1
|
Conejo-García A, Jiménez-Martínez Y, Cámara R, Franco-Montalbán F, Peña-Martín J, Boulaiz H, Carrión MD. New substituted benzoxazine derivatives as potent inducers of membrane permeability and cell death. Bioorg Med Chem 2024; 111:117849. [PMID: 39068873 DOI: 10.1016/j.bmc.2024.117849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 07/17/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024]
Abstract
The search for new agents targeting different forms of cell death is an important research focus for developing new and potent antitumor therapies. As a contribution to this endeavor, we have designed and synthesized a series of new substituted 3,4-dihydro-2H-1,4-benzoxazine derivatives. These compounds have been evaluated for their efficacy against MCF-7 breast cancer and HCT-116 colon cancer cell lines. Overall, substituting this heterocycle led to improved antiproliferative activity compared to the unsubstituted derivative 1. The most active compounds, 2b and 4b, showed IC50 values of 2.27 and 3.26 μM against MCF-7 cells and 4.44 and 7.63 μM against HCT-116 cells, respectively. To investigate the mechanism of action of the target compounds, the inhibition profile of 8 kinases involved in cell signaling was studied highlighting residual activity on HER2 and JNK1 kinases. 2b and 4b showed a consistent binding mode to both receptor kinases, establishing significant interactions with known and catalytically important domains and residues. Compounds 2b and 4b exhibit potent cytotoxic activity by disrupting cell membrane permeability, likely triggering both inflammatory and non-inflammatory cell death mechanisms. This dual capability increases their versatility in the treatment of different stages or types of tumors, providing greater flexibility in clinical applications.
Collapse
Affiliation(s)
- Ana Conejo-García
- Department of Medicinal and Organic Chemistry and Excellence Research Unit of Chemistry Applied to Biomedicine and the Environment, Faculty of Pharmacy, Campus Cartuja s/n, University of Granada, 18071 Granada, Spain; Biosanitary Institute of Granada (ibs.GRANADA), SAS-University of Granada, Avenida de Madrid, 15, 18012 Granada, Spain
| | - Yaiza Jiménez-Martínez
- Department of Anatomy and Human Embryology, Faculty of Medicine, Avenida de la Investigación 11, University of Granada, 18016 Granada, Spain; Biopathology and Regenerative Medicine Institute (IBIMER), Avenida del Conocimiento s/n, 18016 Granada, Spain
| | - Rubén Cámara
- Department of Medicinal and Organic Chemistry and Excellence Research Unit of Chemistry Applied to Biomedicine and the Environment, Faculty of Pharmacy, Campus Cartuja s/n, University of Granada, 18071 Granada, Spain
| | - Francisco Franco-Montalbán
- Department of Medicinal and Organic Chemistry and Excellence Research Unit of Chemistry Applied to Biomedicine and the Environment, Faculty of Pharmacy, Campus Cartuja s/n, University of Granada, 18071 Granada, Spain
| | - Jesús Peña-Martín
- Department of Anatomy and Human Embryology, Faculty of Medicine, Avenida de la Investigación 11, University of Granada, 18016 Granada, Spain; Biopathology and Regenerative Medicine Institute (IBIMER), Avenida del Conocimiento s/n, 18016 Granada, Spain
| | - Houria Boulaiz
- Biosanitary Institute of Granada (ibs.GRANADA), SAS-University of Granada, Avenida de Madrid, 15, 18012 Granada, Spain; Department of Anatomy and Human Embryology, Faculty of Medicine, Avenida de la Investigación 11, University of Granada, 18016 Granada, Spain; Biopathology and Regenerative Medicine Institute (IBIMER), Avenida del Conocimiento s/n, 18016 Granada, Spain.
| | - M Dora Carrión
- Department of Medicinal and Organic Chemistry and Excellence Research Unit of Chemistry Applied to Biomedicine and the Environment, Faculty of Pharmacy, Campus Cartuja s/n, University of Granada, 18071 Granada, Spain; Biosanitary Institute of Granada (ibs.GRANADA), SAS-University of Granada, Avenida de Madrid, 15, 18012 Granada, Spain.
| |
Collapse
|
2
|
Yang X, Wei M, An Y, Liang Q, Nan J, Vijayalakshmi A, Wang Z. Vernodalin Triggers ROS-Mediated Apoptosis in TPC-1 Human PapillaryThyroid Cancer Cells via Suppression of the MAPKs Signaling Pathway. Comb Chem High Throughput Screen 2024; 27:2151-2158. [PMID: 39099452 DOI: 10.2174/0113862073286226240220092357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/28/2024] [Accepted: 02/02/2024] [Indexed: 08/06/2024]
Abstract
BACKGROUND Thyroid Cancer (TC) is an endocrine organ malignancy that has become more common in recent decades. Vernodalin (VN), a cytotoxic sesquiterpene, has been reported to exhibit anticancer properties against human breast and liver cancer cells. However, no study has explored the efficacy of VN with respect to its antiproliferative and apoptotic action on human Papillary Thyroid Cancer cells (PTC). OBJECTIVE The study intended to examine the antitumor and antiproliferative effects of VN and the apoptosis mechanisms underlying its action on TPC-1 human PTC cells. METHODS In this study, we examined the VN cell viability by MTT assay; performed ROS measurement by DCFH staining method, MMP identification by Rh-123 staining method, and apoptotic morphological assay by employing AO/EB and DAPI stain method, and further, p38 MAPK/ERK/JNK cell proliferation markers were determined by western blotting technique. RESULTS The findings showed that VN could inhibit the growth of PTC cells by increasing intracellular ROS, damaging MMP, and stimulating apoptosis in a concentration-dependent manner. The study demonstrated how VN inhibited TPC-1 cell viability by causing ROS-induced cell death via the MAPK signaling pathway. CONCLUSION VN may serve as an agonist to impact apoptosis in PTC cells. In human PTC, VN could play an effective role in chemotherapy. More studies pertaining to animal tumor models are needed to prove its anti-cancer effectiveness in vivo.
