1
|
Foroutan Z, Cicero AFG, Jamialahmadi T, Sahebkar A. Curcuminoids as natural modulators of necroptosis: therapeutic implications. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:1299-1304. [PMID: 39287673 DOI: 10.1007/s00210-024-03455-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 09/11/2024] [Indexed: 09/19/2024]
Abstract
Necroptosis is an emerging form of programmed cell death characterized by necrosis, an inflammatory type of cell death. Necroptosis is primarily initiated by specific mediators that interact with receptor proteins, leading to the activation of protein kinases RIPK1 and RIPK3. These kinases transmit death signals and recruit and phosphorylate mixed lineage kinase domain-like protein (MLKL), which ultimately triggers cell death and necroptosis. Curcuminoids, natural compounds derived from turmeric, have been shown to possess various therapeutic benefits, including neuroprotective, anti-metabolic syndrome, anti-inflammatory, and anti-cancer effects. In this concise overview, we aim to explore the relationship between curcuminoids and the molecular mechanisms of the necroptosis pathway based on recent in vivo and in vitro studies. The available literature indicates that curcuminoids, mainly curcumin, can act as inhibitors of necroptosis in tissue damage scenarios while serving as a necroptosis inducer in cancer cells. Curcuminoids significantly influence key indicators of necroptosis, highlighting their potential to enhance disease treatment. Future studies should focus on further investigating this important component of turmeric to advance therapeutic approaches.
Collapse
Affiliation(s)
- Zahra Foroutan
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Arrigo Francesco Giuseppe Cicero
- Hypertension and Cardiovascular Risk Research Group, Medical and Surgical Sciences Department, Alma Mater Studiorum University of Bologna, Bologna, Italy
- Cardiovascular Medicine Unit, IRCCS AOU Bologna, Bologna, Italy
| | - Tannaz Jamialahmadi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India.
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
2
|
Wang Y, Duan H, Zhang Z, Chen L, Li J. Research Progress on the Application of Natural Medicines in Biomaterial Coatings. MATERIALS (BASEL, SWITZERLAND) 2024; 17:5607. [PMID: 39597430 PMCID: PMC11595593 DOI: 10.3390/ma17225607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 10/29/2024] [Accepted: 11/12/2024] [Indexed: 11/29/2024]
Abstract
With the continuous progress of biomedical technology, biomaterial coatings play an important role in improving the performance of medical devices and promoting tissue repair and regeneration. The application of natural medicine to biological materials has become a hot topic due to its diverse biological activity, low toxicity, and wide range of sources. This article introduces the definition and classification of natural medicines, lists some common natural medicines, such as curcumin, allicin, chitosan, tea polyphenols, etc., and lists some biological activities of some common natural medicines, such as antibacterial, antioxidant, antitumor, and other properties. According to the different characteristics of natural medicines, physical adsorption, chemical grafting, layer-by-layer self-assembly, sol-gel and other methods are combined with biomaterials, which can be used for orthopedic implants, cardiovascular and cerebrovascular stents, wound dressings, drug delivery systems, etc., to exert their biological activity. For example, improving antibacterial properties, promoting tissue regeneration, and improving biocompatibility promote the development of medical health. Although the development of biomaterials has been greatly expanded, it still faces some major challenges, such as whether the combination between the coating and the substrate is firm, whether the drug load is released sustainably, whether the dynamic balance will be disrupted, and so on; a series of problems affects the application of natural drugs in biomaterial coatings. In view of these problems, this paper summarizes some suggestions by evaluating the literature, such as optimizing the binding method and release system; carrying out more clinical application research; carrying out multidisciplinary cooperation; broadening the application of natural medicine in biomaterial coatings; and developing safer, more effective and multi-functional natural medicine coatings through continuous research and innovation, so as to contribute to the development of the biomedical field.
