Cancer Biology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Hiroaki Taniguchi
- Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, 05-552 Jastrzebiec, Poland
| | - Andrei Mocan
- Department of Pharmaceutical Botany, "Iuliu Haţieganu" University of Medicine and Pharmacy, Gheorghe Marinescu 23 Street, 400337 Cluj-Napoca, Romania
| | - Javier Echeverría
- Facultad de Química y Biología, Universidad de Santiago de Chile, Casilla 40, Correo 33, Santiago 9170022, Chile
| | - Vijai K Gupta
- Department of Chemistry and Biotechnology, ERA Chair of Green Chemistry, Tallinn University of Technology, 12618 Tallinn, Estonia
| | - Nikolay T Tzvetkov
- Pharmaceutical Institute, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany; NTZ Lab Ltd., Krasno Selo 198, Sofia 1618, Bulgaria
| | - Atanas G Atanasov
- Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, 05-552 Jastrzebiec, Poland; Department of Pharmacognosy, University of Vienna, Althanstrasse 14, 1090 Vienna, Austria.
| |
Collapse
|
30
|
Spyrelli ED, Kyriazou AV, Virgiliou C, Nakas A, Deda O, Papageorgiou VP, Assimopoulou AN, Gika HG. Metabolic profiling study of shikonin's cytotoxic activity in the Huh7 human hepatoma cell line. MOLECULAR BIOSYSTEMS 2018; 13:841-851. [PMID: 28265634 DOI: 10.1039/c6mb00830e] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Shikonin and its enantiomer alkannin, which are natural products, have been extensively studied in vitro and in vivo for, among others, their antitumor activity. The investigation of the molecular pathways involved in their action is of interest, since they are not yet clearly defined. Metabolic profiling in cells can provide a picture of a cell's phenotype upon intervention, assisting in the elucidation of the mechanism of action. In this study, the cytotoxic effect of shikonin on a human hepatocarcinoma cell line was studied. Huh7 cells were treated with shikonin at 5 μM, and it was found that shikonin markedly inhibited cell growth. Metabolic profiling indicated alterations in the metabolic content of the cells and the culture media upon treatment, detecting the metabolic response of the cells. This study demonstrates the potential of metabolomics to improve knowledge on the mechanisms involved in shikonin's antitumor action.
Collapse
Affiliation(s)
- E D Spyrelli
- Department of Chemical Engineering, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Shikonin causes apoptosis by disrupting intracellular calcium homeostasis and mitochondrial function in human hepatoma cells. Exp Ther Med 2017; 15:1484-1492. [PMID: 29434733 PMCID: PMC5776525 DOI: 10.3892/etm.2017.5591] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 10/27/2017] [Indexed: 11/09/2022] Open
Abstract
Shikonin is known to suppress proliferation and induce apoptosis in a variety of cancer cell lines. In the present study, SMMC-7721 human hepatocellular carcinoma cells were treated with shikonin (1, 2 or 4 µM) for 12–48 h. Cell morphological alterations and DNA damage were determined. Furthermore, changes in cell cycle, mitochondrial transmembrane potential, calcium homeostasis and levels of reactive oxygen species were measured. Shikonin-treated SMMC-7721 cells exhibited morphological changes and DNA damage. Shikonin inhibited cell proliferation causing cell cycle arrest at the G0/G1 phase and induced apoptosis in a dose- and time-dependent manner. Shikonin-induced apoptosis was associated with activation of caspases-3, −8 and −9, elevated levels of intracellular Ca2+ and reactive oxygen species, reduced mitochondrial membrane potential and enhanced efflux of Ca2+ and K+. Gene expression B-cell lymphoma 2 (Bcl-2)-associated X protein (Bax), p53 and caspase-3 was up-regulated, whereas Bcl-2 expression was downregulated. Shikonin caused apoptosis by inhibiting cell cycle progression, disrupting Ca2+ homeostasis, inducing oxidative stress and triggering mitochondrial dysfunction. Activation of caspases-3, −8 and −9, K+ efflux, and regulation of Bax, Bcl-2, p53 and caspase-3 expression are involved in the process. These results provide in-depth insight into the mechanisms of action of shikonin in the treatment of cancer.