Collapse
Affiliation(s)
- Xijia Yang
- Department of General Surgery, Xi'an Gaoxin Hospital, Xi'an, 710000, China
| | - Meng Wei
- Dialysis Department of Nephrology Hospital, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yuan An
- Department of Head and Neck Surgery, Shaanxi Provincial Tumor Hospital, The Affiliated Hospital of Xi'an Jiaotong Univesity, Xi'an, 710061, P.R. China
| | - Qinlong Liang
- Department of Head and Neck Surgery, Shaanxi Provincial Tumor Hospital, The Affiliated Hospital of Xi'an Jiaotong Univesity, Xi'an, 710061, P.R. China
| | - Jing Nan
- Department of Head and Neck Surgery, Shaanxi Provincial Tumor Hospital, The Affiliated Hospital of Xi'an Jiaotong Univesity, Xi'an, 710061, P.R. China
| | - Annamalai Vijayalakshmi
- Galileovasan Offshore and Research And Development Pvt. Ltd, Nagapattinam, Tamil Nadu, 611002, India
| | - Zizhang Wang
- Department of Head and Neck Surgery, Shaanxi Provincial Tumor Hospital, The Affiliated Hospital of Xi'an Jiaotong Univesity, Xi'an, 710061, P.R. China
| |
Collapse
|
3
|
Choi EY, Han EJ, Jeon SJ, Lee SW, Moon JM, Jung SH, Jung JY. Piperlongumine Induces Apoptosis and Cytoprotective Autophagy via the MAPK Signaling Pathway in Human Oral Cancer Cells. Biomedicines 2023; 11:2442. [PMID: 37760883 PMCID: PMC10525218 DOI: 10.3390/biomedicines11092442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 08/27/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023] Open
Abstract
Oral cancer is a malignant tumor that primarily affects areas such as the lips, tongue, buccal mucosa, salivary gland, and gingiva and has a very high malignancy. Piperlongumine (PL), isolated from long pepper (Piper longum L.), is a natural alkaloid with pharmacological effects, such as anti-inflammatory and anti-atherosclerotic effects. The effect and mechanism of PL in oral cancer cell lines has not been explored. Therefore, this study aimed to investigate the mechanism of anticancer effects of PL in the human oral cancer cell lines MC-3 and HSC-4 in vitro. This study demonstrated that PL inhibits cell proliferation by inducing apoptosis and autophagy in human oral cancer cell lines, which was confirmed by the levels of apoptosis- and autophagy-related proteins through Western blotting. Moreover, the pharmacological blockade of autophagy activation by hydroxychloroquine (HCQ), an autophagy inhibitor, significantly improved PL-induced apoptosis in MC-3 cells, suggesting a cytoprotective effect. In addition, activation of the mitogen-activated protein kinase (MAPK) signaling pathway contributed to PL-induced apoptosis. Collectively, the study suggested that combining an autophagy inhibitor with PL treatment can exert effective anticancer properties in oral cancer cells by inducing apoptosis and cytoprotective autophagy via the JNK-mediated MAPK pathway.
Collapse
Affiliation(s)
- Eun-Young Choi
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan-gun 32439, Republic of Korea; (E.-Y.C.); (E.-J.H.); (S.-J.J.); (S.-W.L.); (J.-M.M.); (S.-H.J.)
| | - Eun-Ji Han
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan-gun 32439, Republic of Korea; (E.-Y.C.); (E.-J.H.); (S.-J.J.); (S.-W.L.); (J.-M.M.); (S.-H.J.)
| | - Su-Ji Jeon
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan-gun 32439, Republic of Korea; (E.-Y.C.); (E.-J.H.); (S.-J.J.); (S.-W.L.); (J.-M.M.); (S.-H.J.)
| | - Sang-Woo Lee
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan-gun 32439, Republic of Korea; (E.-Y.C.); (E.-J.H.); (S.-J.J.); (S.-W.L.); (J.-M.M.); (S.-H.J.)
| | - Jun-Mo Moon
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan-gun 32439, Republic of Korea; (E.-Y.C.); (E.-J.H.); (S.-J.J.); (S.-W.L.); (J.-M.M.); (S.-H.J.)
| | - Soo-Hyun Jung
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan-gun 32439, Republic of Korea; (E.-Y.C.); (E.-J.H.); (S.-J.J.); (S.-W.L.); (J.-M.M.); (S.-H.J.)
| | - Ji-Youn Jung
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan-gun 32439, Republic of Korea; (E.-Y.C.); (E.-J.H.); (S.-J.J.); (S.-W.L.); (J.-M.M.); (S.-H.J.)
