1
|
Sirotkin AV, Harrath AH. Apigenin as a Promising Agent for Enhancing Female Reproductive Function and Treating Associated Disorders. Biomedicines 2024; 12:2405. [PMID: 39457717 PMCID: PMC11504338 DOI: 10.3390/biomedicines12102405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 10/13/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Apigenin is an organic flavonoid abundant in some plants such as parsley, chamomile, or celery. Recently, it has been investigated for several of its pharmacological characteristics, such as its ability to act as an antioxidant, reduce inflammation, and inhibit the growth of cancer cells. The purpose of this review is to provide a summary of the existing knowledge regarding the effects of apigenin on female reproductive systems and its dysfunctions. Apigenin can influence reproductive processes by regulating multiple biological events, including oxidative processes, cell proliferation, apoptosis, cell renewal and viability, ovarian blood supply, and the release of reproductive hormones. It could stimulate ovarian folliculogenesis, as well as ovarian and embryonal cell proliferation and viability, which can lead to an increase in fertility and influence the release of reproductive hormones, which may exert its effects on female reproductive health. Furthermore, apigenin could inhibit the activities of ovarian cancer cells and alleviate the pathological changes in the female reproductive system caused by environmental pollutants, harmful medications, cancer, polycystic ovarian syndrome, ischemia, as well as endometriosis. Therefore, apigenin may have potential as a biostimulator for female reproductive processes and as a therapeutic agent for certain reproductive diseases.
Collapse
Affiliation(s)
- Alexander V. Sirotkin
- Faculty of Natural Sciences, Constantine the Philosopher University in Nitra, 949 74 Nitra, Slovakia;
| | - Abdel Halim Harrath
- Zoology Department, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| |
Collapse
|
2
|
Ali ML, Roky AH, Azad SAK, Shaikat AH, Meem JN, Hoque E, Ahasan AMF, Islam MM, Arif MSR, Mostaq MS, Mahmud MZ, Amin MN, Mahmud MA. Autophagy as a targeted therapeutic approach for skin cancer: Evaluating natural and synthetic molecular interventions. CANCER PATHOGENESIS AND THERAPY 2024; 2:231-245. [PMID: 39371094 PMCID: PMC11447340 DOI: 10.1016/j.cpt.2024.01.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 01/22/2024] [Accepted: 01/28/2024] [Indexed: 10/08/2024]
Abstract
Skin cancer, a prevalent malignancy worldwide, poses significant health concerns owing to its increasing incidence. Autophagy, a natural cellular process, is a pivotal event in skin cancer and has advantageous and detrimental effects. This duality has prompted extensive investigations into medical interventions targeting autophagy modulation for their substantial therapeutic potential. This systematic review aimed to investigate the relationship between skin cancer and autophagy and the contribution and mechanism of autophagy modulators in skin cancer. We outlined the effectiveness and safety of targeting autophagy as a promising therapeutic strategy for the treatment of skin cancer. This comprehensive review identified a diverse array of autophagy modulators with promising potential for the treatment of skin cancer. Each of these compounds demonstrates efficacy through distinct physiological mechanisms that have been elucidated in detail. Interestingly, findings from a literature search indicated that none of the natural, synthetic, or semisynthetic compounds exhibited notable adverse effects in either human or animal models. Consequently, this review offers novel mechanistic and therapeutic perspectives on the targeted modulation of autophagy in skin cancer.
Collapse
Affiliation(s)
- Md. Liakot Ali
- Department of Pharmacy, University of Chittagong, Chattogram 4331, Bangladesh
- Pratyasha Health Biomedical Research Center, Dhaka 1230, Bangladesh
| | - Amdad Hossain Roky
- Pratyasha Health Biomedical Research Center, Dhaka 1230, Bangladesh
- Department of Pharmacy, International Islamic University Chittagong, Chattogram 4318, Bangladesh
| | - S.M. Asadul Karim Azad
- Pratyasha Health Biomedical Research Center, Dhaka 1230, Bangladesh
- Department of Pharmacy, International Islamic University Chittagong, Chattogram 4318, Bangladesh
| | - Abdul Halim Shaikat
- Pratyasha Health Biomedical Research Center, Dhaka 1230, Bangladesh
- Department of Pharmacy, International Islamic University Chittagong, Chattogram 4318, Bangladesh
| | - Jannatul Naima Meem
- Department of Pharmacy, University of Chittagong, Chattogram 4331, Bangladesh
- Pratyasha Health Biomedical Research Center, Dhaka 1230, Bangladesh
| | - Emtiajul Hoque
- Pratyasha Health Biomedical Research Center, Dhaka 1230, Bangladesh
- Department of Pharmacy, International Islamic University Chittagong, Chattogram 4318, Bangladesh
| | - Abu Mohammed Fuad Ahasan
- Pratyasha Health Biomedical Research Center, Dhaka 1230, Bangladesh
- Department of Pharmacy, International Islamic University Chittagong, Chattogram 4318, Bangladesh
| | - Mohammed Murshedul Islam
- Pratyasha Health Biomedical Research Center, Dhaka 1230, Bangladesh
- Department of Pharmacy, Daffodil International University, Dhaka 1216, Bangladesh
| | - Md. Saifur Rahaman Arif
- Pratyasha Health Biomedical Research Center, Dhaka 1230, Bangladesh
- Department of Pharmacy, BGC Trust University Bangladesh, Chattogram 4381, Bangladesh
| | - Md. Saqline Mostaq
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71209-0497, USA
| | | | - Mohammad Nurul Amin
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71209-0497, USA
| | - Md. Ashiq Mahmud
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71209-0497, USA
| |
Collapse
|
3
|
Salimizadeh Z, Enferadi ST, Majidizadeh T, Mahjoubi F. Cytotoxicity of alkaloids isolated from Peganum harmala seeds on HCT116 human colon cancer cells. Mol Biol Rep 2024; 51:732. [PMID: 38872006 DOI: 10.1007/s11033-024-09655-7] [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: 11/14/2023] [Accepted: 05/20/2024] [Indexed: 06/15/2024]
Abstract
BACKGROUND The present study aimed to elucidate the potential anticancer activity and mechanism of P. harmala's alkaloid extract, harmine (HAR), and harmaline (HAL) in HCT-116 colorectal cancer cells. METHODS AND RESULTS P. harmala's alkaloid was extracted from harmala seeds. HCT-116 cells were treated with P. harmala's alkaloid extract, HAR and HAL. Cytotoxicity was determined by MTT assay, apoptotic activity detected via flow cytometry and acridine orange (AO)/ethidium bromide (EB) dual staining, and cell cycle distribution analyzed with flow cytometry. The mRNA expression of Bcl-2-associated X protein (Bax) and glycogen synthase kinase-3 beta (GSK3β) was measured by real-time PCR. Furthermore, the expression of Bax, Bcl-2, GSK3β and p53 proteins, were determined by western blotting. The findings indicated that, P. harmala's alkaloids extract, HAR and HAL were significantly cytotoxic toward HCT116 cells after 24 and 48 h of treatment. We showed that P. harmala's alkaloid extract induce apoptosis and cell cycle arrest at G2 phase in the HCT116 cell line. Downregulation of GSK3β and Bcl-2 and upregulation of Bax and p53 were observed. CONCLUSION The findings of this study indicate that the P. harmala's alkaloid extract has anticancer activity and may be further investigated to develop future anticancer chemotherapeutic agents.