Collapse
|
32
|
Li CJ, Liao WT, Wu MY, Chu PY. New Insights into the Role of Autophagy in Tumor Immune Microenvironment. Int J Mol Sci 2017; 18:1566. [PMID: 28753959 PMCID: PMC5536054 DOI: 10.3390/ijms18071566] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 07/11/2017] [Accepted: 07/13/2017] [Indexed: 02/06/2023] Open
Abstract
The tumor microenvironment is a complex system that is affected by various factors, including hypoxia, acidosis, and immune and inflammatory responses, which have significant effects on tumor adhesion, invasion, metastasis, angiogenesis, and autophagy. In this hostile tumor microenvironment, autophagy of tumor cells can promote tumor growth and metastasis. As autophagy is a double-edged sword in tumors, treatment of cancer via regulation of autophagy is extremely complicated. Therefore, understanding the relationship between tumor autophagy and the tumor microenvironment is extremely important. As the immune milieu plays an important role in tumor development, immunotherapy has become a promising form of cancer therapy. A multi-pronged treatment approach using immunotherapy and molecular targets may become the major direction for future cancer treatments. This article reviews existing knowledge regarding the immune factors in the tumor microenvironment and the status of tumor autophagy research.
Collapse
Affiliation(s)
- Chia-Jung Li
- Research Assistant Center, Show Chwan Memorial Hospital, Changhua 500, Taiwan.
| | - Wan-Ting Liao
- Chinese Medicine Department, Show Chwan Memorial Hospital, Changhua 500, Taiwan.
- Graduate Institute of Chinese Medicine, China Medical University, Taichung 404, Taiwan.
| | - Meng-Yu Wu
- Department of Emergency Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan.
| | - Pei-Yi Chu
- Department of Pathology, Show Chwan Memorial Hospital, Changhua 500, Taiwan.
- School of Medicine, College of Medicine, Fu-Jen Catholic University, New Taipei City 242, Taiwan.
- National Institute of Cancer Research, National Health Research Institutes, Tainan 704, Taiwan.
| |
Collapse
|
33
|
Cao ZX, Yang YT, Yu S, Li YZ, Wang WW, Huang J, Xie XF, Xiong L, Lei S, Peng C. Pogostone induces autophagy and apoptosis involving PI3K/Akt/mTOR axis in human colorectal carcinoma HCT116 cells. JOURNAL OF ETHNOPHARMACOLOGY 2017; 202:20-27. [PMID: 27416805 DOI: 10.1016/j.jep.2016.07.028] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 07/07/2016] [Accepted: 07/08/2016] [Indexed: 06/06/2023]
Abstract
ETHNOPHAMACOLOGICAL RELEVANCE Pogostemon cablin is a medicinal herb widely used to treat gastrointestinal diseases in many Asian countries. Pogostone is an important constituent of Pogostemon cablin, and possesses various bioactivitys. In this study, we performed to investigate the anti-colorectal tumor property of Pogostone by inducing aurophagy and apoptosis in human colorectal cancer cells, and to define the potential molecular mechanisms. MATERIALS AND METHODS In vitro, The anti-tumor activity of pogostone was assessed using MTT assay. Autophagy was monitored by transmission electron microscopy observation and mRFP-GFP-LC3 fluorescence analysis in colorectal tumor cell line. Apoptosis was measured by flow cytometry and annexinV-FITC/PI staining. The protein expressions or activition of LC3-Ⅱ, AKT, mTOR, caspase-3 and caspase-7 were detected through western blotting. In vivo, the anti-tumor effect of pogostone was tested with HCT116 colorectal tumor cells transplantation tumor model. The expression of Ki-67 was determined by Immunohistochemistry staining and the apoptosis was evaluated using TUNEL assay. RESULTS In vitro, pogostone exhibits significant anti-tumor activity against human cancer cell lines, especially for HCT116 (18.7±1.93μg/ml). Transmission electron microscopy observation, mRFP-GFP-LC3 fluorescence analysis, flow cytometry and assay and western blotting detection revealed that the anti-colorectal tumor activity of pogostone was dependent on inducing autophagy and apoptosis through up-regulating the expression of LC3-Ⅱ, cleaved caspase-7 and caspase-3, and decreasing the phosphorylation of AKT/mTOR. In vivo, 150mg/kg pogostone inhibited the HCT116 tumor growth in immunodeficient mice with an inhibitory rate of 43.3%, decreased the expression of Ki67, and induced apoptosis in three days. CONCLUSION Pogostone showed anti-colorectal tumor effects by inducing autophagy and apoptosis involving PI3K/Akt/mTOR axis. Thus, pogostone may be a promising lead compound to be further developed for cancer therapy.