- Research Institute for Natural Products, Kongju National University, Yesan-gun 32439, Republic of Korea
| |
Collapse
|
4
|
Patel K, Jain P, Rajput PK, Jangid AK, Solanki R, Kulhari H, Patel S. Human Serum Albumin-based Propulsive Piperlongumine-loaded Nanoparticles: Formulation development, characterization and anti-cancer study. Colloids Surf A Physicochem Eng Asp 2022. [DOI: 10.1016/j.colsurfa.2022.129738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
5
|
Kung FP, Lim YP, Chao WY, Zhang YS, Yu HI, Tai TS, Lu CH, Chen SH, Li YZ, Zhao PW, Yen YP, Lee YR. Piperlongumine, a Potent Anticancer Phytotherapeutic, Induces Cell Cycle Arrest and Apoptosis In Vitro and In Vivo through the ROS/Akt Pathway in Human Thyroid Cancer Cells. Cancers (Basel) 2021; 13:cancers13174266. [PMID: 34503074 PMCID: PMC8428232 DOI: 10.3390/cancers13174266] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/13/2021] [Accepted: 08/20/2021] [Indexed: 01/20/2023] Open
Abstract
Simple Summary There is no effective treatment currently available for patients with anaplastic, recurrent papillary, or follicular thyroid cancers. Reactive oxygen species (ROS) are believed to hold promise as a new therapeutic strategy for multiple human cancers. However, studies on ROS inducers for human thyroid cancer treatment are scarce. This study assesses the anticancer activity and the detailed downstream mechanisms of piperlongumine, a ROS inducer, in human thyroid cancer cells. We demonstrate that piperlongumine inhibits cell proliferation, regulates the cell cycle, and induces cellular apoptosis in various types of human thyroid cancer cells. The antihuman thyroid cancer activity of piperlongumine was through ROS induction, and it further suppressed the downstream Akt signaling pathway to elevate mitochondria-dependent apoptosis. A mouse xenograft study demonstrated that piperlongumine was safe and could inhibit tumorigenesis in vivo. The present study provides strong evidence that piperlongumine can be used as a therapeutic candidate for human thyroid cancers. Abstract Thyroid cancer (TC) is the most common endocrine malignancy, and its global incidence has steadily increased over the past 15 years. TC is broadly divided into well-differentiated, poorly differentiated, and undifferentiated types, depending on the histological and clinical parameters. Thus far, there are no effective treatments for undifferentiated thyroid cancers or advanced and recurrent cancer. Therefore, the development of an effective therapeutic is urgently needed for such patients. Piperlongumine (PL) is a naturally occurring small molecule derived from long pepper; it is selectively toxic to cancer cells by generating reactive oxygen species (ROS). In this study, we demonstrate the potential anticancer activity of PL in four TC cell lines. For this purpose, we cultured TC cell lines and analyzed the following parameters: Cell viability, colony formation, cell cycle, apoptosis, and cellular ROS induction. PL modulated the cell cycle, induced apoptosis, and suppressed tumorigenesis in TC cell lines in a dose- and time-dependent manner through ROS induction. Meanwhile, an intrinsic caspase-dependent apoptosis pathway was observed in the TC cells under PL treatment. The activation of Erk and the suppression of the Akt/mTOR pathways through ROS induction were seen in cells treated with PL. PL-mediated apoptosis in TC cells was through the ROS-Akt pathway. Finally, the anticancer effect and safety of PL were also demonstrated in vivo. Our findings indicate that PL exhibits antitumor activity and has the potential for use as a chemotherapeutic agent against TC. This is the first study to show the sensitivity of TC cell lines to PL.
Collapse
Affiliation(s)
- Fang-Ping Kung
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi 60002, Taiwan; (F.-P.K.); (H.-I.Y.); (T.-S.T.); (C.-H.L.); (Y.-P.Y.)
| | - Yun-Ping Lim
- Department of Pharmacy, College of Pharmacy, China Medical University, Taichung 406040, Taiwan;
- Department of Internal Medicine, China Medical University Hospital, Taichung 404332, Taiwan
- Department of Medical Research, China Medical University Hospital, Taichung 404332, Taiwan
| | - Wen-Ying Chao
- Department of Nursing, Min-Hwei College of Health Care Management, Tainan 73658, Taiwan;
| | - Yi-Sheng Zhang
- Department of Medical Research, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi 60002, Taiwan; (Y.-S.Z.); (S.-H.C.); (Y.-Z.L.); (P.-W.Z.)
| | - Hui-I Yu
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi 60002, Taiwan; (F.-P.K.); (H.-I.Y.); (T.-S.T.); (C.-H.L.); (Y.-P.Y.)
| | - Tsai-Sung Tai
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi 60002, Taiwan; (F.-P.K.); (H.-I.Y.); (T.-S.T.); (C.-H.L.); (Y.-P.Y.)
| | - Chieh-Hsiang Lu
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi 60002, Taiwan; (F.-P.K.); (H.-I.Y.); (T.-S.T.); (C.-H.L.); (Y.-P.Y.)
| | - Shu-Hsin Chen
- Department of Medical Research, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi 60002, Taiwan; (Y.-S.Z.); (S.-H.C.); (Y.-Z.L.); (P.-W.Z.)
| | - Yi-Zhen Li
- Department of Medical Research, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi 60002, Taiwan; (Y.-S.Z.); (S.-H.C.); (Y.-Z.L.); (P.-W.Z.)
| | - Pei-Wen Zhao
- Department of Medical Research, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi 60002, Taiwan; (Y.-S.Z.); (S.-H.C.); (Y.-Z.L.); (P.-W.Z.)
| | - Yu-Pei Yen
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi 60002, Taiwan; (F.-P.K.); (H.-I.Y.); (T.-S.T.); (C.-H.L.); (Y.-P.Y.)
| | - Ying-Ray Lee
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Correspondence:
| |
Collapse
|
6
|
Parama D, Rana V, Girisa S, Verma E, Daimary UD, Thakur KK, Kumar A, Kunnumakkara AB. The promising potential of piperlongumine as an emerging therapeutics for cancer. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2021; 2:323-354. [PMID: 36046754 PMCID: PMC9400693 DOI: 10.37349/etat.2021.00049] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 07/04/2021] [Indexed: 12/24/2022] Open
Abstract
In spite of the immense advancement in the diagnostic and treatment modalities, cancer continues to be one of the leading causes of mortality across the globe, responsible for the death of around 10 million patients every year. The foremost challenges faced in the treatment of this disease are chemoresistance, adverse effects of the drugs, and the high cost of treatment. Though scientific studies over the past few decades have foreseen and are focusing on the cancer-preventive and therapeutic potential of natural products and their underlying mechanism of action, many more of these agents are not still explored. Piperlongumine (PL), or piplartine, is one such alkaloid isolated from Piper longum Linn. which is shown to be safe and has significant potential in the prevention and therapy of cancer. Numerous shreds of evidence have established the ability of this alkaloid and its analogs and nanoformulations in modulating various complex molecular pathways such as phosphatidylinositol-3-kinase/protein kinase B /mammalian target of rapamycin, nuclear factor kappa-B, Janus kinases/signal transducer and activator of transcription 3, etc. and inhibit different hallmarks of cancer such as cell survival, proliferation, invasion, angiogenesis, epithelial-mesenchymal-transition, metastases, etc. In addition, PL was also shown to inhibit radioresistance and chemoresistance and sensitize the cancer cells to the standard chemotherapeutic agents. Therefore, this compound has high potential as a drug candidate for the prevention and treatment of different cancers. The current review briefly reiterates the anti-cancer properties of PL against different types of cancer, which permits further investigation by conducting clinical studies.