Collapse
Affiliation(s)
- Zahra Salimizadeh
- Department of Medical Genetic, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Sattar Tahmasebi Enferadi
- Department of Plant Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Tayebeh Majidizadeh
- Department of Medical Genetic, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Frouzandeh Mahjoubi
- Department of Medical Genetic, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran.
| |
Collapse
|
4
|
Zhu J, Zhu H, Zhu Q, Xu SL, Xiao L, Zhang MY, Gao J. The roles of autophagy, ferroptosis and pyroptosis in the anti-ovarian cancer mechanism of harmine and their crosstalk. Sci Rep 2024; 14:6504. [PMID: 38499622 PMCID: PMC10948856 DOI: 10.1038/s41598-024-57196-7] [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: 03/01/2023] [Accepted: 03/15/2024] [Indexed: 03/20/2024] Open
Abstract
This study aimed to investigate the role of autophagy, ferroptosis, and pyroptosis in the antitumour mechanism of harmine (Har) and its crosstalk in ovarian cancer. By transmission electron microscopy, we found that compared with those in the control group, the cytoplasm of human ovarian cancer cells (SKOV3) treated with Har showed increased numbers of autophagic vesicles, decreased intracellular mitochondrial volume, increased bilayer membrane density, and decreased cristae. Western blot, immunofluorescence, and monodasylcadaverine (MDC) staining all suggested that Har promoted autophagy in SKOV3 cells. LY294002 and siFOXO3 rescued the inhibition of the PI3K/AKT/mTOR/FOXO3 signalling pathway and the promotion of autophagy by Har. Additionally, the levels of ferroptosis- and pyroptosis-related proteins and the levels of Fe2+ , glutathione (GSH), malondialdehyde (MDA), and superoxide dismutase (SOD) suggested that Har promoted ferroptosis and pyroptosis in SKOV3 cells. Interestingly, pretreatment with chloroquine (CQ), erastin, rapamycin (Rap), or ferrostatin-1 (Fer-1) increased or reversed the ferroptosis and pyroptosis promoted by Har, respectively. In vivo, the volume of tumours in the Har group was decreased, and immunohistochemistry revealed decreased levels of Ki-67 and GPX4 and increased levels of ATG5 and NARL3. In conclusion, Har exerts its anti-ovarian cancer effect not only by promoting autophagy by regulating the PI3K/AKT/mTOR/FOXO3 signalling pathway but also by promoting ferroptosis and pyroptosis. Additionally, there is complex crosstalk between autophagy, ferroptosis, and pyroptosis in ovarian cancer.
Collapse
Affiliation(s)
- Jun Zhu
- Jiangxi Medical College, Nanchang University, Nanchang, 330036, Jiangxi, China
- Jiangxi Provincial Key Laboratory of Tumor Metastasis and Precision Therapy, Nanchang, Jiangxi, China
- Department of Gynecologic Oncology, Jiangxi Cancer Hospital, Nanchang, Jiangxi, China
- Nanchang Key Laboratory of Precision Therapy for Gynecological Oncology, Nanchang, Jiangxi, China
| | - Hong Zhu
- Department of Gynecologic Oncology, Jiangxi Cancer Hospital, Nanchang, Jiangxi, China
| | - Qing Zhu
- The 334 Hospital of Nanchang, Nanchang, Jiangxi, China
| | - Shi Lei Xu
- Jiangxi Medical College, Nanchang University, Nanchang, 330036, Jiangxi, China
| | - Lu Xiao
- Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Ming Yue Zhang
- Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Jun Gao
- Department of Gynecologic Oncology, Jiangxi Cancer Hospital, Nanchang, Jiangxi, China.
- Nanchang Key Laboratory of Precision Therapy for Gynecological Oncology, Nanchang, Jiangxi, China.
| |
Collapse
|
5
|
Fan R, Wang S, Wu Y, Feng Y, Gao M, Cao Y, Ma X, Xie S, Wang C, Gao L, Wang Y, Dai F. Activation of endoplasmic reticulum stress by harmine suppresses the growth of esophageal squamous cell carcinoma. Phytother Res 2023; 37:4655-4673. [PMID: 37525965 DOI: 10.1002/ptr.7933] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 05/16/2023] [Accepted: 06/21/2023] [Indexed: 08/02/2023]
Abstract
The worldwide overall 5-year survival rate of esophageal squamous cell carcinoma (ESCC) patients is less than 20%, and novel therapeutic strategies for these patients are urgently needed. Harmine is a natural β-carboline alkaloid, which received great interest in cancer research because of its biological and anti-tumor activities. The aim of this study is to examine the effects of harmine on ESCC and its mechanism. We investigated the effects of harmine on proliferation, cell cycle, apoptosis, and tumor growth in vivo. RNA sequencing (RNA-seq), real-time PCR, and western blotting were used to detect the mechanism. Harmine inhibited ESCC cell growth in vitro and tumor growth in vivo. Differentially expressed genes in harmine-treated ESCC cells were mainly involved in protein processing in the endoplasmic reticulum (ER). Real-time PCR and western blotting confirmed harmine-induced cellular ER stress. CRISPR-Cas9 knockout of C/EBP homologous protein (CHOP) abolished harmine-induced expression of death receptor 5 and apoptosis. Harmine also induced the expression of CHOP-mediated sestrin-2, which in turn contributes to autophagosome formation via suppressing the AMP-activated protein kinase-protein kinase B-mammalian target of rapamycin signaling pathway. In conclusion, our results demonstrate that harmine inhibits the growth of ESCC through its regulation of ER stress, suggesting that it is a promising candidate for ESCC treatment.