Collapse
Affiliation(s)
- Zhi-Xing Cao
- Pharmacy College, Chengdu University of Traditional Chinese Medicine, The Ministry of Education Key Laboratory of Standardization of Chinese Herbal Medicine, Key Laboratory of Systematic Research, Development and Utilization of Chinese Medicine Resources in Sichuan Province, Key Laboratory Breeding Base of Co-founded by Sichuan Province and MOST, PR China
| | - Yu-Ting Yang
- Pharmacy College, Chengdu University of Traditional Chinese Medicine, The Ministry of Education Key Laboratory of Standardization of Chinese Herbal Medicine, Key Laboratory of Systematic Research, Development and Utilization of Chinese Medicine Resources in Sichuan Province, Key Laboratory Breeding Base of Co-founded by Sichuan Province and MOST, PR China
| | - Si Yu
- Pharmacy College, Chengdu University of Traditional Chinese Medicine, The Ministry of Education Key Laboratory of Standardization of Chinese Herbal Medicine, Key Laboratory of Systematic Research, Development and Utilization of Chinese Medicine Resources in Sichuan Province, Key Laboratory Breeding Base of Co-founded by Sichuan Province and MOST, PR China
| | - Yu-Zhi Li
- Pharmacy College, Chengdu University of Traditional Chinese Medicine, The Ministry of Education Key Laboratory of Standardization of Chinese Herbal Medicine, Key Laboratory of Systematic Research, Development and Utilization of Chinese Medicine Resources in Sichuan Province, Key Laboratory Breeding Base of Co-founded by Sichuan Province and MOST, PR China
| | - Wen-Wen Wang
- Pharmacy College, Chengdu University of Traditional Chinese Medicine, The Ministry of Education Key Laboratory of Standardization of Chinese Herbal Medicine, Key Laboratory of Systematic Research, Development and Utilization of Chinese Medicine Resources in Sichuan Province, Key Laboratory Breeding Base of Co-founded by Sichuan Province and MOST, PR China; School of Medical Technology, Chengdu University of Traditional Chinese Medicine, PR China
| | - Jing Huang
- Pharmacy College, Chengdu University of Traditional Chinese Medicine, The Ministry of Education Key Laboratory of Standardization of Chinese Herbal Medicine, Key Laboratory of Systematic Research, Development and Utilization of Chinese Medicine Resources in Sichuan Province, Key Laboratory Breeding Base of Co-founded by Sichuan Province and MOST, PR China; School of Medical Technology, Chengdu University of Traditional Chinese Medicine, PR China
| | - Xiao-Fang Xie
- Pharmacy College, Chengdu University of Traditional Chinese Medicine, The Ministry of Education Key Laboratory of Standardization of Chinese Herbal Medicine, Key Laboratory of Systematic Research, Development and Utilization of Chinese Medicine Resources in Sichuan Province, Key Laboratory Breeding Base of Co-founded by Sichuan Province and MOST, PR China
| | - Liang Xiong
- Pharmacy College, Chengdu University of Traditional Chinese Medicine, The Ministry of Education Key Laboratory of Standardization of Chinese Herbal Medicine, Key Laboratory of Systematic Research, Development and Utilization of Chinese Medicine Resources in Sichuan Province, Key Laboratory Breeding Base of Co-founded by Sichuan Province and MOST, PR China
| | - Song Lei
- Department of pathology, West China Hospital, PR China
| | - Cheng Peng
- Pharmacy College, Chengdu University of Traditional Chinese Medicine, The Ministry of Education Key Laboratory of Standardization of Chinese Herbal Medicine, Key Laboratory of Systematic Research, Development and Utilization of Chinese Medicine Resources in Sichuan Province, Key Laboratory Breeding Base of Co-founded by Sichuan Province and MOST, PR China.