Collapse
Affiliation(s)
- Dey Parama
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India
| | - Varsha Rana
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India
| | - Sosmitha Girisa
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India
| | - Elika Verma
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India
| | - Uzini Devi Daimary
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India
| | - Krishan Kumar Thakur
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India
| | - Aviral Kumar
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India
| | - Ajaikumar B. Kunnumakkara
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India
| |
Collapse
|
7
|
Kulkarni MR, Lad NP, Khedkar VM, Gaikwad ND. Synthesis, in vitro cytotoxicity, and molecular docking study of novel 3,
4‐dihydroisoquinolin
‐1(
2
H
)‐one based piperlongumine analogues. J Heterocycl Chem 2021. [DOI: 10.1002/jhet.4264] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Mahesh R. Kulkarni
- Organic Chemistry Research Centre, Department of Chemistry K.R.T. Arts, B.H. Commerce and A.M. Science College Nashik India
| | - Nitin P. Lad
- Organic Chemistry Research Centre, Department of Chemistry K.R.T. Arts, B.H. Commerce and A.M. Science College Nashik India
| | | | - Nitin D. Gaikwad
- Organic Chemistry Research Centre, Department of Chemistry K.R.T. Arts, B.H. Commerce and A.M. Science College Nashik India
| |
Collapse
|
8
|
Bezerra DP. Piplartine (piperlongumine), oxidative stress, and use in cancer. Cancer 2021. [DOI: 10.1016/b978-0-12-819547-5.00037-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
9
|
Huang JR, Wang ST, Wei MN, Liu K, Fu JW, Xing ZH, Shi Z. Piperlongumine Alleviates Mouse Colitis and Colitis-Associated Colorectal Cancer. Front Pharmacol 2020; 11:586885. [PMID: 33343354 PMCID: PMC7748110 DOI: 10.3389/fphar.2020.586885] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 10/06/2020] [Indexed: 01/05/2023] Open
Abstract
Colorectal cancer is one of the most common and lethal cancers in the world. An important causative factor of colorectal cancer is ulcerative colitis. In this study, we investigated the therapeutic effects of piperlongumine (PL) on the dextran sulfate sodium (DSS)-induced acute colitis and azoxymethane (AOM)/DSS-induced colorectal cancer mouse models. Our results showed that PL could inhibit the inflammation of DSS-induced mouse colitis and reduce the number of large neoplasms (diameter >2 mm) of AOM/DSS-induced mouse colorectal cancer by downregulation of proinflammatory cytokines cyclooxygenase-2 and interleukin-6 and epithelial-mesenchymal transition-related factors, β-catenin, and snail expressions, but fail to improve the colitis symptoms and to decrease the incidence of colonic neoplasms and the number of small neoplasms (diameter <2 mm). These data suggested that PL might be an effective agent in treating colitis and colorectal cancer.
Collapse
Affiliation(s)
- Jia-Rong Huang
- Department of Cell Biology, Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, MOE Key Laboratory of Tumor Molecular Biology, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Sheng-Te Wang
- Department of Cell Biology, Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, MOE Key Laboratory of Tumor Molecular Biology, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Meng-Ning Wei
- Department of Cell Biology, Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, MOE Key Laboratory of Tumor Molecular Biology, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Kun Liu
- Department of Cell Biology, Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, MOE Key Laboratory of Tumor Molecular Biology, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Jing-Wen Fu
- Affiliated High School of South China Normal University, Guangzhou, China
| | - Zi-Hao Xing
- Department of Cell Biology, Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, MOE Key Laboratory of Tumor Molecular Biology, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Zhi Shi
- Department of Cell Biology, Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, MOE Key Laboratory of Tumor Molecular Biology, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou, China
| |
Collapse
|
10
|
Scaria B, Sood S, Raad C, Khanafer J, Jayachandiran R, Pupulin A, Grewal S, Okoko M, Arora M, Miles L, Pandey S. Natural Health Products (NHP's) and Natural Compounds as Therapeutic Agents for the Treatment of Cancer; Mechanisms of Anti-Cancer Activity of Natural Compounds and Overall Trends. Int J Mol Sci 2020; 21:E8480. [PMID: 33187200 PMCID: PMC7697102 DOI: 10.3390/ijms21228480] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/08/2020] [Accepted: 11/09/2020] [Indexed: 12/12/2022] Open
Abstract
Most cancer therapeutics, such as tubulin-targeting chemotherapy drugs, cause cytotoxic, non-selective effects. These harmful side-effects drastically reduce the cancer patient's quality of life. Recently, researchers have focused their efforts on studying natural health products (NHP's) which have demonstrated the ability to selectively target cancer cells in cellular and animal models. However, the major hurdle of clinical validation remains. NHP's warrant further clinical investigation as a therapeutic option since they exhibit low toxicity, while retaining a selective effect. Additionally, they can sensitize cancerous cells to chemotherapy, which enhances the efficacy of chemotherapeutic drugs, indicating that they can be utilized as supplemental therapy. An additional area for further research is the investigation of drug-drug interactions between NHP's and chemotherapeutics. The objectives of this review are to report the most recent results from the field of anticancer NHP research, and to highlight the most recent advancements in possible supplemental therapeutic options.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Siyaram Pandey
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, ON N9B 3P4, Canada; (B.S.); (S.S.); (C.R.); (J.K.); (R.J.); (A.P.); (S.G.); (M.O.); (M.A.); (L.M.)