Collapse
Affiliation(s)
- Ronghui Fan
- Key Laboratory of Natural Medicine and Immuno-Engineering, Henan University, Kaifeng, Henan, China
- School of Life Sciences, Henan University, Kaifeng, Henan, China
| | - Senzhen Wang
- School of Life Sciences, Henan University, Kaifeng, Henan, China
| | - Yalan Wu
- School of Biomedical Sciences, Heart and Vascular Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
- Department of Histology and Embryology, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Yongli Feng
- Key Laboratory of Natural Medicine and Immuno-Engineering, Henan University, Kaifeng, Henan, China
| | - Mengke Gao
- Key Laboratory of Natural Medicine and Immuno-Engineering, Henan University, Kaifeng, Henan, China
| | - Yue Cao
- Key Laboratory of Natural Medicine and Immuno-Engineering, Henan University, Kaifeng, Henan, China
| | - Xiaoxuan Ma
- Key Laboratory of Natural Medicine and Immuno-Engineering, Henan University, Kaifeng, Henan, China
| | - Songqiang Xie
- School of Pharmacy, Henan University, Kaifeng, Henan, China
| | - Chaojie Wang
- Key Laboratory of Natural Medicine and Immuno-Engineering, Henan University, Kaifeng, Henan, China
| | - Lei Gao
- Key Laboratory of Natural Medicine and Immuno-Engineering, Henan University, Kaifeng, Henan, China
- Joint International Research Laboratory of Food & Medicine Resource Function, Henan University, Kaifeng, Henan, China
| | - Yanming Wang
- School of Life Sciences, Henan University, Kaifeng, Henan, China
| | - Fujun Dai
- Key Laboratory of Natural Medicine and Immuno-Engineering, Henan University, Kaifeng, Henan, China
| |
Collapse
|
6
|
Roy T, Boateng ST, Uddin MB, Banang-Mbeumi S, Yadav RK, Bock CR, Folahan JT, Siwe-Noundou X, Walker AL, King JA, Buerger C, Huang S, Chamcheu JC. The PI3K-Akt-mTOR and Associated Signaling Pathways as Molecular Drivers of Immune-Mediated Inflammatory Skin Diseases: Update on Therapeutic Strategy Using Natural and Synthetic Compounds. Cells 2023; 12:1671. [PMID: 37371141 PMCID: PMC10297376 DOI: 10.3390/cells12121671] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/10/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
The dysregulated phosphatidylinositol-3-kinase (PI3K)-Akt-mammalian target of rapamycin (mTOR) signaling pathway has been implicated in various immune-mediated inflammatory and hyperproliferative dermatoses such as acne, atopic dermatitis, alopecia, psoriasis, wounds, and vitiligo, and is associated with poor treatment outcomes. Improved comprehension of the consequences of the dysregulated PI3K/Akt/mTOR pathway in patients with inflammatory dermatoses has resulted in the development of novel therapeutic approaches. Nonetheless, more studies are necessary to validate the regulatory role of this pathway and to create more effective preventive and treatment methods for a wide range of inflammatory skin diseases. Several studies have revealed that certain natural products and synthetic compounds can obstruct the expression/activity of PI3K/Akt/mTOR, underscoring their potential in managing common and persistent skin inflammatory disorders. This review summarizes recent advances in understanding the role of the activated PI3K/Akt/mTOR pathway and associated components in immune-mediated inflammatory dermatoses and discusses the potential of bioactive natural products, synthetic scaffolds, and biologic agents in their prevention and treatment. However, further research is necessary to validate the regulatory role of this pathway and develop more effective therapies for inflammatory skin disorders.
Collapse
Affiliation(s)
- Tithi Roy
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71209, USA; (T.R.); (S.T.B.); (S.B.-M.); (R.K.Y.); (C.R.B.); (J.T.F.); (A.L.W.)
| | - Samuel T. Boateng
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71209, USA; (T.R.); (S.T.B.); (S.B.-M.); (R.K.Y.); (C.R.B.); (J.T.F.); (A.L.W.)
| | - Mohammad B. Uddin
- Department of Toxicology and Cancer Biology, Center for Research on Environmental Diseases, College of Medicine, University of Kentucky, Lexington, KY 40536, USA;
| | - Sergette Banang-Mbeumi
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71209, USA; (T.R.); (S.T.B.); (S.B.-M.); (R.K.Y.); (C.R.B.); (J.T.F.); (A.L.W.)
- Division for Research and Innovation, POHOFI Inc., Madison, WI 53744, USA
- School of Nursing and Allied Health Sciences, Louisiana Delta Community College, Monroe, LA 71203, USA
| | - Rajesh K. Yadav
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71209, USA; (T.R.); (S.T.B.); (S.B.-M.); (R.K.Y.); (C.R.B.); (J.T.F.); (A.L.W.)
| | - Chelsea R. Bock
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71209, USA; (T.R.); (S.T.B.); (S.B.-M.); (R.K.Y.); (C.R.B.); (J.T.F.); (A.L.W.)
| | - Joy T. Folahan
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71209, USA; (T.R.); (S.T.B.); (S.B.-M.); (R.K.Y.); (C.R.B.); (J.T.F.); (A.L.W.)
| | - Xavier Siwe-Noundou
- Department of Pharmaceutical Sciences, School of Pharmacy, Sefako Makgatho Health Sciences University, P.O. Box 218, Pretoria 0208, South Africa;
| | - Anthony L. Walker
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71209, USA; (T.R.); (S.T.B.); (S.B.-M.); (R.K.Y.); (C.R.B.); (J.T.F.); (A.L.W.)
| | - Judy A. King
- Department of Pathology and Translational Pathobiology, LSU Health Shreveport, 1501 Kings Highway, Shreveport, LA 71103, USA;
- College of Medicine, Belmont University, 900 Belmont Boulevard, Nashville, TN 37212, USA
| | - Claudia Buerger
- Department of Dermatology, Venerology and Allergology, Clinic of the Goethe University, 60590 Frankfurt am Main, Germany;
| | - Shile Huang
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71130, USA;
- Department of Hematology and Oncology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71130, USA
- Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA
| | - Jean Christopher Chamcheu
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71209, USA; (T.R.); (S.T.B.); (S.B.-M.); (R.K.Y.); (C.R.B.); (J.T.F.); (A.L.W.)