Collapse
Affiliation(s)
| | | | | | - Lan Chen
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou 450001, China; (Y.W.); (H.D.); (Z.Z.)
| | - Jingan Li
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou 450001, China; (Y.W.); (H.D.); (Z.Z.)
| |
Collapse
|
3
|
Chen F, Zhan J, Liu M, Mamun AA, Huang S, Tao Y, Zhao J, Zhang Y, Xu Y, He Z, Du S, Lu W, Li X, Chen Z, Xiao J. FGF2 Alleviates Microvascular Ischemia-Reperfusion Injury by KLF2-mediated Ferroptosis Inhibition and Antioxidant Responses. Int J Biol Sci 2023; 19:4340-4359. [PMID: 37705747 PMCID: PMC10496511 DOI: 10.7150/ijbs.85692] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 08/08/2023] [Indexed: 09/15/2023] Open
Abstract
An essential pathogenic element of acute limb ischemia/reperfusion (I/R) injury is microvascular dysfunction. The majority of studies indicates that fibroblast growth factor 2 (FGF2) exhibits protective properties in cases of acute I/R injury. Albeit its specific role in the context of acute limb I/R injury is yet unknown. An impressive post-reperfusion increase in FGF2 expression was seen in a mouse model of hind limb I/R, followed by a decline to baseline levels, suggesting a key role for FGF2 in limb survivability. FGF2 appeared to reduce I/R-induced hypoperfusion, tissue edema, skeletal muscle fiber injury, as well as microvascular endothelial cells (ECs) damage within the limb, according to assessments of limb vitality, Western blotting, and immunofluorescence results. The bioinformatics analysis of RNA-sequencing revealed that ferroptosis played a key role in FGF2-facilitated limb preservation. Pharmacological inhibition of NFE2L2 prevented ECs from being affected by FGF2's anti-oxidative and anti-ferroptosis activities. Additionally, silencing of kruppel-like factor 2 (KLF2) by interfering RNA eliminated the antioxidant and anti-ferroptosis effects of FGF2 on ECs. Further research revealed that the AMPK-HDAC5 signal pathway is the mechanism via which FGF2 regulates KLF2 activity. Data from luciferase assays demonstrated that overexpression of HDAC5 prevented KLF2 from becoming activated by FGF2. Collectively, FGF2 protects microvascular ECs from I/R injury by KLF2-mediated ferroptosis inhibition and antioxidant responses.
Collapse
Affiliation(s)
- Fanfeng Chen
- Department of Wound healing, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, China
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, China
| | - Jiayu Zhan
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, China
| | - Mi Liu
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, China
| | - Abdullah Al Mamun
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, China
| | - Shanshan Huang
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, China
| | - Yibing Tao
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, China
| | - Jiaxin Zhao
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, China
| | - Yu Zhang
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, China
| | - Yitie Xu
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, China
| | - Zili He
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, China
| | - Shenghu Du
- Department of Wound healing, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, China
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, China
| | - Wei Lu
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
| | - Xiaokun Li
- Department of Wound healing, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, China
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, China
| | - Zimiao Chen
- Department of Wound healing, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, China
| | - Jian Xiao
- Department of Wound healing, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, China
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, China
| |
Collapse
|
4
|
Bozgeyik E, Bozgeyik I. Unveiling the therapeutic potential of natural-based anticancer compounds inducing non-canonical cell death mechanisms. Pathol Res Pract 2023; 248:154693. [PMID: 37516001 DOI: 10.1016/j.prp.2023.154693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 07/07/2023] [Accepted: 07/13/2023] [Indexed: 07/31/2023]
Abstract
In the Mid-19th century, Rudolf Virchow considered necrosis to be a prominent form of cell death; since then, pathologists have recognized necrosis as both a cause and a consequence of disease. About a century later, the mechanism of apoptosis, another form of cell death, was discovered, and we now know that this process is regulated by several molecular mechanisms that "programme" the cell to die. However, discoveries on cell death mechanisms are not limited to these, and recent studies have allowed the identification of novel cell death pathways that can be molecularly distinguished from necrotic and apoptotic cell death mechanisms. Moreover, the main goal of current cancer therapy is to discover and develop drugs that target apoptosis. However, resistance to chemotherapeutic agents targeting apoptosis is mainly responsible for the failure of clinical therapy and adverse side effects of the chemotherapeutic agents currently in use pose a major threat to the well-being and lives of patients. Therefore, the development of natural-based anticancer drugs with low cellular and organismal side effects is of great interest. In this comprehensive review, we thoroughly examine and discuss natural anticancer compounds that specifically target non-canonical cell death mechanisms.