| |
Collapse
|
11
|
Zhou J, Huang Z, Ni X, Lv C. Piperlongumine induces apoptosis and G 2/M phase arrest in human osteosarcoma cells by regulating ROS/PI3K/Akt pathway. Toxicol In Vitro 2020; 65:104775. [PMID: 31987842 DOI: 10.1016/j.tiv.2020.104775] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 01/11/2020] [Accepted: 01/15/2020] [Indexed: 10/25/2022]
Abstract
Previous research has reported that piperlongumine exerts antitumor properties on several types of tumor cells. However, its effect on osteosarcoma cells remains unknown. This study aimed to investigate the antitumor effects of piperlongumine on osteosarcoma cells (MG63 and U2OS cells) in vitro and determined the underlying mechanism. Cell viability was measured using MTT assay. Cell apoptosis was assessed via AO/EB staining and flow cytometry apoptosis as well as western blot analysis. Cell cycle distribution was detected by flow cytometric cell cycle and western blot analysis. In our research, we found that piperlongumine induced apoptosis and G2/M phase arrest of MG63 cells. Western blot analysis not only confirmed the above results, but also demonstrated that piperlongumine induced apoptosis of osteosarcoma cells by activating Caspase-9-dependent apoptotic pathway. Furthermore, we also found that piperlongumine significantly induced apoptosis and cell cycle arrest of osteosarcoma cells by regulating ROS/PI3K/Akt signaling pathway. In summary, our findings suggested that piperlongumine inhibited osteosarcoma progression by promoting apoptosis of osteosarcoma cells. In addition, the underlying mechanism demonstrated that piperlongumine produced potent antitumor properties in osteosarcoma cells by regulating ROS/PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Jinfeng Zhou
- Department of Orthopedics and Traumatology, the Affiliated Wenzhou Traditional Chinese Medicine Hospital, Zhejiang Chinese Medical University, Wenzhou 325000, Zhejiang, China
| | - Zhengxiang Huang
- Department of Orthopedics, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Xiao Ni
- Department of Orthopedics, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Chen Lv
- Department of Orthopedics, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China.
| |
Collapse
|
12
|
Tripathi SK, Biswal BK. Piperlongumine, a potent anticancer phytotherapeutic: Perspectives on contemporary status and future possibilities as an anticancer agent. Pharmacol Res 2020; 156:104772. [PMID: 32283222 DOI: 10.1016/j.phrs.2020.104772] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 03/19/2020] [Accepted: 03/20/2020] [Indexed: 12/13/2022]
Abstract
Piperlongumine, a white to beige biologically active alkaloid/amide phytochemical, has high pharmacological relevance as an anticancer agent. Piperlongumine has several biological activities, including selective cytotoxicity against multiple cancer cells of different origins at a preclinical level. Several preclinical studies have documented the anticancer potential of piperlongumine through its targeting of multiple molecular mechanisms, such as cell cycle arrest, anti-angiogenesis, anti- invasive and anti-metastasis pathways, autophagy pathways, and intrinsic apoptotic pathways in vitro and in vivo. Mechanistically, piperlongumine inhibits cancer growth by resulting in the accumulation of intracellular reactive oxygen species, decreasing glutathione and chromosomal damage, or modulating key regulatory proteins, including PI3K, AKT, mTOR, NF-kβ, STATs, and cyclin D1. Furthermore, combined treatment with piperlongumine potentiates the anticancer activity of conventional chemotherapeutics and overcomes resistance to chemo- and radio- therapy. Nanoformulation of piperlongumine has been associated with increased aqueous solubility and bioavailability and lower toxicity, thus enhancing therapeutic efficacy in both preclinical and clinical settings. The current review highlights anticancer studies on the occurrence, chemical properties, chemopreventive mechanisms, toxicity, bioavailability, and pharmaceutical relevance of piperlongumine in vitro and in vivo.
Collapse
Affiliation(s)
- Surya Kant Tripathi
- Cancer Drug Resistance Laboratory, Department of Life Science, National Institute of Technology Rourkela, Odisha, 769008, India
| | - Bijesh Kumar Biswal
- Cancer Drug Resistance Laboratory, Department of Life Science, National Institute of Technology Rourkela, Odisha, 769008, India.
| |
Collapse
|
13
|
Cui WQ, Wang ST, Pan D, Chang B, Sang LX. Caffeine and its main targets of colorectal cancer. World J Gastrointest Oncol 2020; 12:149-172. [PMID: 32104547 PMCID: PMC7031145 DOI: 10.4251/wjgo.v12.i2.149] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 10/25/2019] [Accepted: 11/13/2019] [Indexed: 02/05/2023] Open
Abstract
Caffeine is a purine alkaloid and is widely consumed in coffee, soda, tea, chocolate and energy drinks. To date, a growing number of studies have indicated that caffeine is associated with many diseases including colorectal cancer. Caffeine exerts its biological activity through binding to adenosine receptors, inhibiting phosphodiesterases, sensitizing calcium channels, antagonizing gamma-aminobutyric acid receptors and stimulating adrenal hormones. Some studies have indicated that caffeine can interact with signaling pathways such as transforming growth factor β, phosphoinositide-3-kinase/AKT/mammalian target of rapamycin and mitogen-activated protein kinase pathways through which caffeine can play an important role in colorectal cancer pathogenesis, metastasis and prognosis. Moreover, caffeine can act as a general antioxidant that protects cells from oxidative stress and also as a regulatory factor of the cell cycle that modulates the DNA repair system. Additionally, as for intestinal homeostasis, through the interaction with receptors and cytokines, caffeine can modulate the immune system mediating its effects on T lymphocytes, B lymphocytes, natural killer cells and macrophages. Furthermore, caffeine can not only directly inhibit species in the gut microbiome, such as Escherichia coli and Candida albicans but also can indirectly exert inhibition by increasing the effects of other antimicrobial drugs. This review summarizes the association between colorectal cancer and caffeine that is being currently studied.