- Department of Pathology and Translational Pathobiology, LSU Health Shreveport, 1501 Kings Highway, Shreveport, LA 71103, USA;
| |
Collapse
|
7
|
Zani CP, Zani AP, Thomazini CM, Retamiro KM, de Oliveira AR, Gonçalves DL, Sarragiotto MH, Garcia FP, de Oliveira Silva S, Nakamura CV, Ueda-Nakamura T. β-Carboline-α-aminophosphonate Derivative: A Promising Antitumor Agent for Breast Cancer Treatment. Molecules 2023; 28:molecules28093949. [PMID: 37175359 PMCID: PMC10179861 DOI: 10.3390/molecules28093949] [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: 03/22/2023] [Revised: 05/03/2023] [Accepted: 05/04/2023] [Indexed: 05/15/2023] Open
Abstract
Breast cancer is the most common type of cancer and the leading cause of cancer mortality among women worldwide. Considering the limitations of the current treatments available, we analyzed the in vitro cytotoxic potential of ((4-Fluoro-phenyl)-{2-[(1-phenyl-9H-β-carboline-3-carbonyl)-amino]-ethylamino}-methyl)-phosphonic acid dibutyl ester (BCP-1) in breast cancer cells (MCF-7 and MDA-MB-231) and in a non-tumor breast cell line (MCF-10A). BCP-1 has an α-aminophosphonate unit linked to the β-carboline nucleus, and the literature indicates that compounds of these classes have high biological potential. In the present study, the mechanism of action of BCP-1 was investigated through methods of spectrofluorimetry, flow cytometry, and protein expression analysis. It was found that BCP-1 inhibited the proliferation of both cancer cell lines. Furthermore, it induced oxidative stress and cell cycle arrest in G2/M. Upregulation of apoptosis-related proteins such as Bax, cytochrome C, and caspases, as well as a decrease in the anti-apoptotic protein Bcl-2, indicated potential induction of apoptosis in the MDA-MB-231 cells. While in MCF-7 cells, BCP-1 activated the autophagic death pathway, which was demonstrated by an increase in autophagic vacuoles and acidic organelles, in addition to increased expression of LC3I/LC3II and reduced SQSTM1/p62 expression. Further, BCP-1 demonstrated antimetastatic potential by reducing MMP-9 expression and cell migration in both breast cancer cell lines. In conclusion, BCP-1 is a promising candidate for breast cancer chemotherapy.
Collapse
Affiliation(s)
- Caroline Pinto Zani
- Laboratory of Technological Innovation in the Development of Pharmaceuticals and Cosmetics, State University of Maringá, Maringá CEP 87020-900, Paraná, Brazil
| | - Aline Pinto Zani
- Laboratory of Technological Innovation in the Development of Pharmaceuticals and Cosmetics, State University of Maringá, Maringá CEP 87020-900, Paraná, Brazil
| | - Cristiane Melissa Thomazini
- Laboratory of Technological Innovation in the Development of Pharmaceuticals and Cosmetics, State University of Maringá, Maringá CEP 87020-900, Paraná, Brazil
| | - Karina Miyuki Retamiro
- Laboratory of Technological Innovation in the Development of Pharmaceuticals and Cosmetics, State University of Maringá, Maringá CEP 87020-900, Paraná, Brazil
| | | | - Débora Laís Gonçalves
- Department of Chemistry, State University of Maringá, Maringá CEP 87020-900, Paraná, Brazil
| | | | - Francielle Pelegrin Garcia
- Laboratory of Technological Innovation in the Development of Pharmaceuticals and Cosmetics, State University of Maringá, Maringá CEP 87020-900, Paraná, Brazil
| | - Sueli de Oliveira Silva
- Laboratory of Technological Innovation in the Development of Pharmaceuticals and Cosmetics, State University of Maringá, Maringá CEP 87020-900, Paraná, Brazil
| | - Celso Vataru Nakamura
- Laboratory of Technological Innovation in the Development of Pharmaceuticals and Cosmetics, State University of Maringá, Maringá CEP 87020-900, Paraná, Brazil
| | - Tania Ueda-Nakamura
- Laboratory of Technological Innovation in the Development of Pharmaceuticals and Cosmetics, State University of Maringá, Maringá CEP 87020-900, Paraná, Brazil
| |
Collapse
|
8
|
Qin R, You FM, Zhao Q, Xie X, Peng C, Zhan G, Han B. Naturally derived indole alkaloids targeting regulated cell death (RCD) for cancer therapy: from molecular mechanisms to potential therapeutic targets. J Hematol Oncol 2022; 15:133. [PMID: 36104717 PMCID: PMC9471064 DOI: 10.1186/s13045-022-01350-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 09/03/2022] [Indexed: 12/11/2022] Open
Abstract
Regulated cell death (RCD) is a critical and active process that is controlled by specific signal transduction pathways and can be regulated by genetic signals or drug interventions. Meanwhile, RCD is closely related to the occurrence and therapy of multiple human cancers. Generally, RCD subroutines are the key signals of tumorigenesis, which are contributed to our better understanding of cancer pathogenesis and therapeutics. Indole alkaloids derived from natural sources are well defined for their outstanding biological and pharmacological properties, like vincristine, vinblastine, staurosporine, indirubin, and 3,3′-diindolylmethane, which are currently used in the clinic or under clinical assessment. Moreover, such compounds play a significant role in discovering novel anticancer agents. Thus, here we systemically summarized recent advances in indole alkaloids as anticancer agents by targeting different RCD subroutines, including the classical apoptosis and autophagic cell death signaling pathways as well as the crucial signaling pathways of other RCD subroutines, such as ferroptosis, mitotic catastrophe, necroptosis, and anoikis, in cancer. Moreover, we further discussed the cross talk between different RCD subroutines mediated by indole alkaloids and the combined strategies of multiple agents (e.g., 3,10-dibromofascaplysin combined with olaparib) to exhibit therapeutic potential against various cancers by regulating RCD subroutines. In short, the information provided in this review on the regulation of cell death by indole alkaloids against different targets is expected to be beneficial for the design of novel molecules with greater targeting and biological properties, thereby facilitating the development of new strategies for cancer therapy.
Collapse
|
9
|
Luo ML, Huang W, Zhu HP, Peng C, Zhao Q, Han B. Advances in indole-containing alkaloids as potential anticancer agents by regulating autophagy. Biomed Pharmacother 2022; 149:112827. [PMID: 35316753 DOI: 10.1016/j.biopha.2022.112827] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/03/2022] [Accepted: 03/14/2022] [Indexed: 11/02/2022] Open
Abstract
Cancer is a leading cause of death worldwide, and cancer development is often associated with disturbances in the autophagy process. Autophagy is a catabolic process involved in many physiological processes, crucial for cell growth and survival. It is an intracellular lysosomal/vacuolar degradation system. In this system, inner cytoplasmic cell membrane is degraded by lysosomal hydrolases, and the products are released back into the cytoplasm. Indole alkaloids are natural products extensively found in nature and have been proven to possess various pharmacological activities. In recent years, pharmacological studies have demonstrated another potential of indole alkaloids, autophagy regulation. The regulation may contribute to the efficacy of indole alkaloids in preventing and treating cancer. This review summarizes the current understanding of indole alkaloids' effect on tumor cells and autophagy. Then, we focus on mechanisms by which indole alkaloids can target the autophagy process associated with cancer, including the PI3K/Akt/mTOR signaling pathway, MAPK signaling pathway, ROS signaling pathway, Beclin-1, and so on. Literature has been surveyed primarily from 2009 to Nov. 2021, and some semisynthetic or fully synthetic indole derivatives are also discussed.