Collapse
Affiliation(s)
- Esra Bozgeyik
- Department of Medical Services and Techniques, Vocational School of Health Services, Adiyaman University, Adiyaman, Turkey
| | - Ibrahim Bozgeyik
- Department of Medical Biology, Faculty of Medicine, Adiyaman University, Adiyaman, Turkey.
| |
Collapse
|
5
|
Prasad S, Saha P, Chatterjee B, Chaudhary AA, Lall R, Srivastava AK. Complexity of Tumor Microenvironment: Therapeutic Role of Curcumin and Its Metabolites. Nutr Cancer 2022; 75:1-13. [PMID: 35818029 DOI: 10.1080/01635581.2022.2096909] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The tumor microenvironment (TME) is a complex network of cellular and non-cellular components surrounding the tumor. The cellular component includes fibroblasts, adipocytes, endothelial cells, and immune cells, while non-cellular components are tumor vasculature, extracellular matrix and signaling molecules. The tumor cells have constant close interaction with their surrounding TME components that facilitate their growth, survival, and metastasis. Targeting a complex TME network and its interaction with the tumor can offer a novel strategy to disrupt cancer cell progression. Curcumin, from turmeric rhizome, is recognized as a safe and effective natural therapeutic agent against multiple diseases including cancer. Here the effects of curcumin and its metabolites on tumor-TME interaction modulating ability have been described. Curcumin and its metabolites regulate TME by inhibiting the growth of its cellular components such as cancer-associated adipocytes, cancer-associated fibroblast, tumor endothelial cells, tumor-stimulating immune cells, and inducing anticancer immune cells. They also inhibit the interplay of tumor cells to TME by suppressing non-cellular components such as extracellular matrix, and associated tumor promoting signaling-pathways. In addition, curcumin inhibits the inflammatory environment, suppresses angiogenic factors, and increases antioxidant status in TME. Overall, curcumin has the capability to regulate TME components and their interaction with tumor cells.
Collapse
Affiliation(s)
| | - Priyanka Saha
- Cancer Biology & Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, West Bengal, India
| | - Bilash Chatterjee
- Cancer Biology & Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, West Bengal, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| | - Anis Ahmad Chaudhary
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSUI), Riyadh, Saudi Arabia
| | - Rajiv Lall
- Noble Pharma, LLC, Menomonie, Wisconsin, USA
| | - Amit K Srivastava
- Cancer Biology & Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, West Bengal, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| |
Collapse
|
6
|
Abd El-Hameed NM, Abd El-Aleem SA, Khattab MA, Ali AH, Mohammed HH. Curcumin activation of nuclear factor E2-related factor 2 gene (Nrf2): Prophylactic and therapeutic effect in nonalcoholic steatohepatitis (NASH). Life Sci 2021; 285:119983. [PMID: 34599938 DOI: 10.1016/j.lfs.2021.119983] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/11/2021] [Accepted: 09/19/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUNDS Modern dietary habits have been associated with Nonalcoholic Steatohepatitis (NASH). Curcumin is a natural herbal found to suppress cellular oxidative states and could be beneficial in NASH. This study investigates the effect of curcumin in an animal model of NASH. MATERIALS AND METHODS Fifty rats were allocated into five groups. Control, High Fat Diet (HFD), curcumin prophylactic (CP) and therapeutic (CT) groups. HFD regimen was given for 16 weeks. Curcumin was given along with HFD (prophylactic) or after establishment of the model for two weeks (therapeutic). Livers and blood samples were harvested for histological, biochemical, and molecular studies. KEY FINDINGS Livers from HFD groups showed vascular, inflammatory, cellular degenerative and fibrotic changes. The hepatic damage was reflected by the increased serum liver enzymes. HFD groups showed excessive fibrotic change. Interestingly, curcumin administration as prophylactic or therapeutic significantly preserved and/or restored liver structure. This was evidenced by the normalization of the liver enzymes, preservation and/or reversibility of cellular changes and the decrease of the stage of fibrosis. Nuclear factor E2-related factor 2 gene (Nrf2) expression showed no changes in the HFD groups, however it showed upregulation in curcumin treated groups. Thus, the protective and therapeutic effect of curcumin could be induced through upregulation of the Nrf2 gene. Curcumin has a beneficial prophylactic and therapeutic effect that could hinder the development and/or treat NASH in susceptible livers. SIGNIFICANCE Curcumin has a beneficial prophylactic and therapeutic effect that could hinder the development and/or treat NASH in susceptible livers.