| |
Collapse
|
34
|
Liu YH, Weng YP, Lin HY, Tang SW, Chen CJ, Liang CJ, Ku CY, Lin JY. Aqueous extract of Polygonum bistorta modulates proteostasis by ROS-induced ER stress in human hepatoma cells. Sci Rep 2017; 7:41437. [PMID: 28134285 PMCID: PMC5278379 DOI: 10.1038/srep41437] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 12/16/2016] [Indexed: 01/16/2023] Open
Abstract
Hepatocellular carcinoma (HCC) remains the leading cause of cancer mortality with limited therapeutic targets. The endoplasmic reticulum (ER) plays a pivotal role in maintaining proteostasis in normal cells. However, alterations in proteostasis are often found in cancer cells, making it a potential target for therapy. Polygonum bistorta is used in traditional Chinese medicine owing to its anticancer activities, but the molecular and pharmacological mechanisms remain unclear. Using hepatoma cells as a model system, this study demonstrated that P. bistorta aqueous extract (PB) stimulated ER stress by increasing autophagosomes but by blocking degradation, followed by the accumulation of ubiquitinated proteins and cell apoptosis. In addition, an autophagy inhibitor did not enhance ubiquitinated protein accumulation whereas a reactive oxygen species (ROS) scavenger diminished both ubiquitinated protein accumulation and ligand-stimulated epidermal growth factor receptor (EGFR) expression, suggesting that ROS generation by PB may be upstream of PB-triggered cell death. Nevertheless, PB-exerted proteostasis impairment resulted in cytoskeletal changes, impairment of cell adhesion and motility, and inhibition of cell cycle progression. Oral administration of PB delayed tumour growth in a xenograft model without significant body weight loss. These findings indicate that PB may be a potential new alternative or complementary medicine for HCC.
Collapse
Affiliation(s)
- Yu-Huei Liu
- Graduate Institute of Integrated Medicine, China Medical University, Taichung, 404, Taiwan.,Department of Medical Genetics and Medical Research, China Medical University Hospital, Taichung, 404, Taiwan
| | - Yui-Ping Weng
- Graduate Institute of Biological Science and Technology, Chung Hwa University of Medical Technology, Tainan, 717, Taiwan.,Department of Biological Science and Technology, Chung Hwa University of Medical Technology, Tainan, 717, Taiwan
| | - Hsuan-Yuan Lin
- Department of Life Science, National Taiwan Normal University, Taipei, 116, Taiwan
| | - Sai-Wen Tang
- Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, 100, Taiwan
| | - Chao-Jung Chen
- Graduate Institute of Integrated Medicine, China Medical University, Taichung, 404, Taiwan
| | - Chi-Jung Liang
- Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, 100, Taiwan
| | - Chung-Yu Ku
- Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, 100, Taiwan
| | - Jung-Yaw Lin
- Department of Life Science, National Taiwan Normal University, Taipei, 116, Taiwan.,Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, 100, Taiwan
| |
Collapse
|
35
|
Hu B, An HM, Wang SS, Chen JJ, Xu L. Preventive and Therapeutic Effects of Chinese Herbal Compounds against Hepatocellular Carcinoma. Molecules 2016; 21:142. [PMID: 26828466 PMCID: PMC6274246 DOI: 10.3390/molecules21020142] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 01/13/2016] [Accepted: 01/20/2016] [Indexed: 12/14/2022] Open
Abstract
Traditional Chinese Medicines, unique biomedical and pharmaceutical resources, have been widely used for hepatocellular carcinoma (HCC) prevention and treatment. Accumulated Chinese herb-derived compounds with significant anti-cancer effects against HCC have been identified. Chinese herbal compounds are effective in preventing carcinogenesis, inhibiting cell proliferation, arresting cell cycle, inducing apoptosis, autophagy, cell senescence and anoikis, inhibiting epithelial-mesenchymal transition, metastasis and angiogenesis, regulating immune function, reversing drug resistance and enhancing the effects of chemotherapy in HCC. This paper comprehensively reviews these compounds and their effects on HCC. Finally, the perspectives and rational application of herbal compounds for HCC management are discussed.