Collapse
Affiliation(s)
- Meng-Lan Luo
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wei Huang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hong-Ping Zhu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China; Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qian Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Bo Han
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
10
|
Harmine Augments the Cytotoxic and Anti-invasive Potential of Temozolomide Against Glioblastoma Multiforme Cells. Jundishapur J Nat Pharm Prod 2021. [DOI: 10.5812/jjnpp.115464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Background: Glioblastoma multiforme (GBM) is considered the deadliest human cancer. Temozolomide is now a part of postresection standard chemotherapy for this type of cancer. Unfortunately, resistance to temozolomide is a major obstacle to treatment success. Combination therapy with natural anticancer agents increases the activity of temozolomide against cancer cells. Objectives: This study aimed to assess the effects of temozolomide in combination with harmine against GBM cells. Methods: Cancer cells were treated with temozolomide and/or harmine. After 24, 48, 72, and 96 h, the viability of the cells was assessed by the MTT test. The combination index and dose reduction index were determined by CompuSyn software. Tumor invasion potential was investigated by evaluating cell migration, invasion, and adhesion. The real-time PCR technique was done to study the expression pattern of two genes involved in cancer cell invasion. Statistical analysis was performed using one-way analysis of variance and Tukey’s post-hoc test, and differences were considered non-significant at P > 0.05. Results: After treatment with temozolomide, cell viability showed a concentration- and time-dependent decrease, and the cells’ survival rate decreased. The combination of temozolomide and harmine had a synergistic effect. Also, temozolomide and/or harmine treatment decreased cancer cells’ migration, invasion, and adhesion potentials, as well as the expression of metalloproteinases 2 and 9 in T98G cells. Conclusions: The combination of temozolomide and harmine can be promising for the successful treatment of GBM.
Collapse
|
11
|
Arora S, Dharavath RN, Bansal Y, Bishnoi M, Kondepudi KK, Chopra K. Neurobehavioral alterations in a mouse model of chronic partial sleep deprivation. Metab Brain Dis 2021; 36:1315-1330. [PMID: 33740181 DOI: 10.1007/s11011-021-00693-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 02/12/2021] [Indexed: 12/23/2022]
Abstract
The night shift paradigm induces a state of chronic partial sleep deprivation (CPSD) and enhances the vulnerability to neuronal dysfunction. However, the specific neuronal impact of CPSD has not been thoroughly explored to date. In the current study, the night shift condition was mimicked in female Swiss albino mice. The classical sleep deprivation model, i.e., Modified Multiple Platform (MMP) method, was used for 8 h/day from Monday to Friday with Saturday and Sunday as a weekend off for nine weeks. Following nine weeks of night shift schedule, their neurobehavioral profile and physiological parameters were assessed along with the activity of the mitochondrial complexes, oxidative stress, serotonin levels, and inflammatory markers in the brain. Mice showed an overall hyperactive behavioral profile including hyperlocomotion, aggression, and stereotyped behavior accompanied by decreased activity of mitochondrial enzymes and serotonin levels, increased oxidative stress and inflammatory markers in whole brain homogenates. Collectively, the study points towards the occurrence of a hyperactive behavioral profile akin to mania and psychosis as a potential consequence of CPSD.
Collapse
Affiliation(s)
- Shiyana Arora
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences (UIPS), Panjab University, Sector 14, 160014, Chandigarh, India
| | - Ravinder Naik Dharavath
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences (UIPS), Panjab University, Sector 14, 160014, Chandigarh, India
| | - Yashika Bansal
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences (UIPS), Panjab University, Sector 14, 160014, Chandigarh, India
| | - Mahendra Bishnoi
- Food and Nutritional Biotechnology Laboratory, National Agri-Food Biotechnology Institute, SAS Nagar, Punjab, 140306, India
| | - Kanthi Kiran Kondepudi
- Food and Nutritional Biotechnology Laboratory, National Agri-Food Biotechnology Institute, SAS Nagar, Punjab, 140306, India
| | - Kanwaljit Chopra
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences (UIPS), Panjab University, Sector 14, 160014, Chandigarh, India.
| |
Collapse
|
12
|
Jalali A, Dabaghian F, Zarshenas MM. Alkaloids of Peganum harmala: Anticancer Biomarkers with Promising Outcomes. Curr Pharm Des 2021; 27:185-196. [PMID: 33238864 DOI: 10.2174/1381612826666201125103941] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Accepted: 08/24/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Cancer is a serious and growing global health issue worldwide. In the cancerous cells, the normal cell cycle has been disrupted via a series of irreversible changes. Recently, the investigations on herbal medicine and clarifying the phytochemicals potential in treat cancer has been increased. The combination of phytochemicals with conventional cancer treatment approaches can improve outcomes via advancing cell death, restraining cell proliferation and invasion, sensitizing cancerous cells, and promoting the immune system. Therefore, phytochemicals can be introduced as relevant complementary medicaments in cancer therapy. Peganum harmala L. (Zygophyllaceae) as a valuable medicinal herb, possesses various alkaloid ingredient. OBJECTIVE Pointing to the importance of new avenues for cancer management and P. harmala convincing effect in this field, this review strived to collect a frame to epitome possible scopes to develop novel medicines in cancer treatment. METHODS Keywords "Peganum harmala" and cancer, or chemotherapy, or anti-neoplasm were searched through the "Scopus" database up to 29th of February 2020. Papers linking to agriculture, chemistry, environmental, and genetics sciences were omitted and, papers centered on cancer were selected. RESULTS AND DISCUSSION In the current study, 42 related papers to cancer treatment and 22 papers on alkaloid bioactive components are collected from 72 papers. The β-carboline alkaloids derived from P. harmala, especially harmine, demonstrate notable anticancer properties by targeting apoptosis, autophagy, abnormal cell proliferation, angiogenesis, metastasis, and cytotoxicity. Based on the collected information, P. harmala holds significant anticancer activity. Considering the mechanism of the various anticancer drugs and their acting similarity to P. harmala, the alkaloids derived from this herb, particularly harmine, can introduce as a novel anticancer medicine solely or in adjuvant cancer therapy.