Collapse
|
7
|
Liang W, He X, Bi J, Hu T, Sun Y. Role of reactive oxygen species in tumors based on the 'seed and soil' theory: A complex interaction (Review). Oncol Rep 2021; 46:208. [PMID: 34328200 PMCID: PMC8329912 DOI: 10.3892/or.2021.8159] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 06/24/2021] [Indexed: 12/12/2022] Open
Abstract
Tumor microenvironment (TME) can serve as the 'soil' for the growth and survival of tumor cells and function synergically with tumor cells to mediate tumor progression and therapeutic resistance. Reactive oxygen species (ROS) is somewhat of a double‑edged sword for tumors. Accumulating evidence has reported that regulating ROS levels can serve an anti‑tumor role in the TME, including the promotion of cancer cell apoptosis, inhibition of angiogenesis, preventing immune escape, manipulating tumor metabolic reorganization and improving drug resistance. In the present review, the potential role of ROS in anti‑tumor therapy was summarized, including the possibility of directly or indirectly targeting the TME.
Collapse
Affiliation(s)
- Wei Liang
- Department of Radiation Oncology, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine, Affiliated Hospital of Hebei Medical University, Cangzhou, Hebei 061000, P.R. China
| | - Xinying He
- Department of Radiation Oncology, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine, Affiliated Hospital of Hebei Medical University, Cangzhou, Hebei 061000, P.R. China
| | - Jianqiang Bi
- Department of Radiation Oncology, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine, Affiliated Hospital of Hebei Medical University, Cangzhou, Hebei 061000, P.R. China
| | - Tingting Hu
- Department of Radiation Oncology, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine, Affiliated Hospital of Hebei Medical University, Cangzhou, Hebei 061000, P.R. China
| | - Yunchuan Sun
- Department of Radiation Oncology, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine, Affiliated Hospital of Hebei Medical University, Cangzhou, Hebei 061000, P.R. China
| |
Collapse
|
8
|
Electrospun nanofibers enhance trehalose synthesis by regulating gene expression for Micrococcus luteus fermentation. Colloids Surf B Biointerfaces 2021; 202:111714. [PMID: 33765627 DOI: 10.1016/j.colsurfb.2021.111714] [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: 01/17/2021] [Revised: 03/16/2021] [Accepted: 03/17/2021] [Indexed: 11/23/2022]
Abstract
In this study, mesoporous polyacrylonitrile (PAN)/thermoplastic polyurethane (TPU) blended nanofibers were prepared to immobilize Micrococcus luteus for enhancing the conversion of trehalose. The images of SEM showed the cells were adsorbed on the surface and pores due to the unique pore structure. The results of contact angle, Zeta potential and water holding ratio exhibited the good hydrophilicity and stability of PAN/TPU-P2. Besides, it was indicated that the biomass and immobilization efficiency were increased to 0.633 g/L and 0.153 g/g, respectively. It was the most noteworthy that the trehalose yield could reach 23.46 g/L, which was 71.62 % higher than that of the control in the multi-batch fermentation. Moreover, the reactive oxygen species (ROS) level was decreased to 12.8 % while the enzyme concentration was increased to 11.176 mg/mL. Meanwhile, it was also found that PAN/TPU-P2 immobilization substantially increased the expression of target gene MtreY by 3.500 times. In other words, the mechanism by which immobilized cells increased trehalose yield was that PAN/TPU-P regulated gene expression of MtreY. Therefore, this research provided theoretical foundation for the metabolic regulation of sufficient trehalose production by immobilized cells.
Collapse
|
9
|
Ma L, Zhang M, Zhao R, Wang D, Ma Y, Li A. Plant Natural Products: Promising Resources for Cancer Chemoprevention. Molecules 2021; 26:933. [PMID: 33578780 PMCID: PMC7916513 DOI: 10.3390/molecules26040933] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 02/07/2021] [Accepted: 02/08/2021] [Indexed: 01/01/2023] Open
Abstract
Cancer is a major factor threatening human health and life safety, and there is a lack of safe and effective therapeutic drugs. Intervention and prevention in premalignant process are effective ways to reverse carcinogenesis and prevent cancer from occurring. Plant natural products are rich in sources and are a promising source for cancer chemoprevention. This article reviews the chemopreventive effects of natural products, especially focused on polyphenols, flavonoids, monoterpene and triterpenoids, sulfur compounds, and cellulose. Meanwhile, the main mechanisms include induction of apoptosis, antiproliferation and inhibition of metastasis are briefly summarized. In conclusion, this article provides evidence for natural products remaining a prominent source of cancer chemoprevention.