Collapse
Affiliation(s)
- Bing Hu
- Department of Oncology and Institute of Traditional Chinese Medicine in Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| | - Hong-Mei An
- Department of Science & Technology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 202032, China.
| | - Shuang-Shuang Wang
- Department of Oncology and Institute of Traditional Chinese Medicine in Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| | - Jin-Jun Chen
- Department of Plastic & Reconstructive Surgery, Shanghai Key Laboratory of Tissue Engineering, The Ninth People's Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200011, China.
| | - Ling Xu
- Department of Oncology and Institute of Traditional Chinese Medicine in Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| |
Collapse
|
36
|
Lv Y, Xue L, Cai C, Liu QH, Shen J. Deficiency of myotubularin-related protein 14 influences body weight, metabolism, and inflammation in an age-dependent manner. Cell Biosci 2015; 5:69. [PMID: 26697164 PMCID: PMC4687302 DOI: 10.1186/s13578-015-0062-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 12/14/2015] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Myotubularin-related protein 14 (MTMR14) is a novel phosphoinositide phosphatase with roles in the maintenance of normal muscle performance, autophagy, and aging in mice. Our initial pilot study demonstrated that MTMR14 knock out (KO) mice gain weight earlier than their wild-type (WT) littermates, which suggests that this gene may also be involved in metabolism regulation. RESULTS The present study evaluated the role of MTMR14 in the development of aging-associated obesity. We found that aged MTMR14 KO mice fed a normal chow diet exhibited increased serum triglyceride, total cholesterol, and glucose levels compared to age-matched WT controls. Lipid accumulation was also increased in aged KO mice. Several inflammatory cytokines and adipokines were dramatically dysregulated in the metabolic tissues of aged MTMR14 KO mice compared to control mice. Circulating inflammatory cytokines were significantly elevated and plasma adipokine levels were abnormally regulated in aged MTMR14 KO mice. These data suggest that MTMR14 deficiency caused a late-onset inflammation and metabolic dysfunction. Further study demonstrated that this exacerbated metabolic dysfunction and inflammation may be regulated by the phosphoinositide 3 kinase/protein kinase B and extracellular signal-regulated protein kinase signaling pathways. CONCLUSIONS Our current research suggests that MTMR14 deletion induces overweight and adult obesity accompanied by chronic inflammation in an age-dependent manner.
Collapse
Affiliation(s)
- Yin Lv
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, 182 MinZu Ave, Wuhan, 430074 Hubei China
| | - Lu Xue
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, 182 MinZu Ave, Wuhan, 430074 Hubei China
| | - Congli Cai
- Wuhan Youzhiyou Biopharmaceutical Co., Ltd., Wuhan, 430075 China
| | - Qing-Hua Liu
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, 182 MinZu Ave, Wuhan, 430074 Hubei China
| | - Jinhua Shen
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, 182 MinZu Ave, Wuhan, 430074 Hubei China
| |
Collapse
|
37
|
Zhang FY, Hu Y, Que ZY, Wang P, Liu YH, Wang ZH, Xue YX. Shikonin Inhibits the Migration and Invasion of Human Glioblastoma Cells by Targeting Phosphorylated β-Catenin and Phosphorylated PI3K/Akt: A Potential Mechanism for the Anti-Glioma Efficacy of a Traditional Chinese Herbal Medicine. Int J Mol Sci 2015; 16:23823-48. [PMID: 26473829 PMCID: PMC4632727 DOI: 10.3390/ijms161023823] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 08/20/2015] [Accepted: 09/16/2015] [Indexed: 12/26/2022] Open
Abstract