Collapse
Affiliation(s)
- Wen-Qi Cui
- Department of Neurology, Shengjing Hospital, Affiliated Hospital of China Medical University, Shenyang 110004, Liaoning Province, China
- China Medical University 101K class 87, Shenyang 110001, Liaoning Province, China
| | - Shi-Tong Wang
- Department of Cardiovascular Ultrasound, First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
- China Medical University 101K class 87, Shenyang 110001, Liaoning Province, China
| | - Dan Pan
- Department of Geriatrics, First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| | - Bing Chang
- Department of Gastroenterology, First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| | - Li-Xuan Sang
- Department of Geriatrics, First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| |
Collapse
|
14
|
Mohammad J, Singh RR, Riggle C, Haugrud B, Abdalla MY, Reindl KM. JNK inhibition blocks piperlongumine-induced cell death and transcriptional activation of heme oxygenase-1 in pancreatic cancer cells. Apoptosis 2019; 24:730-744. [PMID: 31243599 PMCID: PMC6713602 DOI: 10.1007/s10495-019-01553-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Piperlongumine (PL) is an alkaloid that inhibits glutathione S-transferase pi 1 (GSTP1) activity, resulting in elevated reactive oxygen species (ROS) levels and cancer-selective cell death. We aimed to identify stress-associated molecular responses to PL treatment in pancreatic ductal adenocarcinoma (PDAC) cells. GSTP1 directly interacts with JNK, which is activated by oxidative stress and can lead to decreased cancer cell proliferation and cell death. Therefore, we hypothesized that JNK pathways are activated in response to PL treatment. Our results show PL causes dissociation of GSTP1 from JNK; robust JNK, c-Jun, and early ERK activation followed by suppression; increased expression of cleaved caspase-3 and cleaved PARP; and nuclear translocation of Nrf2 and c-Myc in PDAC cells. Gene expression analysis revealed PL caused a > 20-fold induction of heme oxygenase-1 (HO-1), which we hypothesized was a survival mechanism for PDAC cells under enhanced oxidative stress. HO-1 knockout resulted in enhanced PL-induced PDAC cell death under hypoxic conditions. Similarly, high concentrations of the HO-1 inhibitor, ZnPP (10 µM), sensitized PDAC cells to PL; however, lower concentrations ZnPP (10 nM) and high or low concentrations of SnPP both protected PDAC cells from PL-induced cell death. Interestingly, the JNK inhibitor significantly blocked PL-induced PDAC cell death, Nrf-2 nuclear translocation, and HMOX-1 mRNA expression. Collectively, the results demonstrate JNK signaling contributes to PL-induced PDAC cell death, and at the same time, activates Nrf-2 transcription of HMOX-1 as a compensatory survival mechanism. These results suggest that elevating oxidative stress (using PL) while at the same time impairing antioxidant capacity (inhibiting HO-1) may be an effective therapeutic approach for PDAC.
Collapse
Affiliation(s)
- Jiyan Mohammad
- Department of Biological Sciences, North Dakota State University, Fargo, ND, 58108, USA
| | - Rahul R Singh
- Department of Biological Sciences, North Dakota State University, Fargo, ND, 58108, USA
| | - Cody Riggle
- Department of Biological Sciences, North Dakota State University, Fargo, ND, 58108, USA
| | - Brandon Haugrud
- Department of Biological Sciences, North Dakota State University, Fargo, ND, 58108, USA
| | - Maher Y Abdalla
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Katie M Reindl
- Department of Biological Sciences, North Dakota State University, Fargo, ND, 58108, USA.
| |
Collapse
|
15
|
Piska K, Gunia-Krzyżak A, Koczurkiewicz P, Wójcik-Pszczoła K, Pękala E. Piperlongumine (piplartine) as a lead compound for anticancer agents - Synthesis and properties of analogues: A mini-review. Eur J Med Chem 2018; 156:13-20. [PMID: 30006159 DOI: 10.1016/j.ejmech.2018.06.057] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 06/22/2018] [Accepted: 06/23/2018] [Indexed: 10/28/2022]
Abstract
Piperlongumine, also known as piplartine, is an amide alkaloid of Piper longum L. (long piper), a medical plant known from Ayurvedic medicine. Although was discovered well over fifty years ago, its pharmacological properties have been uncovered in the past decade. In particular, piperlongumine has been most extensively studied as a potential anticancer agent. Piperlongumine has exhibited cytotoxicity against a broad spectrum of human cancer cell lines, as well as demonstrated antitumor activity in rodents. Piperlongumine has also been found to be a proapoptotic, anti-invasive, antiangiogenic agent and synergize with modern chemotherapeutic agents. Because of its clinical potential, several studies were undertaken to obtain piperlongumine analogues, which have exhibited more potent activity or more appropriate drug-like parameters. In this review, the synthesis of piperlongumine analogues and piperlongumine-based hybrid compounds, as well as their anticancer properties and the molecular basis for their activity are explored. General structure-activity relationship conclusions are drawn and directions for the future research are indicated.
Collapse
Affiliation(s)
- Kamil Piska
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Agnieszka Gunia-Krzyżak
- Department of Bioorganic Chemistry, Chair of Organic Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Paulina Koczurkiewicz
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland.
| | - Katarzyna Wójcik-Pszczoła
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Elżbieta Pękala
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| |
Collapse
|
16
|
D'Sousa Costa CO, Araujo Neto JH, Baliza IRS, Dias RB, Valverde LDF, Vidal MTA, Sales CBS, Rocha CAG, Moreira DRM, Soares MBP, Batista AA, Bezerra DP. Novel piplartine-containing ruthenium complexes: synthesis, cell growth inhibition, apoptosis induction and ROS production on HCT116 cells. Oncotarget 2017; 8:104367-104392. [PMID: 29262647 PMCID: PMC5732813 DOI: 10.18632/oncotarget.22248] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 09/29/2017] [Indexed: 12/13/2022] Open
Abstract
Piplartine (piperlongumine) is a plant-derived molecule that has been receiving intense interest due to its anticancer characteristics that target the oxidative stress. In the present paper, two novel piplartine-containing ruthenium complexes [Ru(piplartine)(dppf)(bipy)](PF6)2 (1) and [Ru(piplartine)(dppb)(bipy)](PF6)2 (2) were synthesized and investigated for their cellular and molecular responses on cancer cell lines. We found that both complexes are more potent than metal-free piplartine in a panel of cancer cell lines on monolayer cultures, as well in 3D model of cancer multicellular spheroids formed from human colon carcinoma HCT116 cells. Mechanistic studies uncovered that the complexes reduced the cell growth and caused phosphatidylserine externalization, internucleosomal DNA fragmentation, caspase-3 activation and loss of the mitochondrial transmembrane potential on HCT116 cells. Moreover, the pre-treatment with Z-VAD(OMe)-FMK, a pan-caspase inhibitor, reduced the complexes-induced apoptosis, indicating cell death by apoptosis through caspase-dependent and mitochondrial intrinsic pathways. Treatment with the complexes also caused a marked increase in the production of reactive oxygen species (ROS), including hydrogen peroxide, superoxide anion and nitric oxide, and decreased reduced glutathione levels. Application of N-acetyl-cysteine, an antioxidant, reduced the ROS levels and apoptosis induced by the complexes, indicating activation of ROS-mediated apoptosis pathway. RNA transcripts of several genes, including gene related to the cell cycle, apoptosis and oxidative stress, were regulated under treatment. However, the complexes failed to induce DNA intercalation. In conclusion, the complexes are more potent than piplartine against different cancer cell lines and are able to induce caspase-dependent and mitochondrial intrinsic apoptosis on HCT116 cells by ROS-mediated pathway.