Collapse
Affiliation(s)
- Li Ma
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - MengMeng Zhang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Rong Zhao
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Dan Wang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - YueRong Ma
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Ai Li
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| |
Collapse
|
10
|
Shen H, Shen J, Pan H, Xu L, Sheng H, Liu B, Yao M. Curcumin analog B14 has high bioavailability and enhances the effect of anti-breast cancer cells in vitro and in vivo. Cancer Sci 2020; 112:815-827. [PMID: 33316116 PMCID: PMC7894010 DOI: 10.1111/cas.14770] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 12/06/2020] [Accepted: 12/08/2020] [Indexed: 12/21/2022] Open
Abstract
Curcumin has a variety of anticancer properties, but low bioavailability prevents its use in chemotherapeutic applications. To address this problem, we tested the efficacy of the synthetic curcumin analog B14 in breast cancer cells and explored the mechanism by which B14 inhibits proliferation and metastasis of breast cancer cells. We used the breast cancer cell line MCF-7, MDA-MB-231 to study the anticancer effects of B14 and assessed cell viability, cell migration and invasion, cell cycle, and apoptosis, in addition, the antitumor effect of B14 in vivo was examined in mice bearing MDA-MB-231 cells. We found that, as the concentration of B14 increased, cell viability decreased in a dose-dependent manner. Compound B14 exerted the best antitumor activity and selectivity for MCF-7 and MDA-M-231 cells (IC50 = 8.84 μmol/L and 8.33 μmol/L, respectively), while its IC50 value for MCF-10A breast epithelial cells was 34.96 μmol/L. B14 has been shown to be a multi-targeted drug that alters the expression of cyclin D1, cyclin E1, and cyclin-dependent kinase 2 (CDK2), and ultimately induces G1 phase cell cycle arrest. At the same time, B14 activates the mitochondrial apoptosis pathway in breast cancer cells. Furthermore, B14 was more effective than curcumin in inhibiting cell migration, invasion, and colony formation. In tumor-bearing mice, analog B14 significantly reduced tumor growth and inhibited cell proliferation and angiogenesis. The pharmacokinetic test found that B14 was more stable than curcumin in vivo. Our data reveal the therapeutic potential of the curcumin analog B14 and the underlying mechanisms to fight breast cancer cells.
Collapse
Affiliation(s)
- Hui Shen
- Department of Central Laboratory, The First Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Jianfen Shen
- Department of Central Laboratory, The First Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Huan Pan
- Department of Central Laboratory, The First Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Longsheng Xu
- Department of Central Laboratory, The First Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Han Sheng
- Department of Nursing, The First Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Beibei Liu
- Department of Central Laboratory, The First Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Ming Yao
- Department of Anesthesiology and Pain Medicine, The First Affiliated Hospital of Jiaxing University, Jiaxing, China
| |
Collapse
|
11
|
Free Radicals as a Double-Edged Sword: The Cancer Preventive and Therapeutic Roles of Curcumin. Molecules 2020; 25:molecules25225390. [PMID: 33217990 PMCID: PMC7698794 DOI: 10.3390/molecules25225390] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 11/13/2020] [Accepted: 11/15/2020] [Indexed: 01/07/2023] Open
Abstract
Free radicals, generally composed of reactive oxygen species (ROS) and reactive nitrogen species (RNS), are generated in the body by various endogenous and exogenous systems. The overproduction of free radicals is known to cause several chronic diseases including cancer. However, increased production of free radicals by chemotherapeutic drugs is also associated with apoptosis in cancer cells, indicating the dual nature of free radicals. Among various natural compounds, curcumin manifests as an antioxidant in normal cells that helps in the prevention of carcinogenesis. It also acts as a prooxidant in cancer cells and is associated with inducing apoptosis. Curcumin quenches free radicals, induces antioxidant enzymes (catalase, superoxide dismutase, glutathione peroxidase), and upregulates antioxidative protein markers-Nrf2 and HO-1 that lead to the suppression of cellular oxidative stress. In cancer cells, curcumin aggressively increases ROS that results in DNA damage and subsequently cancer cell death. It also sensitizes drug-resistant cancer cells and increases the anticancer effects of chemotherapeutic drugs. Thus, curcumin shows beneficial effects in prevention, treatment and chemosensitization of cancer cells. In this review, we will discuss the dual role of free radicals as well as the chemopreventive and chemotherapeutic effects of curcumin and its analogues against cancer.