Shikonin is an anthraquinone derivative extracted from the root of lithospermum. Shikonin is traditionally used in the treatment of inflammatory and infectious diseases such as hepatitis. Shikonin also inhibits proliferation and induces apoptosis in various tumors. However, the effect of shikonin on gliomas has not been fully elucidated. In the present study, we aimed to investigate the effects of shikonin on the migration and invasion of human glioblastoma cells as well as the underlying mechanisms. U87 and U251 human glioblastoma cells were treated with shikonin at 2.5, 5, and 7.5 μmol/L and cell viability, migration and invasiveness were assessed with CCK8, scratch wound healing, in vitro Transwell migration, and invasion assays. The expression and activity of matrix metalloproteinase-2 (MMP-2) and matrix metalloproteinase-9 (MMP-9) and the expression of phosphorylated β-catenin (p-β-catenin) and phosphorylated PI3K/Akt were also checked. Results showed that shikonin significantly inhibited the cell proliferation, migration, invasion, and expression of MMP-2 and MMP-9 in U87 and U251 cells. The expression of p-β-catenin showed contrary trends in two cell lines. It was significantly inhibited in U87 cells and promoted in U251 cells. Results in this work indicated that shikonin displayed an inhibitory effect on the migration and invasion of glioma cells by inhibiting the expression and activity of MMP-2 and -9. In addition, shikonin also inhibited the expression of p-PI3K and p-Akt to attenuate cell migration and invasion and MMP-2 and MMP-9 expression in both cell lines, which could be reversed by the PI3K/Akt pathway agonist, insulin-like growth factor-1 (IGF-1).
Collapse
Affiliation(s)
- Feng-Ying Zhang
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang 110122, China.
- Institute of Pathology and Pathophysiology, China Medical University, Shenyang 110122, China.
| | - Yi Hu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Zhong-You Que
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Ping Wang
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang 110122, China.
- Institute of Pathology and Pathophysiology, China Medical University, Shenyang 110122, China.
| | - Yun-Hui Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Zhen-Hua Wang
- Department of Physiology, College of Basic Medicine, China Medical University, Shenyang 110122, China.
| | - Yi-Xue Xue
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang 110122, China.
- Institute of Pathology and Pathophysiology, China Medical University, Shenyang 110122, China.
| |
Collapse
|
38
|
Surface chemistry but not aspect ratio mediates the biological toxicity of gold nanorods in vitro and in vivo. Sci Rep 2015; 5:11398. [PMID: 26096816 PMCID: PMC4476041 DOI: 10.1038/srep11398] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 05/22/2015] [Indexed: 12/14/2022] Open
Abstract
Gold nanorods are a promising nanoscale material in clinical diagnosis and treatment. The physicochemical properties of GNRs, including size, shape and surface features, are crucial factors affecting their cytotoxicity. In this study, we investigated the effects of different aspect ratios and surface modifications on the cytotoxicity and cellular uptake of GNRs in cultured cells and in mice. The results indicated that the surface chemistry but not the aspect ratio of GNRs mediates their biological toxicity. CTAB-GNRs with various aspect ratios had similar abilities to induce cell apoptosis and autophagy by damaging mitochondria and activating intracellular reactive oxygen species (ROS). However, GNRs coated with CTAB/PSS, CTAB/PAH, CTAB/PSS/PAH or CTAB/PAH/PSS displayed low toxicity and did not induce cell death. CTAB/PAH-coated GNRs caused minimally abnormal cell morphology compared with CTAB/PSS and CTAB/PSS/PAH coated GNRs. Moreover, the intravenous injection of CTAB/PAH GNRs enabled the GNRs to reach tumor tissues through blood circulation in animals and remained stable, with a longer half-life compared to the other GNRs. Therefore, our results demonstrated that further coating can prevent cytotoxicity and cell death upon CTAB-coated GNR administration, similar to changing the GNR aspect ratio and CTAB/PAH coated GNRs show superior biological properties with better biocompatibility and minimal cytotoxicity.