Collapse
Affiliation(s)
- Cinara O D'Sousa Costa
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
| | - João H Araujo Neto
- Department of Chemistry, Federal University of São Carlos, São Carlos, São Paulo, 13561-901, Brazil
| | - Ingrid R S Baliza
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
| | - Rosane B Dias
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
| | - Ludmila de F Valverde
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
| | - Manuela T A Vidal
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
| | - Caroline B S Sales
- Department of Biomorphology, Institute of Health Sciences, Federal University of Bahia, Salvador, Bahia, 40110-902, Brazil
| | - Clarissa A G Rocha
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
| | - Diogo R M Moreira
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
| | - Milena B P Soares
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil.,Center of Biotechnology and Cell Therapy, Hospital São Rafael, Salvador, Bahia, 41253-190, Brazil
| | - Alzir A Batista
- Department of Chemistry, Federal University of São Carlos, São Carlos, São Paulo, 13561-901, Brazil
| | - Daniel P Bezerra
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
| |
Collapse
|
17
|
Sun J, Zhang X, Sun Y, Tang ZS, Guo DY. Effects of Hylomecon vernalis ethanol extracts on cell cycle and apoptosis of colon cancer cells. Mol Med Rep 2017; 15:3485-3492. [PMID: 28393197 PMCID: PMC5436294 DOI: 10.3892/mmr.2017.6426] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 01/19/2017] [Indexed: 11/29/2022] Open
Abstract
Hylomecon vernalis Maxim. has traditionally been used to promote blood circulation, alleviate pain, dissipate stasis, and reduce swelling. The aim of the present study was to investigate the effect and potential mechanism of H. vernalis Maxim. ethanol extracts (HVMEE) on the growth and apoptosis of human colon cancer HT-29 and SW620 cells. H. vernalis samples were extracted three times with ethanol, dried, and concentrated into powder. The components of HVMEE were investigated using high performance liquid chromatography in tandem with mass spectrometry analysis. MTT assay was used to investigate the effect of HVMEE on viability of human colon cancer HT-29 and SW620 cells. Apoptosis of HT-29 and SW620 cells was evaluated using flow cytometric analysis. Expression levels of apoptosis and cell cycle-related proteins were assessed by western blot. The findings demonstrated that the alkaloid content of HVMEE was as high as 89.67%, and it effectively inhibited viability in HT-29 and SW620 cells, with IC50 values of 0.105±0.022 mg/ml and 0.146±0.013 mg/ml, respectively. In addition, HVMEE induced apoptosis in HT-29 and SW620 cells, by increasing caspase-3, caspase-9 and BCL2 associated X expression, and reducing Bcl-2 expression. The present study suggests that HVMEE has a potential role in the treatment of colorectal cancer.
Collapse
Affiliation(s)
- Jing Sun
- College of Pharmacy, Shannxi University of Chinese Medicine, Xianyang, Shannxi 712046, P.R. China
| | - Xin Zhang
- College of Pharmacy, Shannxi University of Chinese Medicine, Xianyang, Shannxi 712046, P.R. China
| | - Yang Sun
- Department of Pharmacology, School of Pharmacy, The Fourth Military Medical University, Xi'an, Shannxi 710032, P.R. China
| | - Zhi-Shu Tang
- College of Pharmacy, Shannxi University of Chinese Medicine, Xianyang, Shannxi 712046, P.R. China
| | - Dong-Yan Guo
- College of Pharmacy, Shannxi University of Chinese Medicine, Xianyang, Shannxi 712046, P.R. China
| |
Collapse
|
18
|
Yang Y, Zhou JY, Zhao LJ, Gao BR, Wan XP, Wang JL. Dual-specificity Phosphatase 1 Deficiency Induces Endometrioid Adenocarcinoma Progression via Activation of Mitogen-activated Protein Kinase/Extracellular Signal-regulated Kinase Pathway. Chin Med J (Engl) 2017; 129:1154-60. [PMID: 27174322 PMCID: PMC4878159 DOI: 10.4103/0366-6999.181954] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background: Previously, we reported that dual-specificity phosphatase 1 (DUSP1) was differentially expressed in endometrioid adenocarcinoma (EEA). However, the role of DUSP1 in EEA progression and the relationship between DUSP1 and medroxyprogesterone (MPA) are still unclear. Methods: The expression of DUSP1 in EEA specimens was detected by immunohistochemical analysis. The effect of DUSP1 on cell proliferation was analyzed by Cell Counting Kit 8 and colony formation assay, and cell migration was analyzed by transwell assay. MPA-induced DUSP1 expression in EEA cells was measured by Western blot. Results: DUSP1 expression was deficient in advanced International Federation of Gynecology and Obstetrics stage, high-grade and myometrial invasive EEA. In EEA cell lines (Hec1A, Hec1B, RL952, and Ishikawa), the DUSP1 expression was substantially higher in Ishikawa cells than in other cell lines (P < 0.05). Knockdown of DUSP1 promoted Ishikawa cells proliferation, migration, and activation of mitogen-activated protein kinases/extracellular signal-regulated kinase (MAPK/Erk) pathway. MPA-induced DUSP1 expression and inhibited MAPK/Erk pathway in Ishikawa cells. Conclusions: Our data suggest that DUSP1 deficiency promotes EEA progression via MAPK/Erk pathway, which may be reversed by MPA, suggesting that DUSP1 may serve as a potential therapeutic target for the treatment of EEA.