Collapse
|
12
|
Polychemotherapy with Curcumin and Doxorubicin via Biological Nanoplatforms: Enhancing Antitumor Activity. Pharmaceutics 2020; 12:pharmaceutics12111084. [PMID: 33187385 PMCID: PMC7697177 DOI: 10.3390/pharmaceutics12111084] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 10/31/2020] [Accepted: 11/07/2020] [Indexed: 12/12/2022] Open
Abstract
Doxorubicin (DOX) is a well-known chemotherapeutic agent extensively applied in the field of cancer therapy. However, similar to other chemotherapeutic agents such as cisplatin, paclitaxel, docetaxel, etoposide and oxaliplatin, cancer cells are able to obtain chemoresistance that limits DOX efficacy. In respect to dose-dependent side effect of DOX, enhancing its dosage is not recommended for effective cancer chemotherapy. Therefore, different strategies have been considered for reversing DOX resistance and diminishing its side effects. Phytochemical are potential candidates in this case due to their great pharmacological activities. Curcumin is a potential antitumor phytochemical isolated from Curcuma longa with capacity of suppressing cancer metastasis and proliferation and affecting molecular pathways. Experiments have demonstrated the potential of curcumin for inhibiting chemoresistance by downregulating oncogene pathways such as MMP-2, TGF-β, EMT, PI3K/Akt, NF-κB and AP-1. Furthermore, coadministration of curcumin and DOX potentiates apoptosis induction in cancer cells. In light of this, nanoplatforms have been employed for codelivery of curcumin and DOX. This results in promoting the bioavailability and internalization of the aforementioned active compounds in cancer cells and, consequently, enhancing their antitumor activity. Noteworthy, curcumin has been applied for reducing adverse effects of DOX on normal cells and tissues via reducing inflammation, oxidative stress and apoptosis. The current review highlights the anticancer mechanism, side effects and codelivery of curcumin and DOX via nanovehicles.
Collapse
|
13
|
Ning W, Li S, Yang W, Yang B, Xin C, Ping X, Huang C, Gu Y, Guo L. Blocking exosomal miRNA-153-3p derived from bone marrow mesenchymal stem cells ameliorates hypoxia-induced myocardial and microvascular damage by targeting the ANGPT1-mediated VEGF/PI3k/Akt/eNOS pathway. Cell Signal 2020; 77:109812. [PMID: 33164880 DOI: 10.1016/j.cellsig.2020.109812] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 10/16/2020] [Accepted: 10/16/2020] [Indexed: 01/18/2023]
Abstract
It has been widely reported that exosomes derived from mesenchymal stem cells (MSCs) have a protective effect on myocardial infarction (MI). However, the specific molecules which play a damaging role in MSCs shuttled miRNAs are much less explored. MiRNA-153-3p (miR-153-3p) is a vital miRNA which has been proved to modulate cell proliferation, apoptosis, angiogenesis, peritoneal fibrosis and aortic calcification. Here, we aim to study the effect and mechanism of miR-153-3p in MSC-derived exosomes on hypoxia-induced myocardial and microvascular damage. The exosomes of MSCs were isolated and identified, and the MSCs-exosomes with low expression of miR-153-3p (exo-miR-153-3p-) were constructed to interfere with the endothelial cells and cardiomyocytes in the oxygen-glucose deprivation (OGD) model. The viability, apoptosis, angiogenesis of endothelial cells and cardiomyocytes were determined. Additionally, ANGPT1/VEGF/VEGFR2/PI3K/Akt/eNOS pathway was detected by ELISA and/or western blot. The results illustrated that exo-miR-153-3p- significantly reduced the apoptosis of endothelial cells and cardiomyocytes and promoted their viability. Meanwhile, exo-miR-153-3p- can promote the angiogenesis of endothelial cells. Mechanistically, miR-153-3p regulates the VEGF/VEGFR2/PI3K/Akt/eNOS pathways by targeting ANGPT1. Intervention with VEGFR2 inhibitor (SU1498, 1 μM) remarkably reversed the protective effect of exo-miR-153-3p- in vascular endothelial cells and cardiomyocytes treated by OGD. Collectively, MSCs-derived exosomes with low-expressed miR-153-3p notably promotes the activation of ANGPT1 and the VEGF/VEGFR2 /PI3K/Akt/eNOS pathways, thereby preventing the damages endothelial cells and cardiomyocytes against hypoxia.