Collapse
Affiliation(s)
- Atefeh Jalali
- Medicinal Plants Processing Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Farid Dabaghian
- Department of Pharmacognosy, School of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad M Zarshenas
- Medicinal Plants Processing Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
13
|
Zhu YG, Lv YX, Guo CY, Xiao ZM, Jiang QG, Kuang H, Zhang WH, Hu P. Harmine inhibits the proliferation and migration of glioblastoma cells via the FAK/AKT pathway. Life Sci 2021; 270:119112. [DOI: 10.1016/j.lfs.2021.119112] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 01/09/2021] [Accepted: 01/19/2021] [Indexed: 01/01/2023]
|
14
|
Li J, Liu R. Autophagy in Cellular Stress Responses. OXIDATIVE STRESS 2021:133-154. [DOI: 10.1007/978-981-16-0522-2_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
15
|
Alternative Options for Skin Cancer Therapy via Regulation of AKT and Related Signaling Pathways. Int J Mol Sci 2020; 21:ijms21186869. [PMID: 32962182 PMCID: PMC7560163 DOI: 10.3390/ijms21186869] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/11/2020] [Accepted: 09/17/2020] [Indexed: 12/25/2022] Open
Abstract
Global environmental pollution has led to human exposure to ultraviolet (UV) radiation due to the damaged ozone layer, thereby increasing the incidence and death rate of skin cancer including both melanoma and non-melanoma. Overexpression and activation of V-akt murine thymoma viral oncogene homolog (AKT, also known as protein kinase B) and related signaling pathways are major factors contributing to many cancers including lung cancer, esophageal squamous cell carcinoma and skin cancer. Although BRAF inhibitors are used to treat melanoma, further options are needed due to treatment resistance and poor efficacy. Depletion of AKT expression and activation, and related signaling cascades by its inhibitors, decreases the growth of skin cancer and metastasis. Here we have focused the effects of AKT and related signaling (PI3K/AKT/mTOR) pathways by regulators derived from plants and suggest the need for efficient treatment in skin cancer therapy.
Collapse
|
16
|
Xiang W, Zhang RJ, Jin GL, Tian L, Cheng F, Wang JZ, Xing XF, Xi W, Tang SJ, Chen JF. RCE‑4, a potential anti‑cervical cancer drug isolated from Reineckia carnea, induces autophagy via the dual blockade of PI3K and ERK pathways in cervical cancer CaSki cells. Int J Mol Med 2019; 45:245-254. [PMID: 31746346 DOI: 10.3892/ijmm.2019.4389] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 08/29/2019] [Indexed: 11/06/2022] Open
Abstract
The steroidal saponin RCE‑4 (1β, 3β, 5β, 25S)‑spirostan‑1, 3‑diol 1‑[α‑L‑rhamnopyranosyl‑(1→2)‑β‑D‑xylopyranoside], isolated from Reineckia carnea, exerts significant anti‑cervical cancer activity by inducing apoptosis. The potential effect of RCE‑4 on proliferation inhibition and autophagy induction has rarely been studied. Therefore, the focus of the present study was to investigate the effects of RCE‑4 on proliferation, and to elucidate the detailed mechanisms involved in autophagy induction in cervical cancer cells. CaSki cells were treated with RCE‑4 or/and autophagy inhibitors, and the effect of RCE‑4 on cellular proliferation was assessed by MTT assay. The pro‑autophagic properties of RCE‑4 were subsequently confirmed using monomeric red fluorescent protein‑green fluorescent protein‑microtubule‑associated proteins 1A/1B light chain 3B (LC3) adenoviruses and CYTO‑ID autophagy assays, and by assessing the accumulation of lipid‑modified LC3 (LC3II). The mechanisms of RCE‑4‑induced autophagy were investigated by western blot analysis. The results demonstrated that inhibiting autophagy significantly promoted RCE‑4‑induced cell death, indicating that autophagy served a protective role following RCE‑4 treatment. In addition, RCE‑4‑induced autophagy was reflected by increased expression levels of the serine/threonine‑protein kinase ULK1, phosphorylated (p)‑ULK1, p‑Beclin‑1 and LC3II, the formation of autophagosomes and autolysosomes, and sequestosome 1 (p62) degradation. Subsequent analysis indicated that RCE‑4 activated the AMP‑activated protein kinase (AMPK) pathway by upregulating AMPK and p‑AMPK, and also inhibited the PI3K and extracellular signal‑regulated kinase (ERK) signaling pathways by downregulating p‑PI3K, p‑Akt, p‑mTOR, Ras, c‑Raf, p‑c‑Raf, dual specificity mitogen‑activated protein kinase kinase (MEK)1/2, p‑MEK1/2 and p‑Erk1/2. Additionally, with increased treatment times RCE‑4 may impair lysosomal cathepsin activity and inhibit autophagy flux by suppressing the expression of AMPK, p‑AMPK, ULK1, p‑ULK1 and p‑Beclin‑1, and upregulating that of p62. These results indicated that the dual RCE‑4‑induced inhibition of the PI3K and ERK pathways may result in a more significant anti‑tumor effect and prevent chemoresistance, compared with the inhibition of either single pathway; furthermore, dual blockade of PI3K and ERK, and the AMPK pathway may be involved in the regulation of autophagy caused by RCE‑4. Taken together, RCE‑4 induced autophagy to protect cancer cells against apoptosis, but AMPK‑mediated autophagy was inhibited in the later stages of RCE‑4 treatment. In addition, autophagy inhibition improved the therapeutic effect of RCE‑4. These data highlight RCE‑4 as a potential candidate for cervical cancer treatment.