Collapse
|
39
|
Hu B, Wang SS, Du Q. Traditional Chinese medicine for prevention and treatment of hepatocarcinoma: From bench to bedside. World J Hepatol 2015; 7:1209-1232. [PMID: 26019736 PMCID: PMC4438495 DOI: 10.4254/wjh.v7.i9.1209] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 11/29/2014] [Accepted: 12/19/2014] [Indexed: 02/06/2023] Open
Abstract
Traditional Chinese medicine (TCM) has played a positive role in the management of hepatocarcinoma. Hepatocarcinoma patients may present Qi-stagnation, damp-heat, blood stasis, Qi-deficiency, Yin-deficiency and other TCM syndromes (Zheng). Modern treatments such as surgery, transarterial chemoembolization (TACE) and high intensity focus ultrasound treatment would influence the manifestation of TCM syndromes. Herbs with traditional efficacy of tonifying Qi, blood and Yin, soothing liver-Qi stagnation, clearing heat and detoxifying and dissolving stasis, have been demonstrated to be potent to prevent hepatocarcinogenesis. TCM has been widely used in all aspects of integrative therapy in hepatocarcinoma, including surgical resection, liver transplantation, TACE, local ablative therapies and even as monotherapy for middle-advanced stage hepatocarcinoma. Clinical practices have confirmed that TCM is effective to alleviate clinical symptoms, improve quality of life and immune function, prevent recurrence and metastasis, delay tumor progression, and prolong survival time in hepatocarcinoma patients. The effective mechanism of TCM against hepatocarcinoma is related to inducing apoptosis, autophagy, anoikis and cell senescence, arresting cell cycle, regulating immune function, inhibiting metastasis and angiogenesis, reversing drug resistance and enhancing effects of chemotherapy. Along with the progress of research in this field, TCM will contribute more to the prevention and treatment of hepatocarcinoma.
Collapse
|
40
|
Prieto P, Rosales-Mendoza CE, Terrón V, Toledano V, Cuadrado A, López-Collazo E, Bannenberg G, Martín-Sanz P, Fernández-Velasco M, Boscá L. Activation of autophagy in macrophages by pro-resolving lipid mediators. Autophagy 2015; 11:1729-1744. [PMID: 26506892 PMCID: PMC4824594 DOI: 10.1080/15548627.2015.1078958] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 07/13/2015] [Accepted: 07/28/2015] [Indexed: 02/07/2023] Open
Abstract
The resolution of inflammation is an active process driven by specialized pro-resolving lipid mediators, such as 15-epi-LXA4 and resolvin D1 (RvD1), that promote tissue regeneration. Macrophages regulate the innate immune response being key players during the resolution phase to avoid chronic inflammatory pathologies. Their half-life is tightly regulated to accomplish its phagocytic function, allowing the complete cleaning of the affected area. The balance between apoptosis and autophagy appears to be essential to control the survival of these immune cells within the inflammatory context. In the present work, we demonstrate that 15-epi-LXA4 and RvD1 at nanomolar concentrations promote autophagy in murine and human macrophages. Both compounds induced the MAP1LC3-I to MAP1LC3-II processing and the degradation of SQSTM1 as well as the formation of MAP1LC3(+) autophagosomes, a typical signature of autophagy. Furthermore, 15-epi-LXA4 and RvD1 treatment favored the fusion of the autophagosomes with lysosomes, allowing the final processing of the autophagic vesicles. This autophagic response involves the activation of MAPK1 and NFE2L2 pathways, but by an MTOR-independent mechanism. Moreover, these pro-resolving lipids improved the phagocytic activity of macrophages via NFE2L2. Therefore, 15-epi-LXA4 and RvD1 improved both survival and functionality of macrophages, which likely supports the recovery of tissue homeostasis and avoiding chronic inflammatory diseases.