Collapse
Affiliation(s)
- Wenlong Ning
- Department of Emergency, the First Hospital of Qiqihar, Qiqihar 161005, Heilongjiang, China; Department of Emergency, Affiliated Qiqihar Hospital, Southern Medical University, Qiqihar 161000, Heilongjiang, China
| | - Shuhua Li
- Department of Emergency, the First Hospital of Qiqihar, Qiqihar 161005, Heilongjiang, China; Department of Traditional Chinese Medicine, the First Hospital of Qiqihar, Qiqihar 161005, Heilongjiang, China; Department of Traditional Chinese Medicine, Affiliated Qiqihar Hospital, Southern Medical University, Qiqihar, Heilongjiang, China
| | - Weiguang Yang
- Department of Emergency, the First Hospital of Qiqihar, Qiqihar 161005, Heilongjiang, China; Department of Emergency, Affiliated Qiqihar Hospital, Southern Medical University, Qiqihar 161000, Heilongjiang, China
| | - Bo Yang
- Department of Traditional Chinese Medicine, the First Hospital of Qiqihar, Qiqihar 161005, Heilongjiang, China; Department of Traditional Chinese Medicine, Affiliated Qiqihar Hospital, Southern Medical University, Qiqihar, Heilongjiang, China
| | - Chuanyou Xin
- Department of Emergency, the First Hospital of Qiqihar, Qiqihar 161005, Heilongjiang, China; Department of Emergency, Affiliated Qiqihar Hospital, Southern Medical University, Qiqihar 161000, Heilongjiang, China
| | - Xin Ping
- Department of Emergency, the First Hospital of Qiqihar, Qiqihar 161005, Heilongjiang, China; Department of Emergency, Affiliated Qiqihar Hospital, Southern Medical University, Qiqihar 161000, Heilongjiang, China
| | - Chuanqi Huang
- Department of Emergency, the First Hospital of Qiqihar, Qiqihar 161005, Heilongjiang, China; Department of Emergency, Affiliated Qiqihar Hospital, Southern Medical University, Qiqihar 161000, Heilongjiang, China
| | - Yan Gu
- Department of Emergency, the First Hospital of Qiqihar, Qiqihar 161005, Heilongjiang, China; Department of Emergency, Affiliated Qiqihar Hospital, Southern Medical University, Qiqihar 161000, Heilongjiang, China
| | - Longzhe Guo
- Department of Emergency, the First Hospital of Qiqihar, Qiqihar 161005, Heilongjiang, China; Department of Emergency, Affiliated Qiqihar Hospital, Southern Medical University, Qiqihar 161000, Heilongjiang, China; Department of Anatomy, School of Basic Medical Sciences, Harbin Medical University, Harbin 150081, Heilongjiang, China.
| |
Collapse
|
14
|
Wang J, Toan S, Zhou H. Mitochondrial quality control in cardiac microvascular ischemia-reperfusion injury: New insights into the mechanisms and therapeutic potentials. Pharmacol Res 2020; 156:104771. [PMID: 32234339 DOI: 10.1016/j.phrs.2020.104771] [Citation(s) in RCA: 115] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Revised: 03/13/2020] [Accepted: 03/19/2020] [Indexed: 12/17/2022]
Abstract
Thrombolytic therapy and revascularization strategies create a complete recanalization of the occluded epicardial coronary artery in patients with myocardial infarction (MI). However, about 35 % of patients still experience an impaired myocardial reperfusion, which is termed a no-reflow phenomenon mainly caused by cardiac microvascular ischemia-reperfusion (I/R) injury. Mitochondria are essential for microvascular endothelial cells' survival, both because of their roles as metabolic energy producers and as regulators of programmed cell death. Mitochondrial structure and function are regulated by a mitochondrial quality control (MQC) system, a series of processes including mitochondrial biogenesis, mitochondrial dynamics/mitophagy, mitochondrial proteostasis, and mitochondria-mediated cell death. Our review discusses the MQC mechanisms and how they are linked to cardiac microvascular I/R injury. Additionally, we will summarize the molecular basis that results in defective MQC mechanisms and present potential therapeutic interventions for improving MQC in cardiac microvascular I/R injury.
Collapse
Affiliation(s)
- Jin Wang
- Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing 100853, China
| | - Sam Toan
- Department of Chemical Engineering, University of Minnesota-Duluth, Duluth, MN 55812, USA
| | - Hao Zhou
- Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing 100853, China.
| |
Collapse
|