Collapse
Affiliation(s)
- Wei Xiang
- Hubei Key Laboratory of Natural Products Research and Development, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang, Hubei 443002, P.R. China
| | - Ren-Jing Zhang
- Department of Pathology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Gui-Lan Jin
- The First People's Hospital of Yichang and The People's Hospital of China Three Gorges University, Yichang, Hubei 443002, P.R. China
| | - Li Tian
- Hubei Key Laboratory of Natural Products Research and Development, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang, Hubei 443002, P.R. China
| | - Fan Cheng
- Hubei Key Laboratory of Natural Products Research and Development, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang, Hubei 443002, P.R. China
| | - Jun-Zhi Wang
- Hubei Key Laboratory of Natural Products Research and Development, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang, Hubei 443002, P.R. China
| | - Xiang-Fei Xing
- The First People's Hospital of Yichang and The People's Hospital of China Three Gorges University, Yichang, Hubei 443002, P.R. China
| | - Wei Xi
- The First People's Hospital of Yichang and The People's Hospital of China Three Gorges University, Yichang, Hubei 443002, P.R. China
| | - Shu-Jun Tang
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, IL 60616, USA
| | - Jian-Feng Chen
- Hubei Key Laboratory of Natural Products Research and Development, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang, Hubei 443002, P.R. China
| |
Collapse
|
17
|
Chamcheu JC, Roy T, Uddin MB, Banang-Mbeumi S, Chamcheu RCN, Walker AL, Liu YY, Huang S. Role and Therapeutic Targeting of the PI3K/Akt/mTOR Signaling Pathway in Skin Cancer: A Review of Current Status and Future Trends on Natural and Synthetic Agents Therapy. Cells 2019; 8:cells8080803. [PMID: 31370278 PMCID: PMC6721560 DOI: 10.3390/cells8080803] [Citation(s) in RCA: 130] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 07/26/2019] [Accepted: 07/29/2019] [Indexed: 12/16/2022] Open
Abstract
The mammalian or mechanistic target of rapamycin (mTOR) and associated phosphatidyl-inositiol 3-kinase (PI3K)/protein kinase B (Akt) pathways regulate cell growth, differentiation, migration, and survival, as well as angiogenesis and metabolism. Dysregulation of these pathways is frequently associated with genetic/epigenetic alterations and predicts poor treatment outcomes in a variety of human cancers including cutaneous malignancies like melanoma and non-melanoma skin cancers. Recently, the enhanced understanding of the molecular and genetic basis of skin dysfunction in patients with skin cancers has provided a strong basis for the development of novel therapeutic strategies for these obdurate groups of skin cancers. This review summarizes recent advances in the roles of PI3K/Akt/mTOR and their targets in the development and progression of a broad spectrum of cutaneous cancers and discusses the current progress in preclinical and clinical studies for the development of PI3K/Akt/mTOR targeted therapies with nutraceuticals and synthetic small molecule inhibitors.
Collapse
Affiliation(s)
| | - Tithi Roy
- College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71209-0497, USA
| | | | - Sergette Banang-Mbeumi
- College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71209-0497, USA
- Division for Research and Innovation, POHOFI Inc., P.O. Box 44067, Madison, WI 53744, USA
- School of Nursing and Allied Health Sciences, Louisiana Delta Community College, Monroe, LA 71203, USA
| | | | - Anthony L Walker
- College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71209-0497, USA
| | - Yong-Yu Liu
- College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71209-0497, USA
| | - Shile Huang
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71130-3932, USA
- Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport, LA 71130-3932, USA
| |
Collapse
|
18
|
Ding Y, He J, Huang J, Yu T, Shi X, Zhang T, Yan G, Chen S, Peng C. Harmine induces anticancer activity in breast cancer cells via targeting TAZ. Int J Oncol 2019; 54:1995-2004. [PMID: 31081045 PMCID: PMC6521938 DOI: 10.3892/ijo.2019.4777] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 03/27/2019] [Indexed: 12/21/2022] Open
Abstract
Harmine (HM) is a β-carboline alkaloid found in multiple medicinal plants. It has been used in folk medicine for anticancer therapy; however, the molecular mechanism of HM on human breast cancer remains unclear. Transcriptional co-activator with PDZ-binding motif (TAZ), also known as WW domain-containing transcription regulator 1, serves an important role in the carcinogenesis and progression of breast cancer. The aim of the present study was to elucidate the potential anticancer activity and mechanism of HM in breast cancer, in vitro and in vivo. Cell proliferation was measured using a CCK-8 assay, apoptotic activity was detected by flow cytometry and DAPI staining, and cell migration was examined using a wound healing assay. The expression of proteins, including extracellular signal-regulate kinase (Erk), phosphorylated (p-) Erk, protein kinase B (Akt), p-Akt, B-cell lymphoma 2 (Bcl-2) and Bcl-2-associated X protein (Bax), were determined by western blotting. The mRNA expression of TAZ was detected using reverse transcription-quantitative polymerase chain reaction analysis. The expression of proteins in mouse tumor tissues were examined by immunohistochemistry. HM significantly suppressed cellular proliferation and migration, promoted apoptosis in vitro and inhibited tumor growth in vivo. In addition, HM significantly decreased the expression of TAZ, p-Erk, p-Akt and Bcl-2, but increased that of Bax. The overexpression of TAZ in breast cancer cells inhibited the antitumor effect of HM. In conclusion, HM was found to induce apoptosis and prevent the proliferation and migration of human breast cancer cell lines, possibly via the downregulation of TAZ.
Collapse
Affiliation(s)
- Yu Ding
- Key Laboratory for Molecular Diagnosis of Hubei Province, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Jinrong He
- Key Laboratory for Molecular Diagnosis of Hubei Province, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Juan Huang
- Department of Nephrology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Tong Yu
- Department of Traditional Chinese Medicine, Humanwell Healthcare (Group) Co., Ltd., Wuhan, Hubei 430075, P.R. China
| | - Xiaoyan Shi
- Key Laboratory for Molecular Diagnosis of Hubei Province, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Tianzhu Zhang
- Key Laboratory for Molecular Diagnosis of Hubei Province, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Ge Yan
- Key Laboratory for Molecular Diagnosis of Hubei Province, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Shanshan Chen
- Key Laboratory for Molecular Diagnosis of Hubei Province, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Caixia Peng
- Key Laboratory for Molecular Diagnosis of Hubei Province, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| |
Collapse
|
19
|
β-Carboline and N-hydroxycinnamamide hybrids as anticancer agents for drug-resistant hepatocellular carcinoma. Eur J Med Chem 2019; 168:515-526. [DOI: 10.1016/j.ejmech.2019.02.054] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 01/15/2019] [Accepted: 02/17/2019] [Indexed: 12/20/2022]
|
20
|
Zhao HG, Zhou SL, Lin YY, Wang H, Dai HF, Huang FY. Autophagy plays a protective role against apoptosis induced by toxicarioside N via the Akt/mTOR pathway in human gastric cancer SGC-7901 cells. Arch Pharm Res 2018; 41:986-994. [PMID: 29992400 DOI: 10.1007/s12272-018-1049-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 06/21/2018] [Indexed: 12/12/2022]
Abstract
Toxicarioside N (Tox N), a natural product extract from Antiaris toxicaria, has been reported to induce apoptosis in human gastric cancer cells. However, the mechanism and actual role of autophagy in Tox N-induced apoptosis of human gastric cancer cells remains poorly understood. In the current study, we demonstrated that Tox N could induce autophagy by inhibiting the Akt/mTOR signaling pathway in SGC-7901 cells. Moreover, we found that the inhibition of autophagy by 3-methyladenine, an autophagy inhibitor, enhanced Tox N-induced apoptotic cell death. However, the stimulation of autophagy by rapamycin, an autophagy activator, remarkably suppressed Tox N-induced apoptosis, suggesting that autophagy plays a protective role in Tox N-induced apoptosis. Thus, the results from this study suggested that Tox N combination with an autophagy inhibitor might be a promising strategy to enhance the anticancer activity of Tox N for the treatment of human gastric cancer.