Collapse
Affiliation(s)
- Yuan Yang
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing 100044, China
| | - Jing-Yi Zhou
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing 100044, China
| | - Li-Jun Zhao
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing 100044, China
| | - Bao-Rong Gao
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xiao-Ping Wan
- Department of Gynecology, Tongji University School of Medicine Affiliated Shanghai First Maternity and Infant Hospital, Shanghai 200126, China
| | - Jian-Liu Wang
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing 100044, China
| |
Collapse
|
19
|
Wang Y, Chang J, Liu X, Zhang X, Zhang S, Zhang X, Zhou D, Zheng G. Discovery of piperlongumine as a potential novel lead for the development of senolytic agents. Aging (Albany NY) 2016; 8:2915-2926. [PMID: 27913811 PMCID: PMC5191878 DOI: 10.18632/aging.101100] [Citation(s) in RCA: 166] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 11/04/2016] [Indexed: 04/16/2023]
Abstract
Accumulating evidence indicates that senescent cells play an important role in many age-associated diseases. The pharmacological depletion of senescent cells (SCs) with a "senolytic agent", a small molecule that selectively kills SCs, is a potential novel therapeutic approach for these diseases. Recently, we discovered ABT-263, a potent and highly selective senolytic agent, by screening a library of rationally-selected compounds. With this screening approach, we also identified a second senolytic agent called piperlongumine (PL). PL is a natural product that is reported to have many pharmacological effects, including anti-tumor activity. We show here that PL preferentially killed senescent human WI-38 fibroblasts when senescence was induced by ionizing radiation, replicative exhaustion, or ectopic expression of the oncogene Ras. PL killed SCs by inducing apoptosis, and this process did not require the induction of reactive oxygen species. In addition, we found that PL synergistically killed SCs in combination with ABT-263, and initial structural modifications to PL identified analogs with improved potency and/or selectivity in inducing SC death. Overall, our studies demonstrate that PL is a novel lead for developing senolytic agents.
Collapse
Affiliation(s)
- Yingying Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Jianhui Chang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Xingui Liu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Xuan Zhang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Suping Zhang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Xin Zhang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Daohong Zhou
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Guangrong Zheng
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| |
Collapse
|
20
|
The role of MAPK signaling pathway in the Her-2-positive meningiomas. Oncol Rep 2016; 36:685-95. [PMID: 27279438 PMCID: PMC4933551 DOI: 10.3892/or.2016.4849] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2016] [Accepted: 03/18/2016] [Indexed: 01/16/2023] Open
Abstract
Meningiomas are common types of adult nerve system tumors. Although most cases are considered benign, due to its high rate of recurrence and easy malignant progression to anaplastic meningioma they present a puzzle for the current treatment. The HER-2 oncogene has important value for meningioma cells development and progression. So far, little is known about the effect on the exact underlying signal pathway and molecular mechanisms of HER-2-positive meningioma cells. The goal of the present study was to determine the effects of HER-2 gene and possible involvement of MAPK signal pathway in human malignant meningioma. We applied q-PCR analysis, immunofluorescence (IF) staining, western blot analysis, animal model, MAPK inhibition, MTT assay and cell invasion analysis for the investigation. The results demonstrated that the downregulation of the expression of HER-2 significantly inhibited cell motility and proliferation of human meningioma cells in vivo. Accordingly, in the HER-2-overexpression meningioma cells with the inhibition of ERK1/2, ERK5, JNK, in the cells with the ERK1/2, ERK5 inhibition, protein expression was markedly suppressed as well as the cell proliferation resistance. No difference was observed in the HER-2-overexpression meningioma cells with the inhibition of JNK. These findings suggest that HER-2 gene can affect the proliferation ability of human meningioma cells in vivo and MAPK signal pathway may contribute to the carcinogenesis and development of human meningiomas combinating with HER-2.
Collapse
|
21
|
Dhillon H, Mamidi S, McClean P, Reindl KM. Transcriptome Analysis of Piperlongumine-Treated Human Pancreatic Cancer Cells Reveals Involvement of Oxidative Stress and Endoplasmic Reticulum Stress Pathways. J Med Food 2016; 19:578-85. [PMID: 27119744 PMCID: PMC4904158 DOI: 10.1089/jmf.2015.0152] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 03/14/2016] [Indexed: 12/22/2022] Open
Abstract
Piperlongumine (PL), an alkaloid obtained from long peppers, displays antitumorigenic properties for a variety of human cell- and animal-based models. The aim of this study was to identify the underlying molecular mechanisms for PL anticancer effects on human pancreatic cancer cells. RNA sequencing (RNA-seq) was used to identify the effects of PL on the transcriptome of MIA PaCa-2 human pancreatic cancer cells. PL treatment of pancreatic cancer cells resulted in differential expression of 683 mRNA transcripts with known protein functions, 351 of which were upregulated and 332 of which were downregulated compared to control-treated cells. Transcripts associated with oxidative stress, endoplasmic reticulum (ER) stress, and unfolded protein response pathways were significantly overexpressed with PL treatment. Reverse transcription-quantitative polymerase chain reaction and western blotting were used to validate the RNA-seq results, which included upregulation of HO-1, IRE1α, cytochrome c, and ASNS. The results provide key insight into the mechanisms by which PL alters cancer cell physiology and identify that activation of oxidative stress and ER stress pathways is a critical avenue for PL anticancer effects.
Collapse
Affiliation(s)
- Harsharan Dhillon
- Department of Biological Sciences, North Dakota State University, Fargo, North Dakota, USA
| | - Sujan Mamidi
- Department of Plant Sciences, North Dakota State University, Fargo, North Dakota, USA
| | - Phillip McClean
- Department of Plant Sciences, North Dakota State University, Fargo, North Dakota, USA
| | - Katie M. Reindl
- Department of Biological Sciences, North Dakota State University, Fargo, North Dakota, USA
| |
Collapse
|