Collapse
Affiliation(s)
- Patricia Prieto
- Instituto de Investigaciones Biomédicas Alberto Sols (Centro Mixto CSIC-UAM); Madrid, Spain
| | - César Eduardo Rosales-Mendoza
- Instituto de Investigaciones Biomédicas Alberto Sols (Centro Mixto CSIC-UAM); Madrid, Spain
- Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina; Universidad Autónoma de Nuevo León; Francisco I Madero y Eduardo Aguirre Pequeño S/N, Colonia Mitras Centro; Monterrey, Nuevo León, México
| | - Verónica Terrón
- Instituto de Investigaciones Biomédicas Alberto Sols (Centro Mixto CSIC-UAM); Madrid, Spain
| | - Víctor Toledano
- Instituto de Investigación Hospital Universitario La Paz (IdiPAZ); Madrid, Spain
| | - Antonio Cuadrado
- Instituto de Investigaciones Biomédicas Alberto Sols (Centro Mixto CSIC-UAM); Madrid, Spain
- Instituto de Investigación Hospital Universitario La Paz (IdiPAZ); Madrid, Spain
| | | | - Gerard Bannenberg
- Instituto de Investigaciones Biomédicas Alberto Sols (Centro Mixto CSIC-UAM); Madrid, Spain
| | - Paloma Martín-Sanz
- Instituto de Investigaciones Biomédicas Alberto Sols (Centro Mixto CSIC-UAM); Madrid, Spain
| | | | - Lisardo Boscá
- Instituto de Investigaciones Biomédicas Alberto Sols (Centro Mixto CSIC-UAM); Madrid, Spain
| |
Collapse
|
41
|
Farooqi AA, Fayyaz S, Hou MF, Li KT, Tang JY, Chang HW. Reactive oxygen species and autophagy modulation in non-marine drugs and marine drugs. Mar Drugs 2014; 12:5408-24. [PMID: 25402829 PMCID: PMC4245538 DOI: 10.3390/md12115408] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 11/06/2014] [Accepted: 11/07/2014] [Indexed: 12/11/2022] Open
Abstract
It is becoming more understandable that an existing challenge for translational research is the development of pharmaceuticals that appropriately target reactive oxygen species (ROS)-mediated molecular networks in cancer cells. In line with this approach, there is an overwhelmingly increasing list of many non-marine drugs and marine drugs reported to be involved in inhibiting and suppressing cancer progression through ROS-mediated cell death. In this review, we describe the strategy of oxidative stress-based therapy and connect the ROS modulating effect to the regulation of apoptosis and autophagy. Finally, we focus on exploring the function and mechanism of cancer therapy by the autophagy modulators including inhibitors and inducers from non-marine drugs and marine drugs.
Collapse
Affiliation(s)
- Ammad Ahmad Farooqi
- Laboratory for Translational Oncology and Personalized Medicine, Rashid Latif Medical College, Lahore 54000, Pakistan; E-Mails: (A.A.F.); (S.F.)
| | - Sundas Fayyaz
- Laboratory for Translational Oncology and Personalized Medicine, Rashid Latif Medical College, Lahore 54000, Pakistan; E-Mails: (A.A.F.); (S.F.)
| | - Ming-Feng Hou
- Cancer Center, Kaohsiung Medical University Hospital; Kaohsiung Medical University, Kaohsiung 80708, Taiwan; E-Mail:
- Institute of Clinical Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 80145, Taiwan
| | - Kun-Tzu Li
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; E-Mail:
| | - Jen-Yang Tang
- Department of Radiation Oncology, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 80145, Taiwan
- Department of Radiation Oncology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Radiation Oncology, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
- Authors to whom correspondence should be addressed; E-Mails: (J.Y.T.); (H.W.C.); Tel.: +886-7291-1101 (ext. 8105) (J.Y.T.); +886-7312-1101 (ext. 2691) (H.W.C.); Fax: +886-7213-8400 (J.Y.T.); +886-7312-5339 (H.W.C.)
| | - Hsueh-Wei Chang
- Cancer Center, Kaohsiung Medical University Hospital; Kaohsiung Medical University, Kaohsiung 80708, Taiwan; E-Mail:
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; E-Mail:
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
- Research Center of Environmental Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Authors to whom correspondence should be addressed; E-Mails: (J.Y.T.); (H.W.C.); Tel.: +886-7291-1101 (ext. 8105) (J.Y.T.); +886-7312-1101 (ext. 2691) (H.W.C.); Fax: +886-7213-8400 (J.Y.T.); +886-7312-5339 (H.W.C.)
| |
Collapse
|