Collapse
Affiliation(s)
- Huan-Ge Zhao
- Key Laboratory of Tropical Diseases and Translational Medicine of the Ministry of Education, Hainan Medical College, Haikou, 571199, China
- Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical College, Haikou, 571199, China
| | - Song-Lin Zhou
- Key Laboratory of Tropical Diseases and Translational Medicine of the Ministry of Education, Hainan Medical College, Haikou, 571199, China
- Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical College, Haikou, 571199, China
| | - Ying-Ying Lin
- Key Laboratory of Tropical Diseases and Translational Medicine of the Ministry of Education, Hainan Medical College, Haikou, 571199, China
- Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical College, Haikou, 571199, China
| | - Hua Wang
- Key Laboratory of Tropical Diseases and Translational Medicine of the Ministry of Education, Hainan Medical College, Haikou, 571199, China
- Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical College, Haikou, 571199, China
| | - Hao-Fu Dai
- Institutes of Tropical Bioscience and Biotechnology, Chinese Academy of Tropical Agricultural Sciences, Haikou, 571199, China.
| | - Feng-Ying Huang
- Key Laboratory of Tropical Diseases and Translational Medicine of the Ministry of Education, Hainan Medical College, Haikou, 571199, China.
- Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical College, Haikou, 571199, China.
| |
Collapse
|
21
|
Wei Y, Ma T, Wang H, Xing J, Wang Y, Gu Z, Mu D, Yin Q, Cheng X, Wang C. Extracts of compound Muniziqi granule suppressed uterus contraction and ameliorated oxytocin-induced primary dysmenorrhea. JOURNAL OF ETHNOPHARMACOLOGY 2018; 223:33-40. [PMID: 29783015 DOI: 10.1016/j.jep.2018.05.024] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Revised: 05/16/2018] [Accepted: 05/17/2018] [Indexed: 06/08/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Compound Muniziqi granule (CMG) is usually used as a traditional Uighur medicine to treat acne, chloasma, skin inflammation, primary dysmenorrhea (PDM), and menopausal syndrome. However, there are no sufficient data to support the clinic uses of CMG in PDM. AIM OF THE STUDY This work aims to examine the effect of CMG as a treatment for PDM and reveal its possible therapeutic mechanism. MATERIALS AND METHODS In vivo and in vitro mouse PDM models were utilized in this study. The mouse uterine contraction was induced by oxytocin after progynova or estradiol benzoate pretreatment. CMG, alkaloid extracts from seeds of Peganum harmala (AEP), and 10% and 95% ethanol extracts from seeds of Nigella glandulifera (EEN10 and EEN95) were given to mice in three doses by gavage. The writhing times within 30 min after oxytocin treatment were recorded to evaluate the analgesic effect, and the glutathione peroxidase (GSH-Px), malondialdehyde (MDA), 6-keto-prostaglandin F1α (6-k-PGF1α), prostaglandin F2α (PGF2α), thromboxane B2 (TXB2), and nitric oxide (NO) levels in uterine tissues and PGF2α and MDA in serum were determined. The effects (contractile curve) of CMG, AEP, EEN10, and EEN95 on uterus contraction induced by oxytocin in isolated mouse uterus were recorded. RESULTS In contrast to the control group, CMG, AEP, N10, and N95 could display analgesic activities dose dependently by reducing the writhing response of the PDM model mice. CMG, AEP, EEN10, and EEN95 could also remarkably decrease the level of PGF2α, 6-k-PGF1α, TXB2, NO and MDA in uterine tissues and PGF2α and MDA in serum, whereas the activity of GSH-Px in uterine tissues was increased. Furthermore, CMG, AEP, EEN10, and EEN95 could significantly inhibit the frequency and amplitude of isolated uterus induced by oxytocin in a concentration-dependent manner. CONCLUSIONS CMG exhibited a significant protective effect on experimental PDM. The mechanisms are probably associated with abating lipid peroxidation and over-inflammatory reaction, and alleviating the contraction of isolated mouse uterus. The seeds of P. harmala and N. glandulifera in the CMG may play an important role in exerting protective effects on PDM. This study provides pre-clinic proof to the use of CMG in clinical practice of PDM.
Collapse
Affiliation(s)
- Yue Wei
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine; The MOE Key Laboratory for Standardization of Chinese Medicines; Shanghai Key Laboratory of Compound Chinese Medicines; Shanghai R&D Centre for Standardization of Chinese Medicines, 1200 Cailun Rood, Shanghai 201203, China
| | - Tingyun Ma
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine; The MOE Key Laboratory for Standardization of Chinese Medicines; Shanghai Key Laboratory of Compound Chinese Medicines; Shanghai R&D Centre for Standardization of Chinese Medicines, 1200 Cailun Rood, Shanghai 201203, China
| | - Hanxue Wang
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine; The MOE Key Laboratory for Standardization of Chinese Medicines; Shanghai Key Laboratory of Compound Chinese Medicines; Shanghai R&D Centre for Standardization of Chinese Medicines, 1200 Cailun Rood, Shanghai 201203, China
| | - Jianguo Xing
- The Xinjiang Institute of Materia Medica, Urumqi, Xinjiang 830002, China
| | - Yuwen Wang
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine; The MOE Key Laboratory for Standardization of Chinese Medicines; Shanghai Key Laboratory of Compound Chinese Medicines; Shanghai R&D Centre for Standardization of Chinese Medicines, 1200 Cailun Rood, Shanghai 201203, China
| | - Zhengyi Gu
- The Xinjiang Institute of Materia Medica, Urumqi, Xinjiang 830002, China
| | - Dandan Mu
- Xinjiang Uighur Pharmaceutical Limited Company, Urumqi, Xinjiang 830001, China
| | - Qiang Yin
- Xinjiang Uighur Pharmaceutical Limited Company, Urumqi, Xinjiang 830001, China
| | - Xuemei Cheng
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine; The MOE Key Laboratory for Standardization of Chinese Medicines; Shanghai Key Laboratory of Compound Chinese Medicines; Shanghai R&D Centre for Standardization of Chinese Medicines, 1200 Cailun Rood, Shanghai 201203, China.
| | - Changhong Wang
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine; The MOE Key Laboratory for Standardization of Chinese Medicines; Shanghai Key Laboratory of Compound Chinese Medicines; Shanghai R&D Centre for Standardization of Chinese Medicines, 1200 Cailun Rood, Shanghai 201203, China; The Xinjiang Institute of Materia Medica, Urumqi, Xinjiang 830002, China.
| |
Collapse
|